Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Mol Aspects Med ; 97: 101277, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38788527

ABSTRACT

Excessive accumulation of intermuscular adipose tissue (IMAT) is a common pathological feature in various metabolic and health conditions and can cause muscle atrophy, reduced function, inflammation, insulin resistance, cardiovascular issues, and unhealthy aging. Although IMAT results from fat accumulation in muscle, the mechanisms underlying its onset, development, cellular components, and functions remain unclear. IMAT levels are influenced by several factors, such as changes in the tissue environment, muscle type and origin, extent and duration of trauma, and persistent activation of fibro-adipogenic progenitors (FAPs). FAPs are a diverse and transcriptionally heterogeneous population of stromal cells essential for tissue maintenance, neuromuscular stability, and tissue regeneration. However, in cases of chronic inflammation and pathological conditions, FAPs expand and differentiate into adipocytes, resulting in the development of abnormal and ectopic IMAT. This review discusses the role of FAPs in adipogenesis and how they remodel IMAT. It highlights evidence supporting FAPs and FAP-derived adipocytes as constituents of IMAT, emphasizing their significance in adipose tissue maintenance and development, as well as their involvement in metabolic disorders, chronic pathologies and diseases. We also investigated the intricate molecular pathways and cell interactions governing FAP behavior, adipogenesis, and IMAT accumulation in chronic diseases and muscle deconditioning. Finally, we hypothesize that impaired cellular metabolic flexibility in dysfunctional muscles impacts FAPs, leading to IMAT. A deeper understanding of the biology of IMAT accumulation and the mechanisms regulating FAP behavior and fate are essential for the development of new therapeutic strategies for several debilitating conditions.


Subject(s)
Adipogenesis , Adipose Tissue , Humans , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Stem Cells/metabolism , Stem Cells/cytology , Adipocytes/metabolism , Adipocytes/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Cell Differentiation
2.
Nat Commun ; 14(1): 1980, 2023 04 08.
Article in English | MEDLINE | ID: mdl-37031213

ABSTRACT

Activation of endothelial YAP/TAZ signaling is crucial for physiological and pathological angiogenesis. The mechanisms of endothelial YAP/TAZ regulation are, however, incompletely understood. Here we report that the protocadherin FAT1 acts as a critical upstream regulator of endothelial YAP/TAZ which limits the activity of these transcriptional cofactors during developmental and tumor angiogenesis by promoting their degradation. We show that loss of endothelial FAT1 results in increased endothelial cell proliferation in vitro and in various angiogenesis models in vivo. This effect is due to perturbed YAP/TAZ protein degradation, leading to increased YAP/TAZ protein levels and expression of canonical YAP/TAZ target genes. We identify the E3 ubiquitin ligase Mind Bomb-2 (MIB2) as a FAT1-interacting protein mediating FAT1-induced YAP/TAZ ubiquitination and degradation. Loss of MIB2 expression in endothelial cells in vitro and in vivo recapitulates the effects of FAT1 depletion and causes decreased YAP/TAZ degradation and increased YAP/TAZ signaling. Our data identify a pivotal mechanism of YAP/TAZ regulation involving FAT1 and its associated E3 ligase MIB2, which is essential for YAP/TAZ-dependent angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Trans-Activators , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Proteolysis , Trans-Activators/metabolism , Endothelial Cells/metabolism , YAP-Signaling Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Neovascularization, Pathologic/metabolism , Phosphoproteins/metabolism , Cadherins/metabolism
3.
Development ; 149(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-35050341

ABSTRACT

Angiogenesis is a stepwise process leading to blood vessel formation. In the vertebrate retina, endothelial cells are guided by astrocytes migrating along the inner surface, and the two processes are coupled by a tightly regulated cross-talks between the two cell types. Here, I have investigated how the FAT1 cadherin, a regulator of tissue morphogenesis that governs tissue cross-talk, influences retinal vascular development. Late-onset Fat1 inactivation in the neural lineage in mice, by interfering with astrocyte progenitor migration polarity and maturation, delayed postnatal retinal angiogenesis, leading to persistent vascular abnormalities in adult retinas. Impaired astrocyte migration and polarity were not associated with alterations of retinal ganglion cell axonal trajectories or of the inner limiting membrane. In contrast, inducible Fat1 ablation in postnatal astrocytes was sufficient to alter their migration polarity and proliferation. Altogether, this study uncovers astrocyte-intrinsic and -extrinsic Fat1 activities that influence astrocyte migration polarity, proliferation and maturation, disruption of which impacts retinal vascular development and maintenance.


Subject(s)
Astrocytes/metabolism , Cadherins/metabolism , Neovascularization, Physiologic , Retina/metabolism , Animals , Astrocytes/cytology , Cadherins/genetics , Cell Differentiation , Mice , Retina/cytology , Retina/growth & development , Retinal Vessels/growth & development
4.
Nature ; 589(7842): 448-455, 2021 01.
Article in English | MEDLINE | ID: mdl-33328637

ABSTRACT

FAT1, which encodes a protocadherin, is one of the most frequently mutated genes in human cancers1-5. However, the role and the molecular mechanisms by which FAT1 mutations control tumour initiation and progression are poorly understood. Here, using mouse models of skin squamous cell carcinoma and lung tumours, we found that deletion of Fat1 accelerates tumour initiation and malignant progression and promotes a hybrid epithelial-to-mesenchymal transition (EMT) phenotype. We also found this hybrid EMT state in FAT1-mutated human squamous cell carcinomas. Skin squamous cell carcinomas in which Fat1 was deleted presented increased tumour stemness and spontaneous metastasis. We performed transcriptional and chromatin profiling combined with proteomic analyses and mechanistic studies, which revealed that loss of function of FAT1 activates a CAMK2-CD44-SRC axis that promotes YAP1 nuclear translocation and ZEB1 expression that stimulates the mesenchymal state. This loss of function also inactivates EZH2, promoting SOX2 expression, which sustains the epithelial state. Our comprehensive analysis identified drug resistance and vulnerabilities in FAT1-deficient tumours, which have important implications for cancer therapy. Our studies reveal that, in mouse and human squamous cell carcinoma, loss of function of FAT1 promotes tumour initiation, progression, invasiveness, stemness and metastasis through the induction of a hybrid EMT state.


Subject(s)
Cadherins/deficiency , Epithelial-Mesenchymal Transition/genetics , Gene Deletion , Neoplasm Metastasis/genetics , Neoplasms/genetics , Neoplasms/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Disease Progression , Enhancer of Zeste Homolog 2 Protein/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mesoderm/metabolism , Mesoderm/pathology , Mice , Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Phosphoproteins/analysis , Phosphoproteins/metabolism , Proteomics , SOXB1 Transcription Factors/metabolism , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factors/metabolism , YAP-Signaling Proteins , Zinc Finger E-box-Binding Homeobox 1/metabolism , src-Family Kinases/metabolism
5.
JCI Insight ; 5(23)2020 10 27.
Article in English | MEDLINE | ID: mdl-33108146

ABSTRACT

Pituitary developmental defects lead to partial or complete hormone deficiency and significant health problems. The majority of cases are sporadic and of unknown cause. We screened 28 patients with pituitary stalk interruption syndrome (PSIS) for mutations in the FAT/DCHS family of protocadherins that have high functional redundancy. We identified seven variants, four of which putatively damaging, in FAT2 and DCHS2 in six patients with pituitary developmental defects recruited through a cohort of patients with mostly ectopic posterior pituitary gland and/or pituitary stalk interruption. All patients had growth hormone deficiency and two presented with multiple hormone deficiencies and small glands. FAT2 and DCHS2 were strongly expressed in the mesenchyme surrounding the normal developing human pituitary. We analyzed Dchs2-/- mouse mutants and identified anterior pituitary hypoplasia and partially penetrant infundibular defects. Overlapping infundibular abnormalities and distinct anterior pituitary morphogenesis defects were observed in Fat4-/- and Dchs1-/- mouse mutants but all animal models displayed normal commitment to the anterior pituitary cell type. Together our data implicate FAT/DCHS protocadherins in normal hypothalamic-pituitary development and identify FAT2 and DCHS2 as candidates underlying pituitary gland developmental defects such as ectopic pituitary gland and/or pituitary stalk interruption.


Subject(s)
Cadherin Related Proteins/genetics , Cadherins/genetics , Pituitary Diseases/genetics , Adolescent , Animals , Cadherin Related Proteins/metabolism , Cadherins/metabolism , Female , Humans , Hypothalamus/growth & development , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Pituitary Gland/growth & development , Pituitary Gland/metabolism , Young Adult
6.
Semin Cell Dev Biol ; 104: 14-30, 2020 08.
Article in English | MEDLINE | ID: mdl-32517852

ABSTRACT

For decades, limb development has been a paradigm of three-dimensional patterning. Moreover, as the limb muscles and the other tissues of the limb's musculoskeletal system arise from distinct developmental sources, it has been a prime example of integrative morphogenesis and cross-tissue communication. As the limbs grow, all components of the musculoskeletal system (muscles, tendons, connective tissue, nerves) coordinate their growth and differentiation, ultimately giving rise to a functional unit capable of executing elaborate movement. While the molecular mechanisms governing global three-dimensional patterning and formation of the skeletal structures of the limbs has been a matter of intense research, patterning of the soft tissues is less understood. Here, we review the development of limb muscles with an emphasis on their interaction with other tissue types and the instructive roles these tissues play. Furthermore, we discuss the role of adult correlates of these embryonic accessory tissues in muscle regeneration.


Subject(s)
Extremities/embryology , Muscle Development , Muscle, Skeletal/embryology , Regeneration , Animals , Cell Differentiation , Humans , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism
7.
Front Genet ; 10: 178, 2019.
Article in English | MEDLINE | ID: mdl-30923534

ABSTRACT

Members of the PEA3 transcription factors are emerging as bone fide targets for fibroblast growth factor (FGF) signaling. Among them, ETV4 and ETV5 appear to mediate FGF10 signaling during early embryonic lung development. In this paper, recently obtained Tg(Etv4-GFP) and Etv5 CreERT2-RFP fluorescent reporter lines were generally characterized during early embryonic development and in the context of FGF10 signaling, in particular. We found that both Tg(Etv4-GFP) and Etv5 CreERT2-RFP were primarily expressed in the epithelium of the lung during embryonic development. However, the expression of Etv5 CreERT2-RFP was much higher than that of Tg(Etv4-GFP), and continued to increase during development, whereas Tg(Etv4-GFP) decreased. The expression patterns of the surrogate fluorescent protein GFP and RFP for ETV4 and ETV5, respectively, agreed with known regions of FGF10 signaling in various developing organs, including the lung, where ETV4-GFP was seen primarily in the distal epithelium and to a lesser extent in the surrounding mesenchyme. As expected, ETV5-RFP was restricted to the lung epithelium, showing a decreasing expression pattern from distal buds to proximal conducting airways. FGF10 inhibition experiments confirmed that both Etv4 and Etv5 are downstream of FGF10 signaling. Finally, we also validated that both fluorescent reporters responded to FGF10 inhibition in vitro. In conclusion, these two reporter lines appear to be promising tools to monitor FGF10/FGFR2b signaling in early lung development. These tools will have to be further validated at later stages and in other organs of interest.

8.
Nat Commun ; 9(1): 3691, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209249

ABSTRACT

Spiral ganglion (SG) neurons of the cochlea convey all auditory inputs to the brain, yet the cellular and molecular complexity necessary to decode the various acoustic features in the SG has remained unresolved. Using single-cell RNA sequencing, we identify four types of SG neurons, including three novel subclasses of type I neurons and the type II neurons, and provide a comprehensive genetic framework that define their potential synaptic communication patterns. The connectivity patterns of the three subclasses of type I neurons with inner hair cells and their electrophysiological profiles suggest that they represent the intensity-coding properties of auditory afferents. Moreover, neuron type specification is already established at birth, indicating a neuronal diversification process independent of neuronal activity. Thus, this work provides a transcriptional catalog of neuron types in the cochlea, which serves as a valuable resource for dissecting cell-type-specific functions of dedicated afferents in auditory perception and in hearing disorders.


Subject(s)
Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Cochlea/cytology , Cochlea/metabolism , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Inner/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Spiral Ganglion/cytology , Spiral Ganglion/metabolism , Synaptic Potentials/physiology
9.
PLoS Biol ; 16(5): e2004734, 2018 05.
Article in English | MEDLINE | ID: mdl-29768404

ABSTRACT

Muscle morphogenesis is tightly coupled with that of motor neurons (MNs). Both MNs and muscle progenitors simultaneously explore the surrounding tissues while exchanging reciprocal signals to tune their behaviors. We previously identified the Fat1 cadherin as a regulator of muscle morphogenesis and showed that it is required in the myogenic lineage to control the polarity of progenitor migration. To expand our knowledge on how Fat1 exerts its tissue-morphogenesis regulator activity, we dissected its functions by tissue-specific genetic ablation. An emblematic example of muscle under such morphogenetic control is the cutaneous maximus (CM) muscle, a flat subcutaneous muscle in which progenitor migration is physically separated from the process of myogenic differentiation but tightly associated with elongating axons of its partner MNs. Here, we show that constitutive Fat1 disruption interferes with expansion and differentiation of the CM muscle, with its motor innervation and with specification of its associated MN pool. Fat1 is expressed in muscle progenitors, in associated mesenchymal cells, and in MN subsets, including the CM-innervating pool. We identify mesenchyme-derived connective tissue (CT) as a cell type in which Fat1 activity is required for the non-cell-autonomous control of CM muscle progenitor spreading, myogenic differentiation, motor innervation, and for motor pool specification. In parallel, Fat1 is required in MNs to promote their axonal growth and specification, indirectly influencing muscle progenitor progression. These results illustrate how Fat1 coordinates the coupling of muscular and neuronal morphogenesis by playing distinct but complementary actions in several cell types.


Subject(s)
Cadherins/physiology , Morphogenesis , Motor Neurons/physiology , Muscles/embryology , Neuromuscular Junction/embryology , Animals , Connective Tissue/metabolism , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Mesoderm/physiology , Mice , Mice, Knockout , Muscles/innervation , Pregnancy , Receptor, Platelet-Derived Growth Factor alpha/metabolism
10.
Oncotarget ; 7(46): 74747-74767, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27732969

ABSTRACT

The cytoplasmic tyrosine kinase ABL exerts positive or negative effects in solid tumours according to the cellular context, thus functioning as a "switch modulator". The therapeutic effects of drugs targeting a set of signals encompassing ABL have been explored in several solid tumours. However, the net contribution of ABL inhibition by these agents remains elusive as these drugs also act on other signalling components. Here, using glioblastoma (GBM) as a cellular paradigm, we report that ABL inhibition exacerbates mesenchymal features as highlighted by down-regulation of epithelial markers and up-regulation of mesenchymal markers. Cells with permanent ABL inhibition exhibit enhanced motility and invasive capabilities, while proliferation and tumorigenic properties are reduced. Intriguingly, permanent ABL inhibition also interferes with GBM neurosphere formation and with expression of stemness markers in sphere-cultured GBM cells. Furthermore, we show that the molecular and biological characteristics of GBM cells with impaired ABL are reversible by restoring ABL levels, thus uncovering a remarkable plasticity of GBM cells to ABL threshold. A phospho-signalling screen revealed that loss of tumorigenic and self-renewal properties in GBM cells under permanent ABL inhibition coincide with drastic changes in the expression and/or phosphorylation levels of multiple signalling components. Our findings identify ABL as a crucial player for migration, invasion, proliferation, tumorigenic, and stem-cell like properties of GBM cells. Taken together, this work supports the notion that the oncogenic role of ABL in GBM cells is associated with its capability to coordinate a signalling setting that determines tumorigenic and stem-cell like properties.


Subject(s)
Brain Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Glioblastoma/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction , Biomarkers , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cell Self Renewal/genetics , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-abl/genetics
11.
PLoS Genet ; 11(9): e1005533, 2015.
Article in English | MEDLINE | ID: mdl-26393505

ABSTRACT

The successive events that cells experience throughout development shape their intrinsic capacity to respond and integrate RTK inputs. Cellular responses to RTKs rely on different mechanisms of regulation that establish proper levels of RTK activation, define duration of RTK action, and exert quantitative/qualitative signalling outcomes. The extent to which cells are competent to deal with fluctuations in RTK signalling is incompletely understood. Here, we employ a genetic system to enhance RTK signalling in a tissue-specific manner. The chosen RTK is the hepatocyte growth factor (HGF) receptor Met, an appropriate model due to its pleiotropic requirement in distinct developmental events. Ubiquitously enhanced Met in Cre/loxP-based Rosa26(stopMet) knock-in context (Del-R26(Met)) reveals that most tissues are capable of buffering enhanced Met-RTK signalling thus avoiding perturbation of developmental programs. Nevertheless, this ubiquitous increase of Met does compromise selected programs such as myoblast migration. Using cell-type specific Cre drivers, we genetically showed that altered myoblast migration results from ectopic Met expression in limb mesenchyme rather than in migrating myoblasts themselves. qRT-PCR analyses show that ectopic Met in limbs causes molecular changes such as downregulation in the expression levels of Notum and Syndecan4, two known regulators of morphogen gradients. Molecular and functional studies revealed that ectopic Met expression in limb mesenchyme does not alter HGF expression patterns and levels, but impairs HGF bioavailability. Together, our findings show that myoblasts, in which Met is endogenously expressed, are capable of buffering increased RTK levels, and identify mesenchymal cells as a cell type vulnerable to ectopic Met-RTK signalling. These results illustrate that embryonic cells are sensitive to alterations in the spatial distribution of RTK action, yet resilient to fluctuations in signalling levels of an RTK when occurring in its endogenous domain of activity.


Subject(s)
Embryonic Development/genetics , Hepatocyte Growth Factor/genetics , Myoblasts/metabolism , Proto-Oncogene Proteins c-met/genetics , Animals , Cell Movement/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental , Hepatocyte Growth Factor/metabolism , Mice , Phosphorylation , Proto-Oncogene Proteins c-met/biosynthesis , Signal Transduction , Syndecan-4/biosynthesis , Syndecan-4/genetics
12.
Development ; 142(15): 2564-73, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26116661

ABSTRACT

Regulation of the balance between progenitor self-renewal and differentiation is crucial to development. In the mammalian kidney, reciprocal signalling between three lineages (stromal, mesenchymal and ureteric) ensures correct nephron progenitor self-renewal and differentiation. Loss of either the atypical cadherin FAT4 or its ligand Dachsous 1 (DCHS1) results in expansion of the mesenchymal nephron progenitor pool, called the condensing mesenchyme (CM). This has been proposed to be due to misregulation of the Hippo kinase pathway transcriptional co-activator YAP. Here, we use tissue-specific deletions to prove that FAT4 acts non-autonomously in the renal stroma to control nephron progenitors. We show that loss of Yap from the CM in Fat4-null mice does not reduce the expanded CM, indicating that FAT4 regulates the CM independently of YAP. Analysis of Six2(-/-);Fat4(-/-) double mutants demonstrates that excess progenitors in Fat4 mutants are dependent on Six2, a crucial regulator of nephron progenitor self-renewal. Electron microscopy reveals that cell organisation is disrupted in Fat4 mutants. Gene expression analysis demonstrates that the expression of Notch and FGF pathway components are altered in Fat4 mutants. Finally, we show that Dchs1, and its paralogue Dchs2, function in a partially redundant fashion to regulate the number of nephron progenitors. Our data support a model in which FAT4 in the stroma binds to DCHS1/2 in the mouse CM to restrict progenitor self-renewal.


Subject(s)
Cadherins/metabolism , Cell Differentiation/physiology , Nephrons/ultrastructure , Signal Transduction/physiology , Stem Cells/cytology , Adaptor Proteins, Signal Transducing/metabolism , Analysis of Variance , Animals , Cell Cycle Proteins , Cell Lineage/physiology , Fluorescent Antibody Technique , Gene Expression Profiling , Hippo Signaling Pathway , Immunoblotting , In Situ Hybridization , In Situ Nick-End Labeling , Mice , Mice, Knockout , Microscopy, Electron , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Sequence Analysis, RNA , YAP-Signaling Proteins
13.
Ann Neurol ; 78(3): 387-400, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26018399

ABSTRACT

OBJECTIVE: Facioscapulohumeral muscular dystrophy (FSHD) is linked to either contraction of D4Z4 repeats on chromosome 4 or to mutations in the SMCHD1 gene, both of which result in the aberrant expression of the transcription factor DUX4. However, it is still difficult to correlate these genotypes with the phenotypes observed in patients. Because we have recently shown that mice with disrupted Fat1 functions exhibit FSHD-like phenotypes, we have investigated the expression of the human FAT1 gene in FSHD. METHODS: We first analyzed FAT1 expression in FSHD adult muscles and determined whether FAT1 expression was driven by DUX4. We next determined FAT1 expression levels in 64 muscles isolated from 16 control fetuses. These data were further complemented with analysis of Fat1 expression in developing mouse embryos. RESULTS: We demonstrated that FAT1 expression is independent of DUX4. Moreover, we observed that (1) in control fetal human biopsies or in developing mouse embryos, FAT1 is expressed at lower levels in muscles that are affected at early stages of FSHD progression than in muscles that are affected later or are nonaffected; and (2) in adult muscle biopsies, FAT1 expression is lower in FSHD muscles compared to control muscles. INTERPRETATION: We propose a revised model for FSHD in which FAT1 levels might play a role in determining which muscles will exhibit early and late disease onset, whereas DUX4 may worsen the muscle phenotype.


Subject(s)
Cadherins/biosynthesis , Gene Expression Regulation, Developmental , Muscular Dystrophy, Facioscapulohumeral/diagnosis , Muscular Dystrophy, Facioscapulohumeral/metabolism , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , Adult , Animals , Cells, Cultured , Female , Fetus , Humans , Male , Mice , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Quadriceps Muscle/embryology
14.
Hum Mutat ; 36(4): 443-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25615407

ABSTRACT

Facioscapulohumeralmuscular dystrophy (FSHD) is linked to copy-number reduction (N < 10) of the 4q D4Z4 subtelomeric array, in association with DUX4-permissive haplotypes. This main form is indicated as FSHD1. FSHD-like phenotypes may also appear in the absence of D4Z4 copy-number reduction. Variants of the SMCHD1 gene have been reported to associate with D4Z4 hypomethylation in DUX4-compatible haplotypes, thus defining FSHD2. Recently, mice carrying a muscle-specific knock-out of the protocadherin gene Fat1 or its constitutive hypomorphic allele were shown to develop muscular and nonmuscular defects mimicking human FSHD. Here, we report FAT1 variants in a group of patients presenting with neuromuscular symptoms reminiscent of FSHD. The patients do not carry D4Z4 copy-number reduction, 4q hypomethylation, or SMCHD1 variants. However, abnormal splicing of the FAT1 transcript is predicted for all identified variants. To determine their pathogenicity, we elaborated a minigene approach coupled to an antisense oligonucleotide (AON) assay. In vitro, four out of five selected variants induced partial or complete alteration of splicing by creating new splice sites or modifying splicing regulators. AONs confirmed these effects. Altered transcripts may affect FAT1 protein interactions or stability. Altogether, our data suggest that defective FAT1 is associated with an FSHD-like phenotype.


Subject(s)
Cadherins/genetics , Chromosomes, Human, Pair 4 , Genetic Variation , Muscular Dystrophy, Facioscapulohumeral/diagnosis , Muscular Dystrophy, Facioscapulohumeral/genetics , Phenotype , Adolescent , Adult , Aged , Alleles , Alternative Splicing , Child , Child, Preschool , DNA Methylation , Exons , Gene Expression , Genes, Reporter , Humans , Infant , Infant, Newborn , Middle Aged , Mutation , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Young Adult
15.
Nat Neurosci ; 17(9): 1171-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108913

ABSTRACT

The cadherin Celsr3 regulates the directional growth and targeting of axons in the CNS, but whether it acts in collaboration with or in parallel to other guidance cues is unknown. Furthermore, the function of Celsr3 in the peripheral nervous system is still largely unexplored. Here we show that Celsr3 mediates pathfinding of motor axons innervating the hindlimb. In mice, Celsr3-deficient axons of the peroneal nerve segregate from those of the tibial nerve but fail to extend dorsally, and they stall near the branch point. Mutant axons respond to repulsive ephrinA-EphA forward signaling and glial cell-derived neurotrophic factor (GDNF). However, they are insensitive to attractive EphA-ephrinA reverse signaling. In transfected cells, Celsr3 immunoprecipitates with ephrinA2, ephrinA5, Ret, GDNF family receptor α1 (GFRα1) and Frizzled3 (Fzd3). The function of Celsr3 is Fzd3 dependent but Vangl2 independent. Our results provide evidence that the Celsr3-Fzd3 pathway interacts with EphA-ephrinA reverse signaling to guide motor axons in the hindlimb.


Subject(s)
Axons/physiology , Cadherins/genetics , Hindlimb/innervation , Motor Neurons/physiology , Peroneal Nerve/physiology , Receptors, Cell Surface/genetics , Tibial Nerve/physiology , Animals , Cadherins/metabolism , Cells, Cultured , Clubfoot/embryology , Clubfoot/genetics , Ephrin-A2/metabolism , Ephrin-A5/metabolism , Female , Frizzled Receptors/metabolism , Green Fluorescent Proteins/genetics , HEK293 Cells , Hindlimb/abnormalities , Humans , Male , Mice, Knockout , Motor Neurons/ultrastructure , Peroneal Nerve/cytology , Peroneal Nerve/embryology , Pregnancy , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Tibial Nerve/cytology , Tibial Nerve/embryology
16.
BMC Biol ; 12: 56, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25124859

ABSTRACT

BACKGROUND: Multiple growth factors are known to control several aspects of neuronal biology, consecutively acting as morphogens to diversify neuronal fates, as guidance cues for axonal growth, and as modulators of survival or death to regulate neuronal numbers. The multiplicity of neuronal types is permitted by the combinatorial usage of growth factor receptors, each of which is expressed in distinct and overlapping subsets of neurons, and by the multitasking role of growth factor receptors, which recruit multiple signalling cascades differentially required for distinct biological outcomes. We have explored signalling robustness in cells where a given receptor tyrosine kinase (RTK) elicits qualitatively distinct outcomes. As the HGF/Met system regulates several biological responses in motor neurons (MN) during neuromuscular development, we have investigated the signalling modalities through which the HGF/Met system impacts on MN biology, and the degree of robustness of each of these functions, when challenged with substitutions of signalling pathways. RESULTS: Using a set of mouse lines carrying signalling mutations that change the Met phosphotyrosine binding preferences, we have asked whether distinct functions of Met in several MN subtypes require specific signalling pathways, and to which extent signalling plasticity allows a pleiotropic system to exert distinct developmental outcomes. The differential ability of signalling mutants to promote muscle migration versus axonal growth allowed us to uncouple an indirect effect of HGF/Met signalling on nerve growth through the regulation of muscle size from a direct regulation of motor growth via the PI3 kinase (PI3K), but not Src kinase, pathway. Furthermore, we found that HGF/Met-triggered expansion of Pea3 expression domain in the spinal cord can be accomplished through several alternative signalling cascades, differentially sensitive to the Pea3 dosage. Finally, we show that the regulation of MN survival by HGF/Met can equally be achieved in vitro and in vivo by alternative signalling cascades involving either PI3K-Akt or Src and Mek pathways. CONCLUSIONS: Our findings distinguish MN survival and fate specification, as RTK-triggered responses allowing substitutions of the downstream signalling routes, from nerve growth patterning, which depends on a selective, non-substitutable pathway.


Subject(s)
Body Patterning , Motor Neurons/physiology , Signal Transduction , Animals , Axons/physiology , Cells, Cultured , Embryo, Mammalian , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Transcription Factors/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
17.
PLoS Genet ; 9(6): e1003550, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23785297

ABSTRACT

Generation of skeletal muscles with forms adapted to their function is essential for normal movement. Muscle shape is patterned by the coordinated polarity of collectively migrating myoblasts. Constitutive inactivation of the protocadherin gene Fat1 uncoupled individual myoblast polarity within chains, altering the shape of selective groups of muscles in the shoulder and face. These shape abnormalities were followed by early onset regionalised muscle defects in adult Fat1-deficient mice. Tissue-specific ablation of Fat1 driven by Pax3-cre reproduced muscle shape defects in limb but not face muscles, indicating a cell-autonomous contribution of Fat1 in migrating muscle precursors. Strikingly, the topography of muscle abnormalities caused by Fat1 loss-of-function resembles that of human patients with facioscapulohumeral dystrophy (FSHD). FAT1 lies near the critical locus involved in causing FSHD, and Fat1 mutant mice also show retinal vasculopathy, mimicking another symptom of FSHD, and showed abnormal inner ear patterning, predictive of deafness, reminiscent of another burden of FSHD. Muscle-specific reduction of FAT1 expression and promoter silencing was observed in foetal FSHD1 cases. CGH array-based studies identified deletion polymorphisms within a putative regulatory enhancer of FAT1, predictive of tissue-specific depletion of FAT1 expression, which preferentially segregate with FSHD. Our study identifies FAT1 as a critical determinant of muscle form, misregulation of which associates with FSHD.


Subject(s)
Cadherins/genetics , Muscle Development/genetics , Muscles/physiopathology , Muscular Dystrophy, Facioscapulohumeral/genetics , Adult , Animals , Cadherins/metabolism , Cell Differentiation/genetics , Cells, Cultured , Humans , Mice , Muscles/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts/metabolism , Myoblasts/pathology , Oligonucleotide Array Sequence Analysis , Organ Specificity
18.
Neuron ; 75(6): 1051-66, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22998873

ABSTRACT

The Neurotrophic factor gdnf plays diverse developmental roles, supporting survival and also acting as a chemoattractant for axon and cell migration. We report that in the developing spinal cord, a focal source of gdnf is present in the floor plate (FP) where commissural axons cross the midline. Gdnf has no direct guidance properties but switches on the responsiveness of crossing commissural growth cones to the midline repellent Semaphorin3B by suppressing calpain-mediated processing of the Sema3B signaling coreceptor Plexin-A1. Analysis of single and double mutant mouse models indicates that although gdnf is the principal trigger of Sema3B midline repulsion, it acts with another FP cue, NrCAM. Finally, genetic and in vitro experiments provide evidence that this gdnf effect is RET independent and mediated by NCAM/GFRα1 signaling. This study identifies a regulator of midline crossing and reveals interplays between Semaphorin and gdnf signaling during axon guidance.


Subject(s)
Axons/physiology , Gene Expression Regulation, Developmental/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurons/cytology , Semaphorins/metabolism , Analysis of Variance , Animals , Axons/drug effects , Body Patterning/genetics , Calpain/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Glial Cell Line-Derived Neurotrophic Factor/deficiency , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/genetics , Neurons/drug effects , Receptors, Cell Surface/metabolism , Semaphorins/genetics , Spinal Cord/cytology , Spinal Cord/embryology , Transfection
19.
J Neurosci ; 31(31): 11144-58, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21813676

ABSTRACT

The precise control of motor neuron (MN) death and survival following initial innervation of skeletal muscle targets is a key step in sculpting a functional motor system, but how this is regulated at the level of individual motor pools remains unclear. Hepatocyte growth factor (HGF) and its receptor Met play key developmental roles in both muscle and MNs. We generated mice (termed "Nes-Met") in which met is inactivated from midembryonic stages onward in the CNS only. Adult animals showed motor behavioral defects suggestive of impaired innervation of pectoral muscles. Correspondingly, in neonatal spinal cords of Nes-Met mutants, we observed death of a discrete population of pea3-expressing MNs at brachial levels. Axonal tracing using pea3 reporter mice revealed a novel target muscle of pea3-expressing MNs: the pectoralis minor muscle. In Nes-Met mice, the pectoralis minor pool initially innervated its target muscle, but required HGF/Met for survival, hence for proper maintenance of muscle innervation. In contrast, HGF/Met was dispensable for the survival of neighboring Met-expressing MN pools, despite its earlier functions for their specification and axon growth. Our results demonstrate the exquisite degree to which outcomes of signaling by receptor tyrosine kinases are regulated on a cell-by-cell basis. They also provide a model for one way in which the multiplicity of neurotrophic factors may allow for regulation of MN numbers in a pool-specific manner.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Motor Neurons/drug effects , Motor Neurons/physiology , Nerve Growth Factors/pharmacology , Animals , Animals, Newborn , Axons/physiology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Dose-Response Relationship, Drug , Embryo, Mammalian , Ganglia, Spinal/cytology , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Hand Strength/physiology , Hepatocyte Growth Factor/genetics , Intermediate Filament Proteins/genetics , Male , Mice , Mice, Transgenic , Models, Biological , Motor Activity/genetics , Muscle Strength/genetics , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Mutation/genetics , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nestin , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-met/genetics , Psychomotor Performance/physiology , Reaction Time , Rotarod Performance Test , Spinal Cord/cytology , Transcription Factors/genetics
20.
J Neurosci ; 30(37): 12414-23, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20844136

ABSTRACT

Nociceptors in peripheral ganglia display a remarkable functional heterogeneity. They can be divided into the following two major classes: peptidergic and nonpeptidergic neurons. Although RUNX1 has been shown to play a pivotal role in the specification of nonpeptidergic neurons, the mechanisms driving peptidergic differentiation remain elusive. Here, we show that hepatocyte growth factor (HGF)-Met signaling acts synergistically with nerve growth factor-tyrosine kinase receptor A to promote peptidergic identity in a subset of prospective nociceptors. We provide in vivo evidence that a population of peptidergic neurons, derived from the RUNX1 lineage, require Met activity for the proper extinction of Runx1 and optimal activation of CGRP (calcitonin gene-related peptide). Moreover, we show that RUNX1 in turn represses Met expression in nonpeptidergic neurons, revealing a bidirectional cross talk between Met and RUNX1. Together, our novel findings support a model in which peptidergic versus nonpeptidergic specification depends on a balance between HGF-Met signaling and Runx1 extinction/maintenance.


Subject(s)
Cell Differentiation/physiology , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/physiology , Hepatocyte Growth Factor/physiology , Nociceptors/metabolism , Proto-Oncogene Proteins c-met/physiology , Signal Transduction/physiology , Animals , Cell Lineage/physiology , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/biosynthesis , Ganglia, Spinal/cytology , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Neurological , Neuropeptides/physiology , Nociceptors/cytology , Proto-Oncogene Proteins c-met/deficiency , Proto-Oncogene Proteins c-met/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...