Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cell ; 140(4): 554-66, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20178747

ABSTRACT

Skeletal myogenesis, like hematopoiesis, occurs in successive developmental stages that involve different cell populations and expression of different genes. We show here that the transcription factor nuclear factor one X (Nfix), whose expression is activated by Pax7 in fetal muscle, in turn activates the transcription of fetal specific genes such as MCK and beta-enolase while repressing embryonic genes such as slow myosin. In the case of the MCK promoter, Nfix forms a complex with PKC theta that binds, phosphorylates, and activates MEF2A. Premature expression of Nfix activates fetal and suppresses embryonic genes in embryonic muscle, whereas muscle-specific ablation of Nfix prevents fetal and maintains embryonic gene expression in the fetus. Therefore, Nfix acts as a transcriptional switch from embryonic to fetal myogenesis.


Subject(s)
Muscle Development , Muscle, Skeletal/embryology , NFI Transcription Factors/metabolism , Transcription, Genetic , Animals , Fetus/metabolism , Gene Expression Regulation, Developmental , Humans , Isoenzymes/metabolism , MEF2 Transcription Factors , Mice , Myogenic Regulatory Factors/metabolism , NFATC Transcription Factors/metabolism , PAX7 Transcription Factor/metabolism , Phosphopyruvate Hydratase , Protein Kinase C/metabolism , Protein Kinase C-theta
2.
J Pathol ; 257(3): 352-366, 2022 07.
Article in English | MEDLINE | ID: mdl-35297529

ABSTRACT

Muscular dystrophies are genetic diseases characterized by chronic inflammation and fibrosis. Macrophages are immune cells that sustain muscle regeneration upon acute injury but seem deleterious in the context of chronic muscle injury such as in muscular dystrophies. Here, we observed that the number of macrophages expressing the transcription factor Nfix increases in two distinct mouse models of muscular dystrophies. We showed that the deletion of Nfix in macrophages in dystrophic mice delays the establishment of fibrosis and muscle wasting, and increases grasp force. Macrophages lacking Nfix expressed more TNFα and less TGFß1, thus promoting apoptosis of fibro-adipogenic progenitors. Moreover, pharmacological treatment of dystrophic mice with a ROCK inhibitor accelerated fibrosis through the increase of Nfix expression by macrophages. Thus, we have identified Nfix as a macrophage profibrotic factor in muscular dystrophies, whose inhibition could be a therapeutic route to reduce severity of the dystrophic disease. © 2022 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)
Macrophages , Muscular Dystrophies , NFI Transcription Factors , Animals , Fibrosis , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , NFI Transcription Factors/deficiency , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism
3.
Exp Cell Res ; 410(2): 112968, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34883113

ABSTRACT

Muscular dystrophies (MDs) are heterogeneous diseases, characterized by primary wasting of skeletal muscle, which in severe cases, such as Duchenne Muscular Dystrophy (DMD), leads to wheelchair dependency, respiratory failure, and premature death. Research is ongoing to develop efficacious therapies, particularly for DMD. Most of the efforts, currently focusing on correcting or restoring the primary defect of MDs, are based on gene-addition, exon-skipping, stop codon read-through, and genome-editing. Although promising, most of them revealed several practical limitations. Shared knowledge in the field is that, in order to be really successful, any therapeutic approach has to rely on spared functional muscle tissue, restricting the number of patients eligible for clinical trials to the youngest and less compromised individuals. In line with this, many therapeutic strategies aim to preserve muscle tissue and function. This Review outlines the most interesting and recent studies addressing the secondary outcomes of DMD and how to better deliver the therapeutic agents. In the future, the effective treatment of DMD will likely require combinations of therapies addressing both the primary genetic defect and its consequences.


Subject(s)
Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/therapy , Calcium/metabolism , Energy Metabolism , Humans , Inflammation/pathology , Sodium/metabolism
4.
Development ; 145(21)2018 10 29.
Article in English | MEDLINE | ID: mdl-30266829

ABSTRACT

The transcription factor Nfix belongs to the nuclear factor one family and has an essential role in prenatal skeletal muscle development, where it is a master regulator of the transition from embryonic to foetal myogenesis. Recently, Nfix was shown to be involved in adult muscle regeneration and in muscular dystrophies. Here, we have investigated the signalling that regulates Nfix expression, and show that JunB, a member of the AP-1 family, is an activator of Nfix, which then leads to foetal myogenesis. Moreover, we demonstrate that their expression is regulated through the RhoA/ROCK axis, which maintains embryonic myogenesis. Specifically, RhoA and ROCK repress ERK kinase activity, which promotes JunB and Nfix expression. Notably, the role of ERK in the activation of Nfix is conserved postnatally in satellite cells, which represent the canonical myogenic stem cells of adult muscle. As lack of Nfix in muscular dystrophies rescues the dystrophic phenotype, the identification of this pathway provides an opportunity to pharmacologically target Nfix in muscular dystrophies.


Subject(s)
MAP Kinase Signaling System , Muscle Development , Myoblasts/metabolism , NFI Transcription Factors/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Embryo, Mammalian/metabolism , Female , Fetus/metabolism , Gene Expression Regulation, Developmental , Gene Silencing , Male , Mice , NFI Transcription Factors/genetics , Stem Cells/metabolism , Transcription Factors/metabolism , rho-Associated Kinases/metabolism
5.
Am J Pathol ; 189(2): 354-369, 2019 02.
Article in English | MEDLINE | ID: mdl-30448410

ABSTRACT

In muscular dystrophies, muscle membrane fragility results in a tissue-specific increase of danger-associated molecular pattern molecules (DAMPs) and infiltration of inflammatory cells. The DAMP extracellular ATP (eATP) released by dying myofibers steadily activates muscle and immune purinergic receptors exerting dual negative effects: a direct damage linked to altered intracellular calcium homeostasis in muscle cells and an indirect toxicity through the triggering of the immune response and inhibition of regulatory T cells. Accordingly, pharmacologic and genetic inhibition of eATP signaling improves the phenotype in models of chronic inflammatory diseases. In α-sarcoglycanopathy, eATP effects may be further amplified because α-sarcoglycan extracellular domain binds eATP and displays an ecto-ATPase activity, thus controlling eATP concentration at the cell surface and attenuating the magnitude and/or the duration of eATP-induced signals. Herein, we show that in vivo blockade of the eATP/P2X purinergic pathway by a broad-spectrum P2X receptor-antagonist delayed the progression of the dystrophic phenotype in α-sarcoglycan-null mice. eATP blockade dampened the muscular inflammatory response and enhanced the recruitment of forkhead box protein P3-positive immunosuppressive regulatory CD4+ T cells. The improvement of the inflammatory features was associated with increased strength, reduced necrosis, and limited expression of profibrotic factors, suggesting that pharmacologic purinergic antagonism, altering the innate and adaptive immune component in muscle infiltrates, might provide a therapeutic approach to slow disease progression in α-sarcoglycanopathy.


Subject(s)
Adenosine Triphosphate/immunology , Muscular Dystrophy, Animal , Myofibrils , Sarcoglycans/deficiency , T-Lymphocytes, Regulatory , Adenosine Triphosphate/genetics , Animals , Calcium/immunology , Chronic Disease , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/immunology , Muscular Dystrophy, Animal/pathology , Myofibrils/immunology , Myofibrils/pathology , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/immunology , Sarcoglycans/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
6.
Mol Ther ; 26(4): 1093-1108, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29503200

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal muscle-wasting disease currently without cure. We investigated the use of the PiggyBac transposon for full-length dystrophin expression in murine mesoangioblast (MABs) progenitor cells. DMD murine MABs were transfected with transposable expression vectors for full-length dystrophin and transplanted intramuscularly or intra-arterially into mdx/SCID mice. Intra-arterial delivery indicated that the MABs could migrate to regenerating muscles to mediate dystrophin expression. Intramuscular transplantation yielded dystrophin expression in 11%-44% of myofibers in murine muscles, which remained stable for the assessed period of 5 months. The satellite cells isolated from transplanted muscles comprised a fraction of MAB-derived cells, indicating that the transfected MABs may colonize the satellite stem cell niche. Transposon integration site mapping by whole-genome sequencing indicated that 70% of the integrations were intergenic, while none was observed in an exon. Muscle resistance assessment by atomic force microscopy indicated that 80% of fibers showed elasticity properties restored to those of wild-type muscles. As measured in vivo, transplanted muscles became more resistant to fatigue. This study thus provides a proof-of-principle that PiggyBac transposon vectors may mediate full-length dystrophin expression as well as functional amelioration of the dystrophic muscles within a potential autologous cell-based therapeutic approach of DMD.


Subject(s)
Cell- and Tissue-Based Therapy , DNA Transposable Elements , Gene Transfer Techniques , Genetic Vectors/genetics , Muscular Dystrophy, Duchenne/genetics , Myoblasts/metabolism , Myoblasts/transplantation , Animals , Cell Line , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Dystrophin/genetics , Fluorescent Antibody Technique , Gene Dosage , Gene Expression , Gene Order , Genes, Reporter , Male , Mice , Mice, Inbred mdx , Mice, SCID , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/therapy , Phenotype , Transgenes , Transplantation, Autologous
7.
Genes Dev ; 23(8): 902-5, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19390084

ABSTRACT

Skeletal muscle is a heterogeneous tissue composed of individual muscle fibers, diversified in size, shape, and contractile protein content, to fulfill the different functional needs of the vertebrate body. This heterogeneity derives from and depends at least in part on distinct classes of myogenic progenitors; i.e., embryonic and fetal myoblasts and satellite cells whose origin and lineage relationship have been elusive so far. In this issue of Genes & Development, Hutcheson and colleagues (pp. 997-1013) provide a first answer to this question.


Subject(s)
Muscle Development/physiology , Myoblasts/cytology , PAX7 Transcription Factor/metabolism , Paired Box Transcription Factors/metabolism , Animals , Embryo, Mammalian , Fetus , Mice , PAX3 Transcription Factor , beta Catenin/metabolism
8.
Development ; 140(7): 1528-36, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23482488

ABSTRACT

During mouse skeletal muscle development, the Nfix gene has a pivotal role in regulating fetal-specific transcription. Zebrafish and mice share related programs for muscle development, although zebrafish develops at a much faster rate. In fact, although mouse fetal muscle fibers form after 15 days of development, in fish secondary muscle fibers form by 48 hours post-fertilization in a process that until now has been poorly characterized mechanically. In this work, we studied the zebrafish ortholog Nfix (nfixa) and its role in the proper switch to the secondary myogenic wave. This allowed us to highlight evolutionarily conserved and divergent functions of Nfix. In fact, the knock down of nfixa in zebrafish blocks secondary myogenesis, as in mouse, but also alters primary slow muscle fiber formation. Moreover, whereas Nfix mutant mice are motile, nfixa knockdown zebrafish display impaired motility that probably depends upon disruption of the sarcoplasmic reticulum. We conclude that, during vertebrate evolution, the transcription factor Nfix lost some specific functions, probably as a consequence of the different environment in which teleosts and mammals develop.


Subject(s)
Evolution, Molecular , Muscle, Skeletal/embryology , NFI Transcription Factors/physiology , Vertebrates/genetics , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Conserved Sequence/physiology , Embryo, Mammalian , Embryo, Nonmammalian , Genetic Speciation , Mice , Muscle Development/genetics , Muscle Development/physiology , Muscle, Skeletal/metabolism , NFI Transcription Factors/genetics , Phylogeny , Vertebrates/embryology , Vertebrates/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
9.
Circ Res ; 113(4): 389-98, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23753573

ABSTRACT

RATIONALE: A cell-based biological pacemaker is based on the differentiation of stem cells and the selection of a population displaying the molecular and functional properties of native sinoatrial node (SAN) cardiomyocytes. So far, such selection has been hampered by the lack of proper markers. CD166 is specifically but transiently expressed in the mouse heart tube and sinus venosus, the prospective SAN. OBJECTIVE: We have explored the possibility of using CD166 expression for isolating SAN progenitors from differentiating embryonic stem cells. METHODS AND RESULTS: We found that in embryonic day 10.5 mouse hearts, CD166 and HCN4, markers of the pacemaker tissue, are coexpressed. Sorting embryonic stem cells for CD166 expression at differentiation day 8 selects a population of pacemaker precursors. CD166+ cells express high levels of genes involved in SAN development (Tbx18, Tbx3, Isl-1, Shox2) and function (Cx30.2, HCN4, HCN1, CaV1.3) and low levels of ventricular genes (Cx43, Kv4.2, HCN2, Nkx2.5). In culture, CD166+ cells form an autorhythmic syncytium composed of cells morphologically similar to and with the electrophysiological properties of murine SAN myocytes. Isoproterenol increases (+57%) and acetylcholine decreases (-23%) the beating rate of CD166-selected cells, which express the ß-adrenergic and muscarinic receptors. In cocultures, CD166-selected cells are able to pace neonatal ventricular myocytes at a rate faster than their own. Furthermore, CD166+ cells have lost pluripotency genes and do not form teratomas in vivo. CONCLUSIONS: We demonstrated for the first time the isolation of a nonteratogenic population of cardiac precursors able to mature and form a fully functional SAN-like tissue.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Sinoatrial Node/cytology , Stem Cells/cytology , Acetylcholine/pharmacology , Animals , Biomarkers/metabolism , Cardiotonic Agents/pharmacology , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Coculture Techniques , Embryonic Stem Cells/drug effects , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Isoproterenol/pharmacology , Mice , Models, Animal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Sinoatrial Node/drug effects , Sinoatrial Node/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
10.
Cell Mol Life Sci ; 71(16): 3081-99, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24664432

ABSTRACT

Skeletal myogenesis has been and is currently under extensive study in both mammals and teleosts, with the latter providing a good model for skeletal myogenesis because of their flexible and conserved genome. Parallel investigations of muscle studies using both these models have strongly accelerated the advances in the field. However, when transferring the knowledge from one model to the other, it is important to take into account both their similarities and differences. The main difficulties in comparing mammals and teleosts arise from their different temporal development. Conserved aspects can be seen for muscle developmental origin and segmentation, and for the presence of multiple myogenic waves. Among the divergences, many fish have an indeterminate growth capacity throughout their entire life span, which is absent in mammals, thus implying different post-natal growth mechanisms. This review covers the current state of the art on myogenesis, with a focus on the most conserved and divergent aspects between mammals and teleosts.


Subject(s)
Muscle Development , Muscle, Skeletal/embryology , Muscle, Skeletal/physiology , Animals , Humans , Mammals/embryology , Muscle, Skeletal/cytology , Regeneration , Zebrafish/embryology
11.
Nat Cell Biol ; 9(3): 255-67, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17293855

ABSTRACT

Cells derived from blood vessels of human skeletal muscle can regenerate skeletal muscle, similarly to embryonic mesoangioblasts. However, adult cells do not express endothelial markers, but instead express markers of pericytes, such as NG2 proteoglycan and alkaline phosphatase (ALP), and can be prospectively isolated from freshly dissociated ALP(+) cells. Unlike canonical myogenic precursors (satellite cells), pericyte-derived cells express myogenic markers only in differentiated myotubes, which they form spontaneously with high efficiency. When transplanted into severe combined immune deficient-X-linked, mouse muscular dystrophy (scid-mdx) mice, pericyte-derived cells colonize host muscle and generate numerous fibres expressing human dystrophin. Similar cells isolated from Duchenne patients, and engineered to express human mini-dystrophin, also give rise to many dystrophin-positive fibres in vivo. These data show that myogenic precursors, distinct from satellite cells, are associated with microvascular walls in the human skeletal muscle, may represent a correlate of embryonic 'mesoangioblasts' present after birth and may be a promising candidate for future cell-therapy protocols in patients.


Subject(s)
Adult Stem Cells/cytology , Muscle, Skeletal/cytology , Pericytes/cytology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Adolescent , Adult , Adult Stem Cells/metabolism , Adult Stem Cells/transplantation , Aged , Animals , Antigens, CD/analysis , Cell Culture Techniques/methods , Child , Child, Preschool , Female , Humans , Male , Mice , Mice, Inbred mdx , Mice, Nude , Mice, SCID , Middle Aged , Muscle Proteins/analysis , Muscle Proteins/genetics , Muscle, Skeletal/chemistry , Muscle, Skeletal/physiology , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/surgery , Pericytes/chemistry , Pericytes/transplantation , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation/methods , Treatment Outcome
12.
iScience ; 27(1): 108696, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38205246

ABSTRACT

Muscular dystrophies (MDs) are incurable genetic myopathies characterized by progressive degeneration of skeletal muscles. Dystrophic mice lacking the transcription factor Nfix display morphological and functional improvements of the disease. Recently, we demonstrated that MAPK signaling pathway positively regulates Nfix in muscle development and that Cyanidin, a natural antioxidant molecule, strongly ameliorates the pathology. To explore a synergistic approach aimed at treating MDs, we administered Trametinib, a clinically approved MEK inhibitor, alone or combined with Cyanidin to adult Sgca null mice. We observed that chronic treatment with Trametinib and Cyanidin reduced Nfix in myogenic cells but, unexpectedly, caused ectopic calcifications exclusively in dystrophic muscles. The combined treatment with Cyanidin resulted in histological improvements by preventing Trametinib-induced calcifications in Diaphragm and Soleus. Collectively, this first pilot study revealed that Nfix is modulated by the MAPK pathway in MDs, and that Cyanidin partly rescued the unexpected ectopic calcifications caused by MEK inhibition.

13.
EMBO Rep ; 12(2): 164-71, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21212806

ABSTRACT

Despite having distinct expression patterns and phenotypes in mutant mice, the myogenic regulatory factors Myf5 and MyoD have been considered to be functionally equivalent. Here, we report that these factors have a different response to DNA damage, due to the presence in MyoD and absence in Myf5 of a consensus site for Abl-mediated tyrosine phosphorylation that inhibits MyoD activity in response to DNA damage. Genotoxins failed to repress skeletal myogenesis in MyoD-null embryos; reintroduction of wild-type MyoD, but not mutant Abl phosphorylation-resistant MyoD, restored the DNA-damage-dependent inhibition of muscle differentiation. Conversely, introduction of the Abl-responsive phosphorylation motif converts Myf5 into a DNA-damage-sensitive transcription factor. Gene-dosage-dependent reduction of Abl kinase activity in MyoD-expressing cells attenuated the DNA-damage-dependent inhibition of myogenesis. The presence of a DNA-damage-responsive phosphorylation motif in vertebrate, but not in invertebrate MyoD suggests an evolved response to environmental stress, originated from basic helix-loop-helix gene duplication in vertebrate myogenesis.


Subject(s)
Muscle Development/drug effects , Mutagens/toxicity , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Biological Evolution , Cell Cycle Proteins/metabolism , Cell Differentiation , Cells, Cultured , Coculture Techniques , Cross-Linking Reagents/toxicity , DNA Damage , DNA-Binding Proteins/metabolism , Etoposide/toxicity , Female , Gene Knockdown Techniques , Methyl Methanesulfonate/toxicity , Mice/embryology , Mitomycin/toxicity , MyoD Protein/genetics , Myogenic Regulatory Factor 5/genetics , Phosphorylation , Pregnancy , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-abl/physiology , RNA Interference , Somites/drug effects , Somites/metabolism , Tumor Suppressor Proteins/metabolism
14.
Acta Physiol (Oxf) ; 239(2): e13981, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37186371

ABSTRACT

AIMS: Nfix is a transcription factor belonging to the Nuclear Factor I (NFI) family comprising four members (Nfia, b, c, x). Nfix plays important roles in the development and function of several organs. In muscle development, Nfix controls the switch from embryonic to fetal myogenesis by promoting fast twitching fibres. In the adult muscle, following injury, lack of Nfix impairs regeneration, inducing higher content of slow-twitching fibres. Nfix is expressed also in the heart, but its function has been never investigated before. We studied Nfix role in this organ. METHODS: Using Nfix-null and wild type (WT) mice we analyzed: (1) the expression pattern of Nfix during development by qPCR and (2) the functional alterations caused by its absence, by in vivo telemetry and in vitro patch clamp analysis. RESULTS AND CONCLUSIONS: Nfix expression start in the heart from E12.5. Adult hearts of Nfix-null mice show a hearts morphology and sarcomeric proteins expression similar to WT. However, Nfix-null animals show tachycardia that derives form an intrinsic higher beating rate of the sinus node (SAN). Molecular and functional analysis revealed that sinoatrial cells of Nfix-null mice express a significantly larger L-type calcium current (Cacna1d + Cacna1c). Interestingly, downregulation of Nfix by sh-RNA in primary cultures of neonatal rat ventricular cardiomyocytes induced a similar increase in their spontaneous beating rate and in ICaL current. In conclusion, our data provide the first demonstration of a role of Nfix that, increasing the L-type calcium current, modulates heart rate.

15.
Cell Rep ; 42(1): 111992, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36662619

ABSTRACT

Insights into the evolution of non-model organisms are limited by the lack of reference genomes of high accuracy, completeness, and contiguity. Here, we present a chromosome-level, karyotype-validated reference genome and pangenome for the barn swallow (Hirundo rustica). We complement these resources with a reference-free multialignment of the reference genome with other bird genomes and with the most comprehensive catalog of genetic markers for the barn swallow. We identify potentially conserved and accelerated genes using the multialignment and estimate genome-wide linkage disequilibrium using the catalog. We use the pangenome to infer core and accessory genes and to detect variants using it as a reference. Overall, these resources will foster population genomics studies in the barn swallow, enable detection of candidate genes in comparative genomics studies, and help reduce bias toward a single reference genome.


Subject(s)
Swallows , Animals , Swallows/genetics , Metagenomics , Genome/genetics , Genomics , Chromosomes
16.
J Neuromuscul Dis ; 9(1): 1-23, 2022.
Article in English | MEDLINE | ID: mdl-34542080

ABSTRACT

While skeletal muscle remodeling happens throughout life, diseases that result in its dysfunction are accountable for many deaths. Indeed, skeletal muscle is exceptionally capable to respond to stimuli modifying its homeostasis, such as in atrophy, hypertrophy, regeneration and repair. In particular conditions such as genetic diseases (muscular dystrophies), skeletal muscle's capacity to remodel is strongly affected and undergoes continuous cycles of chronic damage. This induces scarring, fatty infiltration, as well as loss of contractibility and of the ability to generate force. In this context, inflammation, primarily mediated by macrophages, plays a central pathogenic role. Macrophages contribute as the primary regulators of inflammation during skeletal muscle regeneration, affecting tissue-resident cells such as myogenic cells and endothelial cells, but also fibro-adipogenic progenitors, which are the main source of the fibro fatty scar. During skeletal muscle regeneration their function is tightly orchestrated, while in dystrophies their fate is strongly disturbed, resulting in chronic inflammation. In this review, we will discuss the latest findings on the role of macrophages in skeletal muscle diseases, and how they are regulated.


Subject(s)
Inflammation/immunology , Macrophages/physiology , Muscular Dystrophies/immunology , Humans
17.
Front Genet ; 13: 1056114, 2022.
Article in English | MEDLINE | ID: mdl-36685855

ABSTRACT

In 2002 we published an article describing a population of vessel-associated progenitors that we termed mesoangioblasts (MABs). During the past decade evidence had accumulated that during muscle development and regeneration things may be more complex than a simple sequence of binary choices (e.g., dorsal vs. ventral somite). LacZ expressing fibroblasts could fuse with unlabelled myoblasts but not among themselves or with other cell types. Bone marrow derived, circulating progenitors were able to participate in muscle regeneration, though in very small percentage. Searching for the embryonic origin of these progenitors, we identified them as originating at least in part from the embryonic aorta and, at later stages, from the microvasculature of skeletal muscle. While continuing to investigate origin and fate of MABs, the fact that they could be expanded in vitro (also from human muscle) and cross the vessel wall, suggested a protocol for the cell therapy of muscular dystrophies. We tested this protocol in mice and dogs before proceeding to the first clinical trial on Duchenne Muscular Dystrophy patients that showed safety but minimal efficacy. In the last years, we have worked to overcome the problem of low engraftment and tried to understand their role as auxiliary myogenic progenitors during development and regeneration.

18.
J Biol Chem ; 285(45): 34518-27, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20801874

ABSTRACT

Reversible proline-directed phosphorylation at Ser/Thr-Pro motifs has an essential role in myogenesis, a multistep process strictly regulated by several signaling pathways that impinge on two families of myogenic effectors, the basic helix-loop-helix myogenic transcription factors and the MEF2 (myocyte enhancer factor 2) proteins. The question of how these signals are deciphered by the myogenic effectors remains largely unaddressed. In this study, we show that the peptidyl-prolyl isomerase Pin1, which catalyzes the isomerization of phosphorylated Ser/Thr-Pro peptide bonds to induce conformational changes of its target proteins, acts as an inhibitor of muscle differentiation because its knockdown in myoblasts promotes myotube formation. With the aim of clarifying the mechanism of Pin1 function in skeletal myogenesis, we investigated whether MEF2C, a critical regulator of the myogenic program that is the end point of several signaling pathways, might serve as a/the target for the inhibitory effects of Pin1 on muscle differentiation. We show that Pin1 interacts selectively with phosphorylated MEF2C in skeletal muscle cells, both in vitro and in vivo. The interaction with Pin1 requires two novel critical phospho-Ser/Thr-Pro motifs in MEF2C, Ser(98) and Ser(110), which are phosphorylated in vivo. Overexpression of Pin1 decreases MEF2C stability and activity and its ability to cooperate with MyoD to activate myogenic conversion. Collectively, these findings reveal a novel role for Pin1 as a regulator of muscle terminal differentiation and suggest that Pin1-mediated repression of MEF2C function could contribute to this function.


Subject(s)
Cell Proliferation , Muscle Development/physiology , Muscle Fibers, Skeletal/metabolism , Myogenic Regulatory Factors/metabolism , Peptidylprolyl Isomerase/metabolism , Signal Transduction/physiology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , MEF2 Transcription Factors , Mice , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factors/genetics , NIMA-Interacting Peptidylprolyl Isomerase , Peptides/genetics , Peptides/metabolism , Peptidylprolyl Isomerase/genetics , Phosphorylation/physiology , Protein Stability
19.
Talanta ; 235: 122742, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34517610

ABSTRACT

Due to the physiological properties of l-carnosine (l-1), supplementation of this dipeptide has both a nutritional ergogenic application and a therapeutic potential for the treatment of numerous diseases in which ischemic or oxidative stress are involved. Quantitation of carnosine and its analogs in biological matrices results to be crucial for these applications and HPLC-MS procedures with isotope-labeled internal standards are the state-of-the-art approach for this analytical need. The use of these standards allows to account for variations during the sample preparation process, between-sample matrix effects, and variations in instrument performance over analysis time. Although literature reports a number of studies involving carnosine, isotope-labeled derivatives of the dipeptide are not commercially available. In this work we present a fast, flexible, and convenient strategy for the synthesis of the 13C-labeled carnosine analogs and their application as internal standards for the quantitation of carnosine and anserine in a biological matrix.


Subject(s)
Carnosine , Anserine , Chromatography, High Pressure Liquid , Dipeptides , Mass Spectrometry
20.
Nat Commun ; 12(1): 6013, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34650038

ABSTRACT

The transcription factor NF-Y promotes cell proliferation and its activity often declines during differentiation through the regulation of NF-YA, the DNA binding subunit of the complex. In stem cell compartments, the shorter NF-YA splice variant is abundantly expressed and sustains their expansion. Here, we report that satellite cells, the stem cell population of adult skeletal muscle necessary for its growth and regeneration, express uniquely the longer NF-YA isoform, majorly associated with cell differentiation. Through the generation of a conditional knock out mouse model that selectively deletes the NF-YA gene in satellite cells, we demonstrate that NF-YA expression is fundamental to preserve the pool of muscle stem cells and ensures robust regenerative response to muscle injury. In vivo and ex vivo, satellite cells that survive to NF-YA loss exit the quiescence and are rapidly committed to early differentiation, despite delayed in the progression towards later states. In vitro results demonstrate that NF-YA-depleted muscle stem cells accumulate DNA damage and cannot properly differentiate. These data highlight a new scenario in stem cell biology for NF-Y activity, which is required for efficient myogenic differentiation.


Subject(s)
CCAAT-Binding Factor/metabolism , Muscle, Skeletal/metabolism , Regeneration/physiology , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , CCAAT-Binding Factor/genetics , Cell Differentiation/genetics , Cell Proliferation , Gene Expression Regulation , Male , Mice , Mice, Knockout , Muscle Development/genetics , Muscle Development/physiology , Protein Isoforms/genetics , Regeneration/genetics
SELECTION OF CITATIONS
SEARCH DETAIL