Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
Add more filters

Publication year range
1.
J Cell Sci ; 137(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38224152

ABSTRACT

Adult muscle stem cells (MuSCs) are critical for muscle homeostasis and regeneration, and their behavior relies on a finely regulated niche made of specific extracellular matrix (ECM) components and soluble factors. Among ECM proteins, collagen VI (Col6) influences the mechanical properties of the niche and, in turn, MuSC self-renewal capabilities. Here, we investigated whether Col6 can exert a direct function as a biochemical signal for regulating the stemness and differentiation of murine MuSCs and myoblasts. Native Col6, but not its pepsin-resistant fragment, counteracts the early differentiation of myogenic cells by reducing the expression of differentiation marker genes and preserving stemness features, with inhibition of the canonical Wnt pathway. Our data indicate that extracellular Col6 acts as a soluble ligand in delaying early myogenic differentiation by regulating intracellular signals involved in adult myogenesis.


Subject(s)
Collagen , Satellite Cells, Skeletal Muscle , Mice , Animals , Cell Differentiation , Collagen/metabolism , Muscles , Muscle Development/genetics , Muscle, Skeletal/metabolism
2.
Am J Hum Genet ; 109(12): 2230-2252, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36351433

ABSTRACT

EMILIN1 (elastin-microfibril-interface-located-protein-1) is a structural component of the elastic fiber network and localizes to the interface between the fibrillin microfibril scaffold and the elastin core. How EMILIN1 contributes to connective tissue integrity is not fully understood. Here, we report bi-allelic EMILIN1 loss-of-function variants causative for an entity combining cutis laxa, arterial tortuosity, aneurysm formation, and bone fragility, resembling autosomal-recessive cutis laxa type 1B, due to EFEMP2 (FBLN4) deficiency. In both humans and mice, absence of EMILIN1 impairs EFEMP2 extracellular matrix deposition and LOX activity resulting in impaired elastogenesis, reduced collagen crosslinking, and aberrant growth factor signaling. Collagen fiber ultrastructure and histopathology in EMILIN1- or EFEMP2-deficient skin and aorta corroborate these findings and murine Emilin1-/- femora show abnormal trabecular bone formation and strength. Altogether, EMILIN1 connects elastic fiber network with collagen fibril formation, relevant for both bone and vascular tissue homeostasis.


Subject(s)
Bone Diseases, Metabolic , Cutis Laxa , Animals , Humans , Mice , Collagen/genetics , Cutis Laxa/genetics , Elastin/metabolism , Extracellular Matrix Proteins/metabolism
3.
J Pathol ; 260(4): 417-430, 2023 08.
Article in English | MEDLINE | ID: mdl-37272555

ABSTRACT

Despite a number of studies providing evidence that the extracellular matrix (ECM) is an active player in the pathogenesis of intestinal inflammation, knowledge on the actual contribution of specific ECM molecules in the progression of inflammatory bowel disease (IBD) remains scant. Here, we investigated the role of a major ECM protein, collagen VI (ColVI), in gut homeostasis and elucidated the impact of its deregulation on the pathophysiology of IBD. To this end, we combined in vivo and ex vivo studies on wild type and ColVI-deficient (Col6a1-/- ) mice both under physiological conditions and during experimentally induced acute colitis and its subsequent recovery, by means of gut histology and immunostaining, gene expression, bone marrow transplantation, flow cytometry of immune cell subpopulations, and lymph flow assessment. We found that ColVI displayed dynamic expression and ECM deposition during the acute inflammatory and recovery phases of experimentally induced colitis, whereas the genetic ablation of ColVI in Col6a1 null mice impaired the functionality of lymphatic vessels, which in turn affected the resolution of inflammation during colitis. Based on these findings, we investigated ColVI expression and deposition in ileal specimens from two cohorts of patients affected by Crohn's disease (CD) and correlated ColVI abundance to clinical outcome. Our results show that high ColVI immunoreactivity in ileal biopsies of CD patients at diagnosis correlates with increased risk of surgery and that ColVI expression in biopsies taken at the resection margin during surgery, and showing inactive disease, predict disease recurrence. Our data unveil a key role for ColVI in the intestinal microenvironment, where it is involved in lymphangiogenesis and intestinal inflammation. Altogether, these findings point at the dysregulation of ColVI expression as a novel factor contributing to the onset and maintenance of inflammation in CD via mechanisms impinging on the modulation of inflammatory cell recruitment and function. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Colitis , Crohn Disease , Inflammatory Bowel Diseases , Animals , Mice , Lymphangiogenesis , Collagen Type VI/genetics , Colitis/chemically induced , Colitis/genetics , Mice, Knockout , Inflammation , Drainage
4.
Cell Mol Life Sci ; 80(8): 233, 2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37505240

ABSTRACT

Microenvironmental factors are known fundamental regulators of the phenotype and aggressiveness of glioblastoma (GBM), the most lethal brain tumor, characterized by fast progression and marked resistance to treatments. In this context, the extracellular matrix (ECM) is known to heavily influence the behavior of cancer cells from several origins, contributing to stem cell niches, influencing tumor invasiveness and response to chemotherapy, mediating survival signaling cascades, and modulating inflammatory cell recruitment. Here, we show that collagen VI (COL6), an ECM protein widely expressed in both normal and pathological tissues, has a distinctive distribution within the GBM mass, strongly correlated with the most aggressive and phenotypically immature cells. Our data demonstrate that COL6 sustains the stem-like properties of GBM cells and supports the maintenance of an aggressive transcriptional program promoting cancer cell proliferation and survival. In particular, we identified a specific subset of COL6-transcriptionally co-regulated genes, required for the response of cells to replicative stress and DNA damage, supporting the concept that COL6 is an essential stimulus for the activation of GBM cell response and resistance to chemotherapy, through the ATM/ATR axis. Altogether, these findings indicate that COL6 plays a pivotal role in GBM tumor biology, exerting a pleiotropic action across different GBM hallmarks, including phenotypic identity and gene transcription, as well as response to treatments, thus providing valuable information for the understanding of the complex microenvironmental cues underlying GBM malignancy.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Collagen/metabolism , Signal Transduction , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Neoplastic Stem Cells/metabolism
5.
Biol Res ; 56(1): 19, 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37106439

ABSTRACT

BACKGROUND: AMBRA1 is an intrinsically disordered protein, working as a scaffold molecule to coordinate, by protein-protein interaction, many cellular processes, including autophagy, mitophagy, apoptosis and cell cycle progression. The zebrafish genome contains two ambra1 paralogous genes (a and b), both involved in development and expressed at high levels in the gonads. Characterization of the zebrafish paralogous genes mutant lines generated by CRISPR/Cas9 approach showed that ambra1b knockout leads to an all-male population. RESULTS: We demonstrated that the silencing of the ambra1b gene determines a reduction of primordial germ cells (PGCs), a condition that, in the zebrafish, leads to the development of all-male progeny. PGC reduction was confirmed by knockdown experiments and rescued by injection of ambra1b and human AMBRA1 mRNAs, but not ambra1a mRNA. Moreover, PGC loss was not rescued by injection with human AMBRA1 mRNA mutated in the CUL4-DDB1 binding region, thus suggesting that interaction with this complex is involved in PGC protection from loss. Results from zebrafish embryos injected with murine Stat3 mRNA and stat3 morpholino suggest that Ambra1b could indirectly regulate this protein through CUL4-DDB1 interaction. According to this, Ambra1+/- mice showed a reduced Stat3 expression in the ovary together with a low number of antral follicles and an increase of atretic follicles, indicating a function of Ambra1 in the ovary of mammals as well. Moreover, in agreement with the high expression of these genes in the testis and ovary, we found significant impairment of the reproductive process and pathological alterations, including tumors, mainly limited to the gonads. CONCLUSIONS: By exploiting ambra1a and ambra1b knockout zebrafish lines, we prove the sub-functionalization between the two paralogous zebrafish genes and uncover a novel function of Ambra1 in the protection from excessive PGC loss, which seems to require binding with the CUL4-DDB1 complex. Both genes seem to play a role in the regulation of reproductive physiology.


Subject(s)
Sex Differentiation , Zebrafish , Animals , Female , Humans , Male , Mice , Adaptor Proteins, Signal Transducing/metabolism , Germ Cells/metabolism , Mammals/genetics , Mammals/metabolism , Reproduction , RNA, Messenger/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
Am J Pathol ; 190(2): 426-441, 2020 02.
Article in English | MEDLINE | ID: mdl-31837950

ABSTRACT

Collagen VI (COL6) is known for its role in a spectrum of congenital muscular dystrophies, which are often accompanied by respiratory dysfunction. However, little is known regarding the function of COL6 in the lung. We confirmed the presence of COL6 throughout the basement membrane region of mouse lung tissue. Lung structure and organization were studied in a previously described Col6a1-/- mouse, which does not produce detectable COL6 in the lung. The Col6a1-/- mouse displayed histopathologic alveolar and airway abnormalities. The airspaces of Col6a1-/- lungs appeared simplified, with larger (29%; P < 0.01) and fewer (31%; P < 0.001) alveoli. These airspace abnormalities included reduced isolectin B4+ alveolar capillaries and surfactant protein C-positive alveolar epithelial type-II cells. Alterations in lung function consistent with these histopathologic changes were evident. Col6a1-/- mice also displayed multiple airway changes, including increased branching (59%; P < 0.001), increased mucosal thickness (34%; P < 0.001), and increased epithelial cell density (13%; P < 0.001). Comprehensive transcriptome analysis revealed that the loss of COL6 is associated with reductions in integrin-paxillin-phosphatidylinositol 3-kinase signaling in vivo. In vitro, COL6 promoted steady-state phosphorylated paxillin levels and reduced cell density (16% to 28%; P < 0.05) at confluence. Inhibition of phosphatidylinositol 3-kinase, or its downstream effectors, resulted in increased cell density to a level similar to that seen on matrices lacking COL6.


Subject(s)
Basement Membrane/pathology , Collagen Type VI/physiology , Epithelial Cells/pathology , Lung/pathology , Pulmonary Alveoli/pathology , Animals , Basement Membrane/metabolism , Cell Size , Epithelial Cells/metabolism , Female , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveoli/metabolism , Signal Transduction
7.
Hum Mol Genet ; 26(9): 1643-1655, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28334757

ABSTRACT

Morphogens release and activity can be negatively affected by an impaired glycosaminoglycans (GAGs) turnover and proteoglycans assembly in the extracellular matrix, leading to altered tissue morphogenesis. In this work, we show that loss of Iduronate-2-sulfatase (IDS) activity, affecting GAGs catabolism and responsible for a life-threatening valvulopathy in mucopolysaccharidosis type II (MPSII), triggers early Sonic Hedgehog (Shh) and Wnt/ß-catenin signaling defects, leading to aberrant heart development and atrioventricular valve formation in a zebrafish model. In addition, we consistently found impaired Shh signaling activity and cardiac electrophysiological abnormalities in IDS knockout mice at postnatal stages before any evident massive GAGs accumulation. These results suggest that IDS activity substantially affect cardiac morphogenesis through impaired Shh signaling and document an unexplored role of the enzyme in the fine-tuning of cell signaling pathways.


Subject(s)
Glycoproteins/metabolism , Mucopolysaccharidosis II/metabolism , Animals , Disease Models, Animal , Glycosaminoglycans/metabolism , Hedgehog Proteins/metabolism , Iduronate Sulfatase , Mice , Mice, Knockout , Myocardium/cytology , Myocardium/metabolism , Proteoglycans/metabolism , Wnt Signaling Pathway , Zebrafish/metabolism , Zebrafish Proteins/metabolism , beta Catenin
8.
J Cell Sci ; 130(19): 3203-3211, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28883093

ABSTRACT

The notochord is a midline structure common to all chordate animals; it provides mechanical and signaling cues for the developing embryo. In vertebrates, the notochord plays key functions during embryogenesis, being a source of developmental signals that pattern the surrounding tissues. It is composed of a core of vacuolated cells surrounded by an epithelial-like sheath of cells that secrete a thick peri-notochordal basement membrane made of different extracellular matrix (ECM) proteins. The correct deposition and organization of the ECM is essential for proper notochord morphogenesis and function. Work carried out in the past two decades has allowed researchers to dissect the contribution of different ECM components to this embryonic tissue. Here, we will provide an overview of these genetic and mechanistic studies. In particular, we highlight the specific functions of distinct matrix molecules in regulating notochord development and notochord-derived signals. Moreover, we also discuss the involvement of ECM synthesis and its remodeling in the pathogenesis of chordoma, a malignant bone cancer that originates from remnants of notochord remaining after embryogenesis.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Notochord/embryology , Organogenesis/physiology , Animals , Humans , Notochord/cytology
9.
Arterioscler Thromb Vasc Biol ; 38(10): 2484-2497, 2018 10.
Article in English | MEDLINE | ID: mdl-30354220

ABSTRACT

Objective- EMILIN-1 (elastin microfibrils interface located protein-1) protein inhibits pro-TGF-ß (transforming growth factor-ß) proteolysis and limits TGF-ß bioavailability in vascular extracellular matrix. Emilin1-/- null mice display increased vascular TGF-ß signaling and are hypertensive. Because EMILIN-1 is expressed in vessels from embryonic life to adulthood, we aimed at unravelling whether the hypertensive phenotype of Emilin1-/- null mice results from a developmental defect or lack of homeostatic role in the adult. Approach and Results- By using a conditional gene targeting inactivating EMILIN-1 in smooth muscle cells of adult mice, we show that increased blood pressure in mice with selective smooth muscle cell ablation of EMILIN-1 depends on enhanced myogenic tone. Mechanistically, we unveil that higher TGF-ß signaling in smooth muscle cells stimulates HB-EGF (heparin-binding epidermal growth factor) expression and subsequent transactivation of EGFR (epidermal growth factor receptor). With increasing intraluminal pressure in resistance arteries, the cross talk established by TGF-ß and EGFR signals recruits TRPC6 (TRP [transient receptor potential] classical type 6) and TRPM4 (TRP melastatin type 4) channels, lastly stimulating voltage-dependent calcium channels and potentiating myogenic tone. We found reduced EMILIN-1 and enhanced myogenic tone, dependent on increased TGF-ß-EGFR signaling, in resistance arteries from hypertensive patients. Conclusions- Taken together, our findings implicate an unexpected role of the TGF-ß-EGFR pathway in hypertension with current translational perspectives.


Subject(s)
ErbB Receptors/metabolism , Hypertension/metabolism , Membrane Glycoproteins/metabolism , Mesenteric Arteries/metabolism , Transforming Growth Factor beta1/metabolism , Vasoconstriction , Animals , Blood Pressure , Calcium Channels/metabolism , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Female , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Hypertension/genetics , Hypertension/physiopathology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Signal Transduction , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , TRPM Cation Channels/metabolism , Transforming Growth Factor beta1/pharmacology , Vasoconstriction/drug effects
10.
J Cell Sci ; 129(8): 1671-84, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26945058

ABSTRACT

Collagen VI myopathies are genetic disorders caused by mutations in collagen 6 A1, A2 and A3 genes, ranging from the severe Ullrich congenital muscular dystrophy to the milder Bethlem myopathy, which is recapitulated by collagen-VI-null (Col6a1(-/-)) mice. Abnormalities in mitochondria and autophagic pathway have been proposed as pathogenic causes of collagen VI myopathies, but the link between collagen VI defects and these metabolic circuits remains unknown. To unravel the expression profiling perturbation in muscles with collagen VI myopathies, we performed a deep RNA profiling in both Col6a1(-/-)mice and patients with collagen VI pathology. The interactome map identified common pathways suggesting a previously undetected connection between circadian genes and collagen VI pathology. Intriguingly, Bmal1(-/-)(also known as Arntl) mice, a well-characterized model displaying arrhythmic circadian rhythms, showed profound deregulation of the collagen VI pathway and of autophagy-related genes. The involvement of circadian rhythms in collagen VI myopathies is new and links autophagy and mitochondrial abnormalities. It also opens new avenues for therapies of hereditary myopathies to modulate the molecular clock or potential gene-environment interactions that might modify muscle damage pathogenesis.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Clocks/physiology , Collagen Type VI/genetics , Contracture/genetics , Mitochondria/physiology , Muscular Dystrophies/congenital , Mutation/genetics , Sclerosis/genetics , Animals , Autophagy/genetics , Gene Expression Profiling , Humans , Mice , Mice, Knockout , Microarray Analysis , Muscular Dystrophies/genetics , RNA/analysis
11.
Acta Neuropathol ; 136(3): 483-499, 2018 09.
Article in English | MEDLINE | ID: mdl-29752552

ABSTRACT

The synaptic cleft of the neuromuscular junction (NMJ) consists of a highly specialized extracellular matrix (ECM) involved in synapse maturation, in the juxtaposition of pre- to post-synaptic areas, and in ensuring proper synaptic transmission. Key components of synaptic ECM, such as collagen IV, perlecan and biglycan, are binding partners of one of the most abundant ECM protein of skeletal muscle, collagen VI (ColVI), previously never linked to NMJ. Here, we demonstrate that ColVI is itself a component of this specialized ECM and that it is required for the structural and functional integrity of NMJs. In vivo, ColVI deficiency causes fragmentation of acetylcholine receptor (AChR) clusters, with abnormal expression of NMJ-enriched proteins and re-expression of fetal AChRγ subunit, both in Col6a1 null mice and in patients affected by Ullrich congenital muscular dystrophy (UCMD), the most severe form of ColVI-related myopathies. Ex vivo muscle preparations from ColVI null mice revealed altered neuromuscular transmission, with electrophysiological defects and decreased safety factor (i.e., the excess current generated in response to a nerve impulse over that required to reach the action potential threshold). Moreover, in vitro studies in differentiated C2C12 myotubes showed the ability of ColVI to induce AChR clustering and synaptic gene expression. These findings reveal a novel role for ColVI at the NMJ and point to the involvement of NMJ defects in the etiopathology of ColVI-related myopathies.


Subject(s)
Collagen Type VI/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophies/metabolism , Neuromuscular Junction/metabolism , Receptors, Cholinergic/metabolism , Sclerosis/metabolism , Animals , Collagen Type VI/genetics , Extracellular Matrix/metabolism , Humans , Mice , Mice, Knockout , Muscular Dystrophies/genetics , Sclerosis/genetics
12.
J Biol Chem ; 291(10): 5247-58, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26742845

ABSTRACT

Collagen VI, a collagen with uncharacteristically large N- and C-terminal non-collagenous regions, forms a distinct microfibrillar network in most connective tissues. It was long considered to consist of three genetically distinct α chains (α1, α2, and α3). Intracellularly, heterotrimeric molecules associate to form dimers and tetramers, which are then secreted and assembled to microfibrils. The identification of three novel long collagen VI α chains, α4, α5, and α6, led to the question if and how these may substitute for the long α3 chain in collagen VI assembly. Here, we studied structural features of the novel long chains and analyzed the assembly of these into tetramers and microfibrils. N- and C-terminal globular regions of collagen VI were recombinantly expressed and studied by small angle x-ray scattering (SAXS). Ab initio models of the N-terminal globular regions of the α4, α5, and α6 chains showed a C-shaped structure similar to that found for the α3 chain. Single particle EM nanostructure of the N-terminal globular region of the α4 chain confirmed the C-shaped structure revealed by SAXS. Immuno-EM of collagen VI extracted from tissue revealed that like the α3 chain the novel long chains assemble to homotetramers that are incorporated into mixed microfibrils. Moreover, SAXS models of the C-terminal globular regions of the α1, α2, α4, and α6 chains were generated. Interestingly, the α1, α2, and α4 C-terminal globular regions dimerize. These self-interactions may play a role in tetramer formation.


Subject(s)
Collagen Type IV/chemistry , Protein Multimerization , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Protein Folding , Protein Structure, Tertiary
13.
J Cell Sci ; 128(19): 3525-31, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26377767

ABSTRACT

Collagen VI represents a remarkable extracellular matrix molecule, and in the past few years, studies of this molecule have revealed its involvement in a wide range of tissues and pathological conditions. In addition to its complex multi-step pathway of biosynthesis and assembly that leads to the formation of a characteristic and distinctive network of beaded microfilaments in the extracellular matrix, collagen VI exerts several key roles in different tissues. These range from unique biomechanical roles to cytoprotective functions in different cells, including myofibers, chondrocytes, neurons, fibroblasts and cardiomyocytes. Indeed, collagen VI has been shown to exert a surprisingly broad range of cytoprotective effects, which include counteracting apoptosis and oxidative damage, favoring tumor growth and progression, regulating autophagy and cell differentiation, and even contributing to the maintenance of stemness. In this Cell Science at a Glance article and the accompanying poster, we present the current knowledge of collagen VI, and in particular, discuss its relevance in stemness and in preserving the mechanical properties of tissues, as well as its links with human disorders.


Subject(s)
Collagen Type VI/metabolism , Extracellular Matrix/metabolism , Muscle, Skeletal/metabolism , Animals , Humans
14.
Exp Dermatol ; 26(5): 435-438, 2017 05.
Article in English | MEDLINE | ID: mdl-27892605

ABSTRACT

EMILIN3 is an extracellular matrix glycoprotein that displays a dynamic and restricted expression pattern in connective tissues during post-natal life. In this study, we report the characterization of EMILIN3 deposition in the skin. In addition, to unravel the functions of this protein in skin homeostasis, we generated Emilin3 null mice and provide evidence that EMILIN3 is dispensable for hair follicle growth and maintenance throughout adult life.


Subject(s)
Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Hair Follicle/growth & development , Animals , Hair Follicle/metabolism , Mice
15.
Hum Mol Genet ; 23(20): 5353-63, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24852368

ABSTRACT

Ullrich congenital muscular dystrophy (UCMD) and Bethlem myopathy (BM) are inherited muscle diseases due to mutations in the genes encoding the extracellular matrix protein collagen (Col) VI. Opening of the cyclosporin A-sensitive mitochondrial permeability transition pore (PTP) is a causative event in disease pathogenesis, and a potential target for therapy. Here, we have tested the effect of N-methyl-4-isoleucine-cyclosporin (NIM811), a non-immunosuppressive cyclophilin inhibitor, in a zebrafish model of ColVI myopathy obtained by deletion of the N-terminal region of the ColVI α1 triple helical domain, a common mutation of UCMD. Treatment with antisense morpholino sequences targeting col6a1 exon 9 at the 1-4 cell stage (within 1 h post fertilization, hpf) caused severe ultrastructural and motor abnormalities as assessed by electron and fluorescence microscopy, birefringence, spontaneous coiling events and touch-evoked responses measured at 24-48 hpf. Structural and functional abnormalities were largely prevented when NIM811--which proved significantly more effective than cyclosporin A--was administered at 21 hpf, while FK506 was ineffective. Beneficial effects of NIM811 were also detected (i) in primary muscle-derived cell cultures from UCMD and BM patients, where the typical mitochondrial alterations and depolarizing response to rotenone and oligomycin were significantly reduced; and (ii) in the Col6a1(-/-) myopathic mouse model, where apoptosis was prevented and muscle strength was increased. Since the PTP of zebrafish shares its key regulatory features with the mammalian pore, our results suggest that early treatment with NIM811 should be tested as a potential therapy for UCMD and BM.


Subject(s)
Collagen Type VI/genetics , Collagen Type VI/metabolism , Cyclosporine/administration & dosage , Muscular Dystrophies/drug therapy , Muscular Dystrophies/pathology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Cyclosporine/therapeutic use , Disease Models, Animal , Humans , Mice , Mitochondria/metabolism , Muscle Strength/drug effects , Muscular Dystrophies/congenital , Muscular Dystrophies/genetics , Zebrafish
16.
J Cell Sci ; 127(Pt 4): 828-44, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24357721

ABSTRACT

The annexins are an evolutionarily conserved family of phospholipid-binding proteins of largely unknown function. We observed that the AnxA2(-/-) lung basement membrane specifically lacks collagen VI (COL6), and postulated that ANXA2 directs bronchial epithelial cell secretion of COL6, an unusually large multimeric protein. COL6 serves to anchor cells to basement membranes and, unlike other collagens, undergoes multimerization prior to secretion. Here, we show that AnxA2(-/-) mice have reduced exercise tolerance with impaired lung tissue elasticity, which was phenocopied in Col6a1(-/-) mice. In vitro, AnxA2(-/-) fibroblasts retained COL6 within intracellular vesicles and adhered poorly to their matrix unless ANXA2 expression was restored. In vivo, AnxA2(-/-) bronchial epithelial cells underwent apoptosis and disadhesion. Immunoprecipitation and immunoelectron microscopy revealed that ANXA2 associates with COL6 and the SNARE proteins SNAP-23 and VAMP2 at secretory vesicle membranes of bronchial epithelial cells, and that absence of ANXA2 leads to retention of COL6 in a late-Golgi, VAMP2-positive compartment. These results define a new role for ANXA2 in the COL6 secretion pathway, and further show that this pathway establishes cell-matrix interactions that underlie normal pulmonary function and epithelial cell survival.


Subject(s)
Annexin A2/physiology , Apoptosis , Collagen Type VI/metabolism , Epithelial Cells/metabolism , Animals , Basement Membrane , Bronchi/metabolism , Bronchi/pathology , Cell Shape , Cell Survival , Cells, Cultured , Collagen Type VI/genetics , Elasticity , Epithelial Cells/physiology , Exercise Tolerance , Golgi Apparatus/metabolism , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Physical Exertion , Protein Transport , Pulmonary Ventilation , Respiratory Mucosa/pathology , Vesicle-Associated Membrane Protein 2/metabolism
17.
Development ; 140(22): 4594-601, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24131633

ABSTRACT

The notochord is a transient and essential structure that provides both mechanical and signaling cues to the developing vertebrate embryo. In teleosts, the notochord is composed of a core of large vacuolated cells and an outer layer of cells that secrete the notochord sheath. In this work, we have identified the extracellular matrix glycoprotein Emilin3 as a novel essential component of the zebrafish notochord sheath. The development of the notochord sheath is impaired in Emilin3 knockdown embryos. The patterning activity of the notochord is also affected by Emilin3, as revealed by the increase of Hedgehog (Hh) signaling in Emilin3-depleted embryos and the decreased Hh signaling in embryos overexpressing Emilin3 in the notochord. In vitro and in vivo experiments indicate that Emilin3 modulates the availability of Hh ligands by interacting with the permissive factor Scube2 in the notochord sheath. Overall, this study reveals a new role for an EMILIN protein and reinforces the concept that structure and function of the notochord are strictly linked.


Subject(s)
Antigens, Surface/metabolism , Extracellular Matrix Proteins/metabolism , Hedgehog Proteins/metabolism , Membrane Glycoproteins/metabolism , Notochord/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Body Patterning/drug effects , Body Patterning/genetics , Down-Regulation/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Models, Biological , Morpholinos/pharmacology , Notochord/cytology , Notochord/drug effects , Notochord/embryology , Protein Binding/drug effects , Protein Binding/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects
18.
Acta Neuropathol ; 132(1): 127-44, 2016 07.
Article in English | MEDLINE | ID: mdl-26971100

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is a neuromuscular disease caused by the expansion of a polyglutamine tract in the androgen receptor (AR). The mechanism by which expansion of polyglutamine in AR causes muscle atrophy is unknown. Here, we investigated pathological pathways underlying muscle atrophy in SBMA knock-in mice and patients. We show that glycolytic muscles were more severely affected than oxidative muscles in SBMA knock-in mice. Muscle atrophy was associated with early-onset, progressive glycolytic-to-oxidative fiber-type switch. Whole genome microarray and untargeted lipidomic analyses revealed enhanced lipid metabolism and impaired glycolysis selectively in muscle. These metabolic changes occurred before denervation and were associated with a concurrent enhancement of mechanistic target of rapamycin (mTOR) signaling, which induced peroxisome proliferator-activated receptor γ coactivator 1 alpha (PGC1α) expression. At later stages of disease, we detected mitochondrial membrane depolarization, enhanced transcription factor EB (TFEB) expression and autophagy, and mTOR-induced protein synthesis. Several of these abnormalities were detected in the muscle of SBMA patients. Feeding knock-in mice a high-fat diet (HFD) restored mTOR activation, decreased the expression of PGC1α, TFEB, and genes involved in oxidative metabolism, reduced mitochondrial abnormalities, ameliorated muscle pathology, and extended survival. These findings show early-onset and intrinsic metabolic alterations in SBMA muscle and link lipid/glucose metabolism to pathogenesis. Moreover, our results highlight an HFD regime as a promising approach to support SBMA patients.


Subject(s)
Diet, High-Fat/adverse effects , Glycolysis , Muscle, Skeletal/metabolism , Muscular Disorders, Atrophic/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Atrophy/metabolism , Atrophy/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Disease Models, Animal , Disease Progression , Female , Glycolysis/physiology , Humans , Lipid Metabolism/physiology , Male , Membrane Potential, Mitochondrial/physiology , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Disorders, Atrophic/pathology , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Random Allocation , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction
19.
Cell Mol Life Sci ; 72(16): 2989-3008, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25833128

ABSTRACT

The notochord is an embryonic midline structure common to all members of the phylum Chordata, providing both mechanical and signaling cues to the developing embryo. In vertebrates, the notochord arises from the dorsal organizer and it is critical for proper vertebrate development. This evolutionary conserved structure located at the developing midline defines the primitive axis of embryos and represents the structural element essential for locomotion. Besides its primary structural function, the notochord is also a source of developmental signals that patterns surrounding tissues. Among the signals secreted by the notochord, Hedgehog proteins play key roles during embryogenesis. The Hedgehog signaling pathway is a central regulator of embryonic development, controlling the patterning and proliferation of a wide variety of organs. In this review, we summarize the current knowledge on notochord structure and functions, with a particular emphasis on the key developmental events that take place in vertebrates. Moreover, we discuss some genetic studies highlighting the phenotypic consequences of impaired notochord development, which enabled to understand the molecular basis of different human congenital defects and diseases.


Subject(s)
Basement Membrane/metabolism , Biological Evolution , Cell Communication/physiology , Models, Biological , Notochord/anatomy & histology , Notochord/embryology , Somites/embryology , Animals , Extracellular Matrix Proteins/metabolism , Hedgehog Proteins/metabolism , Humans , Intervertebral Disc/embryology , Species Specificity , Spine/embryology
SELECTION OF CITATIONS
SEARCH DETAIL