Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Circ Res ; 133(11): 927-943, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37846569

ABSTRACT

BACKGROUND: Cardiac ventricles provide the contractile force of the beating heart throughout life. How the primitive endocardium-layered myocardial projections called trabeculae form and mature into the adult ventricles is of great interest for biology and regenerative medicine. Trabeculation is dependent on the signaling protein Nrg1 (neuregulin-1). However, the mechanism of action of Nrg1 and its role in ventricular wall maturation are poorly understood. METHODS: We investigated the functions and downstream mechanisms of Nrg1 signaling during ventricular chamber development using confocal imaging, transcriptomics, and biochemical approaches in mice with cardiac-specific inactivation or overexpression of Nrg1. RESULTS: Analysis of cardiac-specific Nrg1 mutant mice showed that the transcriptional program underlying cardiomyocyte-oriented cell division and trabeculae formation depends on endocardial Nrg1 to myocardial ErbB2 (erb-b2 receptor tyrosine kinase 2) signaling and phospho-Erk (phosphorylated extracellular signal-regulated kinase; pErk) activation. Early endothelial loss of Nrg1 and reduced pErk activation diminished cardiomyocyte Pard3 and Crumbs2 (Crumbs Cell Polarity Complex Component 2) protein and altered cytoskeletal gene expression and organization. These alterations are associated with abnormal gene expression related to mitotic spindle organization and a shift in cardiomyocyte division orientation. Nrg1 is crucial for trabecular growth and ventricular wall thickening by regulating an epithelial-to-mesenchymal transition-like process in cardiomyocytes involving migration, adhesion, cytoskeletal actin turnover, and timely progression through the cell cycle G2/M phase. Ectopic cardiac Nrg1 overexpression and high pErk signaling caused S-phase arrest, sustained high epithelial-to-mesenchymal transition-like gene expression, and prolonged trabeculation, blocking compact myocardium maturation. Myocardial trabecular patterning alterations resulting from above- or below-normal Nrg1-dependent pErk activation were concomitant with sarcomere actin cytoskeleton disorganization. The Nrg1 loss- and gain-of-function transcriptomes were enriched for Yap1 (yes-associated protein-1) gene signatures, identifying Yap1 as a potential downstream effector. Furthermore, biochemical and imaging data reveal that Nrg1 influences pErk activation and Yap1 nuclear-cytoplasmic distribution during trabeculation. CONCLUSIONS: These data establish the Nrg1-ErbB2/ErbB4-Erk axis as a crucial regulator of cardiomyocyte cell cycle progression and migration during ventricular development.


Subject(s)
Myocytes, Cardiac , Neuregulin-1 , Animals , Mice , Myocytes, Cardiac/metabolism , Neuregulin-1/genetics , Myocardium/metabolism , Heart Ventricles/metabolism , Cell Division
3.
Circulation ; 147(1): 47-65, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36325906

ABSTRACT

BACKGROUND: The complex genetics underlying human cardiac disease is evidenced by its heterogenous manifestation, multigenic basis, and sporadic occurrence. These features have hampered disease modeling and mechanistic understanding. Here, we show that 2 structural cardiac diseases, left ventricular noncompaction (LVNC) and bicuspid aortic valve, can be caused by a set of inherited heterozygous gene mutations affecting the NOTCH ligand regulator MIB1 (MINDBOMB1) and cosegregating genes. METHODS: We used CRISPR-Cas9 gene editing to generate mice harboring a nonsense or a missense MIB1 mutation that are both found in LVNC families. We also generated mice separately carrying these MIB1 mutations plus 5 additional cosegregating variants in the ASXL3, APCDD1, TMX3, CEP192, and BCL7A genes identified in these LVNC families by whole exome sequencing. Histological, developmental, and functional analyses of these mouse models were carried out by echocardiography and cardiac magnetic resonance imaging, together with gene expression profiling by RNA sequencing of both selected engineered mouse models and human induced pluripotent stem cell-derived cardiomyocytes. Potential biochemical interactions were assayed in vitro by coimmunoprecipitation and Western blot. RESULTS: Mice homozygous for the MIB1 nonsense mutation did not survive, and the mutation caused LVNC only in heteroallelic combination with a conditional allele inactivated in the myocardium. The heterozygous MIB1 missense allele leads to bicuspid aortic valve in a NOTCH-sensitized genetic background. These data suggest that development of LVNC is influenced by genetic modifiers present in affected families, whereas valve defects are highly sensitive to NOTCH haploinsufficiency. Whole exome sequencing of LVNC families revealed single-nucleotide gene variants of ASXL3, APCDD1, TMX3, CEP192, and BCL7A cosegregating with the MIB1 mutations and LVNC. In experiments with mice harboring the orthologous variants on the corresponding Mib1 backgrounds, triple heterozygous Mib1 Apcdd1 Asxl3 mice showed LVNC, whereas quadruple heterozygous Mib1 Cep192 Tmx3;Bcl7a mice developed bicuspid aortic valve and other valve-associated defects. Biochemical analysis suggested interactions between CEP192, BCL7A, and NOTCH. Gene expression profiling of mutant mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes revealed increased cardiomyocyte proliferation and defective morphological and metabolic maturation. CONCLUSIONS: These findings reveal a shared genetic substrate underlying LVNC and bicuspid aortic valve in which MIB1-NOTCH variants plays a crucial role in heterozygous combination with cosegregating genetic modifiers.


Subject(s)
Bicuspid Aortic Valve Disease , Cardiomyopathies , Heart Defects, Congenital , Induced Pluripotent Stem Cells , Humans , Animals , Mice , Heart Defects, Congenital/complications , Cardiomyopathies/etiology , Myocytes, Cardiac , Aortic Valve/diagnostic imaging , Transcription Factors , Chromosomal Proteins, Non-Histone
4.
Basic Res Cardiol ; 117(1): 62, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36445563

ABSTRACT

Aortic stenosis (AS) is associated with left ventricular (LV) hypertrophy and heart failure (HF). There is a lack of therapies able to prevent/revert AS-induced HF. Beta3 adrenergic receptor (ß3AR) signaling is beneficial in several forms of HF. Here, we studied the potential beneficial effect of ß3AR overexpression on AS-induced HF. Selective ß3AR stimulation had a positive inotropic effect. Transgenic mice constitutively overexpressing human ß3AR in the heart (c-hß3tg) were protected from the development of HF in response to induced AS, and against cardiomyocyte mitochondrial dysfunction (fragmented mitochondria with remodeled cristae and metabolic reprogramming featuring altered substrate use). Similar beneficial effects were observed in wild-type mice inoculated with adeno-associated virus (AAV9) inducing cardiac-specific overexpression of human ß3AR before AS induction. Moreover, AAV9-hß3AR injection into wild-type mice at late disease stages, when cardiac hypertrophy and metabolic reprogramming are already advanced, reversed the HF phenotype and restored balanced mitochondrial dynamics, demonstrating the potential of gene-therapy-mediated ß3AR overexpression in AS. Mice with cardiac specific ablation of Yme1l (cYKO), characterized by fragmented mitochondria, showed an increased mortality upon AS challenge. AAV9-hß3AR injection in these mice before AS induction reverted the fragmented mitochondria phenotype and rescued them from death. In conclusion, our results step out that ß3AR overexpression might have translational potential as a therapeutic strategy in AS-induced HF.


Subject(s)
Aortic Valve Stenosis , Heart Failure , Humans , Mice , Animals , Receptors, Adrenergic, beta-3 , Mitochondrial Dynamics , Hypertrophy, Left Ventricular , Myocytes, Cardiac , Mice, Transgenic , Metalloendopeptidases
5.
Cell Death Dis ; 12(8): 729, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294700

ABSTRACT

Bone morphogenetic protein (Bmp) signaling is critical for organismal development and homeostasis. To elucidate Bmp2 function in the vascular/hematopoietic lineages we generated a new transgenic mouse line in which ectopic Bmp2 expression is controlled by the Tie2 promoter. Tie2CRE/+;Bmp2tg/tg mice develop aortic valve dysfunction postnatally, accompanied by pre-calcific lesion formation in valve leaflets. Remarkably, Tie2CRE/+;Bmp2tg/tg mice develop extensive soft tissue bone formation typical of acquired forms of heterotopic ossification (HO) and genetic bone disorders, such as Fibrodysplasia Ossificans Progressiva (FOP). Ectopic ossification in Tie2CRE/+;Bmp2tg/tg transgenic animals is accompanied by increased bone marrow hematopoietic, fibroblast and osteoblast precursors and circulating pro-inflammatory cells. Transplanting wild-type bone marrow hematopoietic stem cells into lethally irradiated Tie2CRE/+;Bmp2tg/tg mice significantly delays HO onset but does not prevent it. Moreover, transplanting Bmp2-transgenic bone marrow into wild-type recipients does not result in HO, but hematopoietic progenitors contribute to inflammation and ectopic bone marrow colonization rather than to endochondral ossification. Conversely, aberrant Bmp2 signaling activity is associated with fibroblast accumulation, skeletal muscle fiber damage, and expansion of a Tie2+ fibro-adipogenic precursor cell population, suggesting that ectopic bone derives from a skeletal muscle resident osteoprogenitor cell origin. Thus, Tie2CRE/+;Bmp2tg/tg mice recapitulate HO pathophysiology, and might represent a useful model to investigate therapies seeking to mitigate disorders associated with aberrant extra-skeletal bone formation.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Lineage , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Receptor, TIE-2/metabolism , Animals , Aortic Valve/diagnostic imaging , Aortic Valve/pathology , Aortic Valve/physiopathology , Bone Marrow Transplantation , Bone Morphogenetic Protein 2/blood , Calcinosis/diagnostic imaging , Calcinosis/pathology , Calcinosis/physiopathology , Chondrogenesis , Endothelial Cells/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Kaplan-Meier Estimate , Mice, Inbred C57BL , Mice, Transgenic , Muscle Cells/pathology , Ossification, Heterotopic/blood , Ossification, Heterotopic/diagnostic imaging , Osteogenesis , Tomography, X-Ray Computed
6.
Elife ; 82019 12 04.
Article in English | MEDLINE | ID: mdl-31789590

ABSTRACT

Coronaries are essential for myocardial growth and heart function. Notch is crucial for mouse embryonic angiogenesis, but its role in coronary development remains uncertain. We show Jag1, Dll4 and activated Notch1 receptor expression in sinus venosus (SV) endocardium. Endocardial Jag1 removal blocks SV capillary sprouting, while Dll4 inactivation stimulates excessive capillary growth, suggesting that ligand antagonism regulates coronary primary plexus formation. Later endothelial ligand removal, or forced expression of Dll4 or the glycosyltransferase Mfng, blocks coronary plexus remodeling, arterial differentiation, and perivascular cell maturation. Endocardial deletion of Efnb2 phenocopies the coronary arterial defects of Notch mutants. Angiogenic rescue experiments in ventricular explants, or in primary human endothelial cells, indicate that EphrinB2 is a critical effector of antagonistic Dll4 and Jag1 functions in arterial morphogenesis. Thus, coronary arterial precursors are specified in the SV prior to primary coronary plexus formation and subsequent arterial differentiation depends on a Dll4-Jag1-EphrinB2 signaling cascade.


Subject(s)
Coronary Vessels/growth & development , Coronary Vessels/metabolism , Ephrin-B2/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/metabolism , Membrane Proteins/metabolism , Signal Transduction , Animals , Endocardium/metabolism , Endothelium, Vascular/metabolism , Heart Ventricles/growth & development , Heart Ventricles/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia/metabolism , Hypoxia/physiopathology , Ligands , Mice , Morphogenesis , Mutation/genetics , NFATC Transcription Factors/metabolism , Neovascularization, Physiologic , Receptors, Notch/metabolism , Stress, Physiological , Transcriptome/genetics , Vascular Remodeling
7.
Circulation ; 140(14): 1188-1204, 2019 10.
Article in English | MEDLINE | ID: mdl-31567019

ABSTRACT

BACKGROUND: Arrhythmogenic cardiomyopathy/arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited cardiac disease characterized by fibrofatty replacement of the myocardium, resulting in heart failure and sudden cardiac death. The most aggressive arrhythmogenic cardiomyopathy/ARVC subtype is ARVC type 5 (ARVC5), caused by a p.S358L mutation in TMEM43 (transmembrane protein 43). The function and localization of TMEM43 are unknown, as is the mechanism by which the p.S358L mutation causes the disease. Here, we report the characterization of the first transgenic mouse model of ARVC5. METHODS: We generated transgenic mice overexpressing TMEM43 in either its wild-type or p.S358L mutant (TMEM43-S358L) form in postnatal cardiomyocytes under the control of the α-myosin heavy chain promoter. RESULTS: We found that mice expressing TMEM43-S358L recapitulate the human disease and die at a young age. Mutant TMEM43 causes cardiomyocyte death and severe fibrofatty replacement. We also demonstrate that TMEM43 localizes at the nuclear membrane and interacts with emerin and ß-actin. TMEM43-S358L shows partial delocalization to the cytoplasm, reduced interaction with emerin and ß-actin, and activation of glycogen synthase kinase-3ß (GSK3ß). Furthermore, we show that targeting cardiac fibrosis has no beneficial effect, whereas overexpression of the calcineurin splice variant calcineurin Aß1 results in GSK3ß inhibition and improved cardiac function and survival. Similarly, treatment of TMEM43 mutant mice with a GSK3ß inhibitor improves cardiac function. Finally, human induced pluripotent stem cells bearing the p.S358L mutation also showed contractile dysfunction that was partially restored after GSK3ß inhibition. CONCLUSIONS: Our data provide evidence that TMEM43-S358L leads to sustained cardiomyocyte death and fibrofatty replacement. Overexpression of calcineurin Aß1 in TMEM43 mutant mice or chemical GSK3ß inhibition improves cardiac function and increases mice life span. Our results pave the way toward new therapeutic approaches for ARVC5.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Ventricular Dysfunction/pathology , Animals , Calcineurin/genetics , Calcineurin/metabolism , Cell Differentiation , Cell Survival/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Heart Ventricles/physiopathology , Humans , Induced Pluripotent Stem Cells/cytology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Severity of Illness Index , Ventricular Dysfunction/mortality
8.
Development ; 145(13)2018 07 02.
Article in English | MEDLINE | ID: mdl-29853617

ABSTRACT

Signaling interactions between the myocardium and endocardium pattern embryonic cardiac regions, instructing their development to fulfill specific functions in the mature heart. We show that ectopic Bmp2 expression in the mouse chamber myocardium changes the transcriptional signature of adjacent chamber endocardial cells into valve tissue, and enables them to undergo epithelial-mesenchyme transition. This induction is independent of valve myocardium specification and requires high levels of Notch1 activity. Biochemical experiments suggest that Bmp2-mediated Notch1 induction is achieved through transcriptional activation of the Notch ligand Jag1, and physical interaction of Smad1/5 with the intracellular domain of the Notch1 receptor. Thus, widespread myocardial Bmp2 and endocardial Notch signaling drive presumptive ventricular endocardium to differentiate into valve endocardium. Understanding the molecular basis of valve development is instrumental to designing therapeutic strategies for congenital heart valve defects.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Embryo, Mammalian/embryology , Endocardium/embryology , Heart Valves/embryology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Bone Morphogenetic Protein 2/genetics , Embryo, Mammalian/cytology , Endocardium/cytology , Heart Valves/cytology , Mice , Mice, Transgenic , Myocardium/cytology , Myocardium/metabolism , Receptors, Notch/genetics , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism
9.
Cell Death Dis ; 9(3): 399, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540665

ABSTRACT

During mammalian heart development, restricted myocardial Bmp2 expression is a key patterning signal for atrioventricular canal specification and the epithelial-mesenchyme transition that gives rise to the valves. Using a mouse transgenic line conditionally expressing Bmp2, we show that widespread Bmp2 expression in the myocardium leads to valve and chamber dysmorphogenesis and embryonic death by E15.5. Transgenic embryos show thickened valves, ventricular septal defect, enlarged trabeculae and dilated ventricles, with an endocardium able to undergo EMT both in vivo and in vitro. Gene profiling and marker analysis indicate that cellular proliferation is increased in transgenic embryos, whereas chamber maturation and patterning are impaired. Similarly, forced Bmp2 expression stimulates proliferation and blocks cardiomyocyte differentiation of embryoid bodies. These data show that widespread myocardial Bmp2 expression directs ectopic valve primordium formation and maintains ventricular myocardium and cardiac progenitors in a primitive, proliferative state, identifying the potential of Bmp2 in the expansion of immature cardiomyocytes.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Proliferation , Epithelial-Mesenchymal Transition , Myocardium/metabolism , Myocytes, Cardiac/cytology , Animals , Bone Morphogenetic Protein 2/genetics , Gene Expression Regulation, Developmental , Heart/embryology , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Signal Transduction
10.
Stud Health Technol Inform ; 205: 131-5, 2014.
Article in English | MEDLINE | ID: mdl-25160160

ABSTRACT

The world concern about the costs of the health care systems has raised the importance of counting on precise and interpretable tools, that help the health care institution's managers to make decisions to optimize the use of health resources. In this paper we propose a new Classification based on Association Rules (CAR) algorithm that improves the interpretability of the results, making it specially useful for decision making. Changing the usual way to obtain the rules we follow four goals: first to improve the interpretability of the result by obtaining rules meaningful and interpretable by themselves, secondly to reduce the complexity of the result obtaining a lower number of rules; thirdly, to obtain simpler rules, with less size in number of antecedents; and finally to avoid the usual over-fitting problem of the classification methods by obtaining a generic final result set, where specific rules for specific cases are avoided unless they are necessary. To prove the utility of our proposal we have used it in an example of decision support regarding the planning of the surgery rooms.


Subject(s)
Algorithms , Decision Making, Organizational , Decision Support Systems, Management/organization & administration , Decision Support Techniques , Efficiency, Organizational , Hospital Administration/methods , Operating Rooms/organization & administration
11.
Nat Med ; 19(2): 193-201, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23314057

ABSTRACT

Left ventricular noncompaction (LVNC) causes prominent ventricular trabeculations and reduces cardiac systolic function. The clinical presentation of LVNC ranges from asymptomatic to heart failure. We show that germline mutations in human MIB1 (mindbomb homolog 1), which encodes an E3 ubiquitin ligase that promotes endocytosis of the NOTCH ligands DELTA and JAGGED, cause LVNC in autosomal-dominant pedigrees, with affected individuals showing reduced NOTCH1 activity and reduced expression of target genes. Functional studies in cells and zebrafish embryos and in silico modeling indicate that MIB1 functions as a dimer, which is disrupted by the human mutations. Targeted inactivation of Mib1 in mouse myocardium causes LVNC, a phenotype mimicked by inactivation of myocardial Jagged1 or endocardial Notch1. Myocardial Mib1 mutants show reduced ventricular Notch1 activity, expansion of compact myocardium to proliferative, immature trabeculae and abnormal expression of cardiac development and disease genes. These results implicate NOTCH signaling in LVNC and indicate that MIB1 mutations arrest chamber myocardium development, preventing trabecular maturation and compaction.


Subject(s)
Cardiomyopathies/etiology , Heart Ventricles , Mutation , Receptors, Notch/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/genetics , Amino Acid Sequence , Animals , Cardiomyopathies/genetics , Female , HEK293 Cells , Heart/embryology , Heart Ventricles/embryology , Humans , Male , Mice , Molecular Sequence Data , Protein Multimerization , Ubiquitin-Protein Ligases/physiology , Zebrafish
12.
J Clin Invest ; 120(10): 3493-507, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20890042

ABSTRACT

Cardiac valve formation is crucial for embryonic and adult heart function. Valve malformations constitute the most common congenital cardiac defect, but little is known about the molecular mechanisms regulating valve formation and homeostasis. Here, we show that endocardial Notch1 and myocardial Bmp2 signal integration establish a valve-forming field between 2 chamber developmental domains. Patterning occurs through the activation of endocardial epithelial-to-mesenchymal transition (EMT) exclusively in prospective valve territories. Mice with constitutive endocardial Notch1 activity ectopically express Hey1 and Heyl. They also display an activated mesenchymal gene program in ventricles and a partial (noninvasive) EMT in vitro that becomes invasive upon BMP2 treatment. Snail1, TGF-ß2, or Notch1 inhibition reduces BMP2-induced ventricular transformation and invasion, whereas BMP2 treatment inhibits endothelial Gsk3ß, stabilizing Snail1 and promoting invasiveness. Integration of Notch and Bmp2 signals is consistent with Notch1 signaling being attenuated after myocardial Bmp2 deletion. Notch1 activation in myocardium extends Hey1 expression to nonchamber myocardium, represses Bmp2, and impairs EMT. In contrast, Notch deletion abrogates endocardial Hey gene transcription and extends Bmp2 expression to the ventricular endocardium. This embryonic Notch1-Bmp2-Snail1 relationship may be relevant in adult valve disease, in which decreased NOTCH signaling causes valve mesenchyme cell formation, fibrosis, and calcification.


Subject(s)
Bone Morphogenetic Protein 2/physiology , Heart Valves/embryology , Mesoderm/metabolism , Receptor, Notch1/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Cell Cycle Proteins/analysis , Epithelial Cells/pathology , Gene Expression Regulation, Developmental , Humans , Mesoderm/pathology , Mice , Repressor Proteins/analysis , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/physiology , Transforming Growth Factor beta2/physiology
13.
Am J Pathol ; 170(5): 1659-68, 2007 May.
Article in English | MEDLINE | ID: mdl-17456771

ABSTRACT

Walker-Warburg syndrome (WWS) is the most severe of a group of congenital disorders that have in common defects in the O-glycosylation of alpha-dystroglycan. WWS is characterized by congenital muscular dystrophy coupled with severe ocular and brain malformations. Moreover, in at least one-fifth of the reported cases, mutations in the POMT1 gene are responsible for this disease. During embryonic development (E8.5 to E11.5), the mouse Pomt1 gene is expressed in the tissues most severely affected in WWS, the muscle, eye, and brain. In this study, we show that mPomt1 expression is maintained in the muscle and eye in later developmental stages and, notably, that its expression is particularly strong in regions of brain and cerebellum that, when affected, could generate the defects observed in patients with WWS. We show that the Pomt1 protein is localized to the sarcoplasmic reticulum of muscle tissue cells in adult mice, where alpha-dystroglycan is O-glycosylated. Furthermore, the Pomt1 protein is localized to the acrosome of maturing spermatids, where alpha-dystroglycan is not glycosylated, so that Pomt1 might have a different target for O-mannosylation in the testes. This expression pattern in the testes could also be related to the gonadal anomalies observed in some patients with WWS.


Subject(s)
Abnormalities, Multiple/enzymology , Brain/enzymology , Mannosyltransferases/biosynthesis , Muscle, Skeletal/enzymology , Muscular Dystrophy, Animal/enzymology , Acrosome/enzymology , Animals , Blotting, Western , Brain/embryology , Brain/growth & development , Dystroglycans/metabolism , Embryo, Mammalian , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , In Situ Hybridization , Male , Mice , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Myocardium/enzymology , RNA, Messenger/analysis , Sarcoplasmic Reticulum/enzymology
14.
Proc Natl Acad Sci U S A ; 101(39): 14126-31, 2004 Sep 28.
Article in English | MEDLINE | ID: mdl-15383666

ABSTRACT

O-mannosylation is an important protein modification in eukaryotes that is initiated by an evolutionarily conserved family of protein O-mannosyltransferases. The first mammalian protein O-mannosyltransferase gene described was the human POMT1. Mutations in the hPOMT1 gene are responsible for Walker-Warburg syndrome (WWS), a severe recessive congenital muscular dystrophy associated with defects in neuronal migration that produce complex brain and eye abnormalities. During embryogenesis, the murine Pomt1 gene is prominently expressed in the neural tube, the developing eye, and the mesenchyme. These sites of expression correlate with those in which the main tissue alterations are observed in WWS patients. We have inactivated a Pomt1 allele by gene targeting in embryonic stem cells and produced chimeras transmitting the defect allele to offspring. Although heterozygous mice were viable and fertile, the total absence of Pomt1(-/-) pups in the progeny of heterozygous intercrosses indicated that this genotype is embryonic lethal. An analysis of the mutant phenotype revealed that homozygous Pomt1(-/-) mice suffer developmental arrest around embryonic day (E) 7.5 and die between E7.5 and E9.5. The Pomt1(-/-) embryos present defects in the formation of Reichert's membrane, the first basement membrane to form in the embryo. The failure of this membrane to form appears to be the result of abnormal glycosylation and maturation of dystroglycan that may impair recruitment of laminin, a structural component required for the formation of Reichert's membrane in rodents. The targeted disruption of mPomt1 represents an example of an engineered deletion of a known glycosyltransferase involved in O-mannosyl glycan synthesis.


Subject(s)
Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Fetal Death/genetics , Mannosyltransferases/genetics , Abnormalities, Multiple/enzymology , Animals , Base Sequence , Brain/abnormalities , Brain/embryology , Extracellular Matrix/physiology , Eye Abnormalities/genetics , Female , Fetal Death/embryology , Gene Expression/physiology , Gene Targeting , Glycosylation , Hematoxylin/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Laminin/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Pregnancy , Recombination, Genetic , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...