Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Front Mol Biosci ; 10: 1169658, 2023.
Article in English | MEDLINE | ID: mdl-37342207

ABSTRACT

Despite the identification of numerous molecular pathways modulating cardiac hypertrophy its pathogenesis is not completely understood. In this study we define an unexpected role for Fibin ("fin bud initiation factor homolog") in cardiomyocyte hypertrophy. Via gene expression profiling in hypertrophic murine hearts after transverse aortic constriction we found a significant induction of Fibin. Moreover, Fibin was upregulated in another mouse model of cardiac hypertrophy (calcineurin-transgenics) as well as in patients with dilated cardiomyopathy. Immunoflourescence microscopy revealed subcellular localization of Fibin at the sarcomeric z-disc. Overexpression of Fibin in neonatal rat ventricular cardiomyocytes revealed a strong anti-hypertrophic effect through inhibiting both, NFAT- and SRF-dependent signalling. In contrast, transgenic mice with cardiac-restricted overexpression of Fibin developed dilated cardiomyopathy, accompanied by induction of hypertrophy-associated genes. Moreover, Fibin overexpression accelerated the progression to heart failure in the presence of prohypertrophic stimuli such as pressure overload and calcineurin overexpression. Histological and ultrastructural analyses surprisingly showed large protein aggregates containing Fibin. On the molecular level, aggregate formation was accompanied by an induction of the unfolded protein response subsequent UPR-mediated apoptosis and autophagy. Taken together, we identified Fibin as a novel potent negative regulator of cardiomyocyte hypertrophy in vitro. Yet, heart-specific Fibin overexpression in vivo causes development of a protein-aggregate-associated cardiomyopathy. Because of close similarities to myofibrillar myopathies, Fibin represents a candidate gene for cardiomyopathy and Fibin transgenic mice may provide additional mechanistic insight into aggregate formation in these diseases.

2.
Cell Mol Life Sci ; 78(16): 6033-6049, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34274976

ABSTRACT

Melanocytes are pigmented cells residing mostly in the skin and hair follicles of vertebrates, where they contribute to colouration and protection against UV-B radiation. However, the spectrum of their functions reaches far beyond that. For instance, these pigment-producing cells are found inside the inner ear, where they contribute to the hearing function, and in the heart, where they are involved in the electrical conductivity and support the stiffness of cardiac valves. The embryonic origin of such extracutaneous melanocytes is not clear. We took advantage of lineage-tracing experiments combined with 3D visualizations and gene knockout strategies to address this long-standing question. We revealed that Schwann cell precursors are recruited from the local innervation during embryonic development and give rise to extracutaneous melanocytes in the heart, brain meninges, inner ear, and other locations. In embryos with a knockout of the EdnrB receptor, a condition imitating Waardenburg syndrome, we observed only nerve-associated melanoblasts, which failed to detach from the nerves and to enter the inner ear. Finally, we looked into the evolutionary aspects of extracutaneous melanocytes and found that pigment cells are associated mainly with nerves and blood vessels in amphibians and fish. This new knowledge of the nerve-dependent origin of extracutaneous pigment cells might be directly relevant to the formation of extracutaneous melanoma in humans.


Subject(s)
Brain/physiology , Ear, Inner/physiology , Heart/physiology , Meninges/physiology , Nervous System/physiopathology , Schwann Cells/physiology , Amphibians/metabolism , Amphibians/physiology , Animals , Brain/metabolism , Cell Lineage/physiology , Ear, Inner/metabolism , Embryonic Development/physiology , Female , Fishes/metabolism , Fishes/physiology , Melanocytes/metabolism , Melanocytes/physiology , Meninges/metabolism , Mice , Nervous System/metabolism , Pregnancy , Receptor, Endothelin B/metabolism , Schwann Cells/metabolism
3.
JACC Basic Transl Sci ; 6(4): 365-380, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33997522

ABSTRACT

Autophagy is a cellular degradation process that has been implicated in diverse disease processes. The authors provide evidence that FYCO1, a component of the autophagic machinery, is essential for adaptation to cardiac stress. Although the absence of FYCO1 does not affect basal autophagy in isolated cardiomyocytes, it abolishes induction of autophagy after glucose deprivation. Likewise, Fyco1-deficient mice subjected to starvation or pressure overload are unable to respond with induction of autophagy and develop impaired cardiac function. FYCO1 overexpression leads to induction of autophagy in isolated cardiomyocytes and transgenic mouse hearts, thereby rescuing cardiac dysfunction in response to biomechanical stress.

4.
Nutr Res ; 87: 49-56, 2021 03.
Article in English | MEDLINE | ID: mdl-33601214

ABSTRACT

Infancy may represent a sensitive window for establishing food preferences that could affect the individual's long-term potential to establish healthy eating patterns. Our study was based on the hypothesis that preserving the natural flavor of the ingredients of commercially prepared complementary foods would increase the acceptance of new foods, especially vegetables. Frozen vegetable-based meals for infants were developed to preserve the natural taste of the ingredients better than sterilization of meals in jars. In a 3-month randomized, controlled intervention study, 51 infants were fed either frozen menus (intervention group) or commercial sterilized meals in jars (control group) on at least 5 days per week. Then the acceptability of a known vegetable-based puree was tested in comparison to an unknown puree, measuring the quantities consumed and also the mother's assessment of the infants' liking. In conclusion, the results of this study clearly indicated that infants fed vegetable-based frozen meals for 3 months accepted a new vegetable better than infants fed sterilized commercial meals in jars.


Subject(s)
Food Preferences , Frozen Foods , Infant Food , Infant Nutritional Physiological Phenomena , Vegetables , Feeding Behavior , Female , Humans , Infant , Male , Taste
5.
Nat Commun ; 10(1): 5243, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748531

ABSTRACT

Familial exudative vitreoretinopathy (FEVR) is a human disease characterized by defective retinal angiogenesis and associated complications that can result in vision loss. Defective Wnt/ß-catenin signaling is an established cause of FEVR, whereas other molecular alterations contributing to the disease remain insufficiently understood. Here, we show that integrin-linked kinase (ILK), a mediator of cell-matrix interactions, is indispensable for retinal angiogenesis. Inactivation of the murine Ilk gene in postnatal endothelial cells results in sprouting defects, reduced endothelial proliferation and disruption of the blood-retina barrier, resembling phenotypes seen in established mouse models of FEVR. Retinal vascularization defects are phenocopied by inducible inactivation of the gene for α-parvin (Parva), an interactor of ILK. Screening genomic DNA samples from exudative vitreoretinopathy patients identifies three distinct mutations in human ILK, which compromise the function of the gene product in vitro. Together, our data suggest that defective cell-matrix interactions are linked to Wnt signaling and FEVR.


Subject(s)
Blood-Retinal Barrier/metabolism , Endothelial Cells/metabolism , Familial Exudative Vitreoretinopathies/genetics , Neovascularization, Physiologic/genetics , Protein Serine-Threonine Kinases/genetics , Retinal Vessels/growth & development , Animals , Endothelial Cells/cytology , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Microfilament Proteins/genetics , Phenotype , Wnt Signaling Pathway/genetics
6.
Cell Rep ; 27(4): 1293-1306.e6, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31018141

ABSTRACT

Gene expression profiles of more than 10,000 individual microglial cells isolated from cortex and hippocampus of male and female AppNL-G-F mice over time demonstrate that progressive amyloid-ß accumulation accelerates two main activated microglia states that are also present during normal aging. Activated response microglia (ARMs) are composed of specialized subgroups overexpressing MHC type II and putative tissue repair genes (Dkk2, Gpnmb, and Spp1) and are strongly enriched with Alzheimer's disease (AD) risk genes. Microglia from female mice progress faster in this activation trajectory. Similar activated states are also found in a second AD model and in human brain. Apoe, the major genetic risk factor for AD, regulates the ARMs but not the interferon response microglia (IRMs). Thus, the ARMs response is the converging point for aging, sex, and genetic AD risk factors.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Biomarkers/metabolism , Brain/pathology , Disease Models, Animal , Microglia/pathology , Plaque, Amyloid/pathology , Aging/genetics , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , Biomarkers/analysis , Brain/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Presenilins/physiology , Sex Characteristics
8.
Cell ; 175(2): 400-415.e13, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30173915

ABSTRACT

Macrophages are highly heterogeneous tissue-resident immune cells that perform a variety of tissue-supportive functions. The current paradigm dictates that intestinal macrophages are continuously replaced by incoming monocytes that acquire a pro-inflammatory or tissue-protective signature. Here, we identify a self-maintaining population of macrophages that arise from both embryonic precursors and adult bone marrow-derived monocytes and persists throughout adulthood. Gene expression and imaging studies of self-maintaining macrophages revealed distinct transcriptional profiles that reflect their unique localization (i.e., closely positioned to blood vessels, submucosal and myenteric plexus, Paneth cells, and Peyer's patches). Depletion of self-maintaining macrophages resulted in morphological abnormalities in the submucosal vasculature and loss of enteric neurons, leading to vascular leakage, impaired secretion, and reduced intestinal motility. These results provide critical insights in intestinal macrophage heterogeneity and demonstrate the strategic role of self-maintaining macrophages in gut homeostasis and intestinal physiology.


Subject(s)
Intestines/immunology , Macrophages/immunology , Animals , Body Patterning/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Gastrointestinal Motility/immunology , Gastrointestinal Motility/physiology , Homeostasis , Inflammation/immunology , Intestinal Mucosa/immunology , Intestine, Small/metabolism , Mice , Monocytes/metabolism , Neurons/metabolism , Phagocytes/immunology , Transcriptome
9.
Nat Commun ; 8(1): 1574, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29146905

ABSTRACT

Pericytes adhere to the abluminal surface of endothelial tubules and are required for the formation of stable vascular networks. Defective endothelial cell-pericyte interactions are frequently observed in diseases characterized by compromised vascular integrity such as diabetic retinopathy. Many functional properties of pericytes and their exact role in the regulation of angiogenic blood vessel growth remain elusive. Here we show that pericytes promote endothelial sprouting in the postnatal retinal vasculature. Using genetic and pharmacological approaches, we show that the expression of vascular endothelial growth factor receptor 1 (VEGFR1) by pericytes spatially restricts VEGF signalling. Angiogenic defects caused by pericyte depletion are phenocopied by intraocular injection of VEGF-A or pericyte-specific inactivation of the murine gene encoding VEGFR1. Our findings establish that pericytes promote endothelial sprouting, which results in the loss of side branches and the enlargement of vessels when pericyte function is impaired or lost.


Subject(s)
Endothelial Cells/metabolism , Eye/blood supply , Neovascularization, Physiologic/physiology , Pericytes/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Capillaries/cytology , Capillaries/growth & development , Cell Line , Diphtheria Toxin/toxicity , Endothelial Cells/cytology , Heparin-binding EGF-like Growth Factor/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Pericytes/cytology , Receptor, Platelet-Derived Growth Factor beta/genetics , Retina/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/genetics
10.
Nat Cell Biol ; 19(8): 915-927, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28714968

ABSTRACT

Endothelial sprouting and proliferation are tightly coordinated processes mediating the formation of new blood vessels during physiological and pathological angiogenesis. Endothelial tip cells lead sprouts and are thought to suppress tip-like behaviour in adjacent stalk endothelial cells by activating Notch. Here, we show with genetic experiments in postnatal mice that the level of active Notch signalling is more important than the direct Dll4-mediated cell-cell communication between endothelial cells. We identify endothelial expression of VEGF-A and of the chemokine receptor CXCR4 as key processes controlling Notch-dependent vessel growth. Surprisingly, genetic experiments targeting endothelial tip cells in vivo reveal that they retain their function without Dll4 and are also not replaced by adjacent, Dll4-positive cells. Instead, activation of Notch directs tip-derived endothelial cells into developing arteries and thereby establishes that Dll4-Notch signalling couples sprouting angiogenesis and artery formation.


Subject(s)
Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Receptor, Notch1/metabolism , Retinal Artery/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell Communication , Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Cells, Cultured , Female , Gene Expression Regulation , Genotype , Intracellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptor, Notch1/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Retinal Artery/cytology , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
Appetite ; 101: 184-91, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26928790

ABSTRACT

Food neophobia (FN) is described as the rejection to eat unknown foods. Because only little is known about the role of FN in adolescence the aim of this study was to examine potential determinants of FN and associations with dietary habits of DONALD study participants. FN was measured with Pliner's and Hobden's Food Neophobia Scale (FN Score (FNS): range 10-70) in 166 10-18-year-old adolescents. Participants' age, sex, body weight status and duration of breast-feeding as well as parents' FN and educational status were considered as determinants. Energy intake, distribution of macronutrients and two variety scores were calculated from 3-day weighed dietary records. Multivariable general linear models were performed for data analyses. Boys and girls did not differ in their FNS (median (Min-Max): boys 31 (10-58), girls 32 (14-59)). Increasing age (p = 0.010) and duration of total breast-feeding (p = 0.006) were associated with decreasing FNS in girls only. FN was further positively associated with parental FN in the total sample (p = 0.004). FN was negatively associated with protein intake in the total sample (p = 0.017). The overall low level of FN in the DONALD study can be ascribed to the low level of FN in adolescence in general. Congruently with other studies, age and breast-feeding duration were identified as determinants of girls' FN and parental FN was identified as determinant of FN in the total sample. Further, our results indicate that FN leads to reduced protein intakes. Dietary variety was not strongly affected, possibly because of a broad variety of food supply in Germany.


Subject(s)
Choice Behavior , Diet , Food Preferences/psychology , Adolescent , Body Mass Index , Body Weight , Child , Cross-Sectional Studies , Diet Records , Educational Status , Female , Fruit , Germany , Humans , Male , Nutrition Assessment , Parent-Child Relations , Parents/psychology , Surveys and Questionnaires , Vegetables
12.
Nat Commun ; 5: 5758, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25502622

ABSTRACT

Tissue vascularization entails the formation of a blood vessel plexus, which remodels into arteries and veins. Here we show, by using time-lapse imaging of zebrafish fin regeneration and genetic lineage tracing of endothelial cells in the mouse retina, that vein-derived endothelial tip cells contribute to emerging arteries. Our movies uncover that arterial-fated tip cells change migration direction and migrate backwards within the expanding vascular plexus. This behaviour critically depends on chemokine receptor cxcr4a function. We show that the relevant Cxcr4a ligand Cxcl12a selectively accumulates in newly forming bone tissue even when ubiquitously overexpressed, pointing towards a tissue-intrinsic mode of chemokine gradient formation. Furthermore, we find that cxcr4a mutant cells can contribute to developing arteries when in association with wild-type cells, suggesting collective migration of endothelial cells. Together, our findings reveal specific cell migratory behaviours in the developing blood vessel plexus and uncover a conserved mode of artery formation.


Subject(s)
Arteries/growth & development , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Neovascularization, Physiologic , Receptors, CXCR4/metabolism , Veins/growth & development , Zebrafish Proteins/metabolism , Animal Fins/blood supply , Animal Fins/cytology , Animal Fins/growth & development , Animal Fins/metabolism , Animals , Animals, Genetically Modified , Arteries/cytology , Arteries/metabolism , Cell Lineage/genetics , Cell Movement , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/growth & development , Gene Expression Regulation, Developmental , Mice , Receptors, CXCR4/genetics , Retina/cytology , Retina/growth & development , Retina/metabolism , Signal Transduction , Time-Lapse Imaging , Veins/cytology , Veins/metabolism , Video Recording , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
13.
Dev Cell ; 25(4): 427-34, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23664862

ABSTRACT

At sites of angiogenesis, the expression of the key angiogenesis regulator vascular endothelial growth factor (VEGF) and its main receptor, VEGF receptor 2 (VEGFR-2), are strongly upregulated. Whereas the processes controlling VEGF expression are well described, the mechanisms underlying VEGFR-2 upregulation have remained unclear. We found that endothelial VEGFR-2 expression is strongly reduced in the absence of the G protein G13, resulting in an impaired responsiveness to VEGF-A, a phenotype that can be rescued by normalization of VEGFR-2 levels. G13-mediated VEGFR-2 expression involved activation of the small GTPase RhoA and transcription factor NF-κB, the latter acting via a specific binding site at position -84 of the VEGFR-2 promoter. Mice with endothelial cell-specific loss of G13 showed reduced VEGFR-2 expression at sites of angiogenesis and attenuated VEGF effects, resulting in impaired retinal angiogenesis and tumor vascularization. Taken together, we identified G-protein-mediated signaling via G13 as a critical regulator of VEGFR-2 expression during angiogenesis.


Subject(s)
Endothelium/blood supply , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Alleles , Animals , Cell Line, Tumor , Endothelium/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Human Umbilical Vein Endothelial Cells , Humans , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Neovascularization, Pathologic/genetics , Promoter Regions, Genetic , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Tamoxifen/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
14.
Nat Protoc ; 5(9): 1518-34, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20725067

ABSTRACT

The retina is a powerful experimental system for the analysis of angiogenic blood vessel growth in the postnatal organisms. The three-dimensional architecture of the vessel network and processes as diverse as endothelial cell (EC) proliferation, sprouting, perivascular cell recruitment, vessel remodeling or maturation can be investigated at high resolution. The characterization of physiological and pathological angiogenic processes in mice has been greatly facilitated by inducible and cell type-specific loss-of-function and gain-of-function genetics. In this paper, we provide a detailed protocol for tamoxifen-inducible gene deletion in neonatal mice, as well as for retina dissection, whole-mount immunostaining and the quantitation of EC sprouting and proliferation. These methods have been optimized by our laboratory and yield reliable results. The entire protocol takes approximately 10 d to complete.


Subject(s)
Gene Targeting/methods , Neovascularization, Physiologic , Retinal Vessels/growth & development , Tamoxifen/pharmacology , Animals , Cell Proliferation , Dissection/methods , Endothelial Cells/cytology , Endothelial Cells/physiology , Gene Deletion , Immunohistochemistry/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence/methods , Plant Lectins/analysis , Retina/growth & development , beta-Galactosidase/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...