Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 15(2): 169, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395995

ABSTRACT

Phosphodiesterase 2A (Pde2A) is a dual-specific PDE that breaks down both cAMP and cGMP cyclic nucleotides. We recently highlighted a direct relationship between Pde2A impairment, a consequent increase of cAMP, and the appearance of mouse congenital heart defects (CHDs). Here we aimed to characterize the pathways involved in the development of CHDs and in their prevention by pharmacological approaches targeting cAMP and cGMP signaling. Transcriptome analysis revealed a modulation of more than 500 genes affecting biological processes involved in the immune system, cardiomyocyte development and contractility, angiogenesis, transcription, and oxidative stress in hearts from Pde2A-/- embryos. Metoprolol and H89 pharmacological administration prevented heart dilatation and hypertabeculation in Pde2A-/- embryos. Metoprolol was also able to partially impede heart septum defect and oxidative stress at tissue and molecular levels. Amelioration of cardiac defects was also observed by using the antioxidant NAC, indicating oxidative stress as one of the molecular mechanisms underpinning the CHDs. In addition, Sildenafil treatment recovered cardiac defects suggesting the requirement of cAMP/cGMP nucleotides balance for the correct heart development.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2 , Heart Defects, Congenital , Mice , Animals , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Metoprolol , Signal Transduction , Cyclic GMP/metabolism , Heart Defects, Congenital/genetics , Heart Defects, Congenital/prevention & control , Oxidative Stress
2.
Cell Mol Life Sci ; 80(8): 202, 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37442828

ABSTRACT

The epidermal growth factor receptor (EGFR) is one of the main tumor drivers and is an important therapeutic target for many cancers. Calcium is important in EGFR signaling pathways. Sorcin is one of the most important calcium sensor proteins, overexpressed in many tumors, that promotes cell proliferation, migration, invasion, epithelial-to-mesenchymal transition, malignant progression and resistance to chemotherapeutic drugs. The present work elucidates a functional mechanism that links calcium homeostasis to EGFR signaling in cancer. Sorcin and EGFR expression are significantly correlated and associated with reduced overall survival in cancer patients. Mechanistically, Sorcin directly binds EGFR protein in a calcium-dependent fashion and regulates calcium (dys)homeostasis linked to EGF-dependent EGFR signaling. Moreover, Sorcin controls EGFR proteostasis and signaling and increases its phosphorylation, leading to increased EGF-dependent migration and invasion. Of note, silencing of Sorcin cooperates with EGFR inhibitors in the regulation of migration, highlighting calcium signaling pathway as an exploitable target to enhance the effectiveness of EGFR-targeting therapies.


Subject(s)
Epidermal Growth Factor , Neoplasms , Humans , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/metabolism , Calcium , Signal Transduction , ErbB Receptors/genetics , ErbB Receptors/metabolism , Cell Line, Tumor , Cell Movement
3.
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.

4.
Int J Mol Sci ; 24(1)2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36614143

ABSTRACT

Phosphodiesterase 5A (PDE5A) is involved in cGMP hydrolysis, regulating many physiological processes. Increased activity of PDE5A has been found in several pathological conditions, and the pharmacological inhibition of PDE5 has been demonstrated to have several therapeutic applications. We have identified the presence of three different Pde5a isoforms in cardiomyocytes, and we have found that the expression of specific Pde5a isoforms may have a causal role in the onset of pathological responses in these cells. In our previous study, we demonstrated that PDE5A inhibition could ameliorate muscular dystrophy by acting at different levels, as assessed by the altered genomic response of muscular cells following treatment with the PDE5A inhibitor tadalafil. Thus, considering the importance of PDE5A in various pathophysiological conditions, we further investigated the regulation of this enzyme. Here, we analysed the expression of Pde5a isoforms in the pathophysiology of skeletal muscle. We found that skeletal muscle tissues and myogenic cells express Pde5a1 and Pde5a2 isoforms, and we observed an increased expression of Pde5a1 in damaged skeletal muscles, while Pde5a2 levels remained unchanged. We also cloned and characterized the promoters that control the transcription of Pde5a isoforms, investigating which of the transcription factors predicted by bioinformatics analysis could be involved in their modulation. In conclusion, we found an overexpression of Pde5a1 in compromised muscle and identified an involvement of MyoD and Runx1 in Pde5a1 transcriptional activity.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases , Signal Transduction , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Cyclic GMP/metabolism , Muscle, Skeletal/metabolism
5.
Clin Epigenetics ; 13(1): 173, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34530916

ABSTRACT

BACKGROUND: Thymic epithelial tumors (TETs) are rare neoplasms, originating from epithelial thymic cells. The oncogenic potential of these rare neoplasms is still largely undefined, and a deeper molecular characterization could result in a relevant advance in their management, greatly improving diagnosis, prognosis and treatment choice. Deregulation of N6-methyladenosine (m6A) RNA modification, catalyzed by the METTL3/METTL14 methyltransferase complex, is emerging as a relevant event in cell differentiation and carcinogenesis. Various studies have reported that altered expression of METTL3 is associated with an aggressive malignant phenotype and favors migration and invasiveness, but its role in Thymic Tumors remains unknown. RESULTS: In this study, we characterized that METTL3 contributes to Thymic Epithelial Tumor phenotype. We evidenced that METTL3 is overexpressed in tumor tissue compared to normal counterpart. Silencing of METTL3 expression in thymic carcinoma cells results in reduced cell proliferation and overall translation rate. Of note, METTL3 is responsible for the induction of c-MYC expression in TET cells. Specifically, high expression of c-MYC protein is enabled by lncRNA MALAT1, which is methylated and delocalized by METTL3. Interestingly, blocking of c-MYC by using JQ1 inhibitor cooperates with METTL3 depletion in the inhibition of proliferation and induction of cell death. CONCLUSION: This study highlighted METTL3 as a tumor promoter in Thymic tumors and c-MYC as a promising target to be exploited for the treatment of TET.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation, Neoplastic/genetics , Methyltransferases/genetics , Neoplasms, Glandular and Epithelial/genetics , Proto-Oncogene Proteins c-myc/genetics , Thymus Neoplasms/genetics , Transcription Factors/genetics , Cells, Cultured , Humans
7.
Cell Death Dis ; 11(11): 959, 2020 11 07.
Article in English | MEDLINE | ID: mdl-33161413

ABSTRACT

Long non-coding RNAs are emerging as new molecular players involved in many biological processes, such as proliferation, apoptosis, cell cycle, migration, and differentiation. Their aberrant expression has been reported in variety of diseases. The aim of this study is the identification and functional characterization of clinically relevant lncRNAs responsible for the inhibition of miR-145-5p, a key tumor suppressor in thymic epithelial tumors (TETs). Starting from gene expression analysis by microarray in a cohort of fresh frozen thymic tumors and normal tissues, we identified LINC00174 as upregulated in TET. Interestingly, LINC00174 expression is positively correlated with a 5-genes signature in TETs. Survival analyses, performed on the TCGA dataset, showed that LINC00174 and its associated 5-genes signature are prognostic in TETs. Specifically, we show that LINC00174 favors the expression of SYBU, FEM1B, and SCD5 genes by sponging miR-145-5p, a well-known tumor suppressor microRNA downregulated in a variety of tumors, included TETs. Functionally, LINC00174 impacts on cell migration and lipid metabolism. Specifically, SCD5, one of the LINC00174-associated genes, is implicated in the control of lipid metabolism and promotes thymic cancer cells migration. Our study highlights that LINC00174 and its associated gene signature are relevant prognostic indicators in TETs. Of note, we here show that a key controller of lipid metabolism, SCD5, augments the migration ability of TET cells, creating a link between lipids and motility, and highlighting these pathways as relevant targets for the development of novel therapeutic approaches for TET.


Subject(s)
Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic , Lipid Metabolism , Neoplasms, Glandular and Epithelial/pathology , RNA, Long Noncoding/genetics , Thymus Neoplasms/pathology , Apoptosis , Biomarkers, Tumor/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Proliferation , Gene Expression Profiling , Humans , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Prognosis , Survival Rate , Thymus Neoplasms/genetics , Thymus Neoplasms/metabolism , Tumor Cells, Cultured
8.
Int J Mol Sci ; 21(8)2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32326334

ABSTRACT

Phosphodiesterase 2A (PDE2A) is a cAMP-cGMP hydrolyzing enzyme essential for mouse development and the PDE2A knockout model (PDE2A-/-) is embryonic lethal. Notably, livers of PDE2A-/- embryos at embryonic day 14.5 (E14.5) have extremely reduced size. Morphological, cellular and molecular analyses revealed loss of integrity in the PDE2A-/- liver niche that compromises the hematopoietic function and maturation. Hematopoietic cells isolated from PDE2A-/- livers are instead able to differentiate in in vitro assays, suggesting the absence of blood cell-autonomous defects. Apoptosis was revealed in hepatoblasts and at the endothelial and stromal compartments in livers of PDE2A-/- embryos. The increase of the intracellular cAMP level and of the inducible cAMP early repressor (ICER) in liver of PDE2A-/- embryos might explain the impairment of liver development by downregulating the expression of the anti-apoptotic gene Bcl2. In summary, we propose PDE2A as an essential gene for integrity maintenance of liver niche and the accomplishment of hematopoiesis.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Hematopoiesis/genetics , Liver/embryology , Liver/metabolism , Organogenesis/genetics , Animals , Apoptosis/genetics , Biomarkers , Cell Differentiation , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Genotype , Immunohistochemistry , Mice , Mice, Transgenic , Mutation , Stem Cells/cytology , Stem Cells/metabolism , Stromal Cells/metabolism
9.
Stem Cells Int ; 2019: 7548160, 2019.
Article in English | MEDLINE | ID: mdl-31827532

ABSTRACT

Satellite cells (SC) are the stem cells of skeletal muscles. They are quiescent in adult animals but resume proliferation to allow muscle hypertrophy or regeneration after injury. The mechanisms balancing quiescence, self-renewal, and differentiation of SC are difficult to analyze in vivo owing to their complexity and in vitro because the staminal character of SC is lost when they are removed from the niche and is not adequately reproduced in the culture models currently available. To overcome these difficulties, we set up a culture model of the myogenic C2C12 cell line in suspension. When C2C12 cells are cultured in suspension, they enter a state of quiescence and form three-dimensional aggregates (myospheres) that produce the extracellular matrix and express markers of quiescent SC. In the initial phase of culture, a portion of the cells fuses in syncytia and abandons the myospheres. The remaining cells are mononucleated and quiescent but resume proliferation and differentiation when plated in a monolayer. The notch pathway controls the quiescent state of the cells as shown by the fact that its inhibition leads to the resumption of differentiation. Within this context, notch3 appears to play a central role in the activity of this pathway since the expression of notch1 declines soon after aggregation. In summary, the culture model of C2C12 in suspension may be used to study the cellular interactions of muscle stem cells and the pathways controlling SC quiescence entrance and maintenance.

10.
Front Physiol ; 10: 897, 2019.
Article in English | MEDLINE | ID: mdl-31354530

ABSTRACT

Sarcopenia is the age-related progressive loss of skeletal muscle mass and strength finally leading to poor physical performance. Impaired myogenesis contributes to the pathogenesis of sarcopenia, while mitochondrial dysfunctions are thought to play a primary role in skeletal muscle loss during aging. Here we studied the link between myogenesis and metabolism. In particular, we analyzed the effect of the metabolic modulator trimetazidine (TMZ) on myogenesis in aging. We show that reprogramming the metabolism by TMZ treatment for 12 consecutive days stimulates myogenic gene expression in skeletal muscle of 22-month-old mice. Our data also reveal that TMZ increases the levels of mitochondrial proteins and stimulates the oxidative metabolism in aged muscles, this finding being in line with our previous observations in cachectic mice. Moreover, we show that, besides TMZ also other types of metabolic modulators (i.e., 5-Aminoimidazole-4-Carboxamide Ribofuranoside-AICAR) can stimulate differentiation of skeletal muscle progenitors in vitro. Overall, our results reveal that reprogramming the metabolism stimulates myogenesis while triggering mitochondrial proteins synthesis in vivo during aging. Together with the previously reported ability of TMZ to increase muscle strength in aged mice, these new data suggest an interesting non-invasive therapeutic strategy which could contribute to improving muscle quality and neuromuscular communication in the elderly, and counteracting sarcopenia.

11.
J Cell Sci ; 132(5)2019 03 04.
Article in English | MEDLINE | ID: mdl-30745336

ABSTRACT

Ataxia telangiectasia is a rare, multi system disease caused by ATM kinase deficiency. Atm-knockout mice recapitulate premature aging, immunodeficiency, cancer predisposition, growth retardation and motor defects, but not cerebellar neurodegeneration and ataxia. We explored whether Atm loss is responsible for skeletal muscle defects by investigating myofiber morphology, oxidative/glycolytic activity, myocyte ultrastructural architecture and neuromuscular junctions. Atm-knockout mice showed reduced muscle and fiber size. Atrophy, protein synthesis impairment and a switch from glycolytic to oxidative fibers were detected, along with an increase of in expression of slow and fast myosin types (Myh7, and Myh2 and Myh4, respectively) in tibialis anterior and solei muscles isolated from Atm-knockout mice. Transmission electron microscopy of tibialis anterior revealed misalignments of Z-lines and sarcomeres and mitochondria abnormalities that were associated with an increase in reactive oxygen species. Moreover, neuromuscular junctions appeared larger and more complex than those in Atm wild-type mice, but with preserved presynaptic terminals. In conclusion, we report for the first time that Atm-knockout mice have clear morphological skeletal muscle defects that will be relevant for the investigation of the oxidative stress response, motor alteration and the interplay with peripheral nervous system in ataxia telangiectasia.


Subject(s)
Aging, Premature/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia/metabolism , Immunologic Deficiency Syndromes/genetics , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Neoplasms/genetics , Animals , Ataxia Telangiectasia/physiopathology , Cells, Cultured , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Muscle, Skeletal/abnormalities , Muscle, Skeletal/ultrastructure , Reactive Oxygen Species/metabolism , Sarcomeres/ultrastructure
12.
Cell Death Dis ; 10(1): 17, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30622242

ABSTRACT

To perform their regulatory functions, microRNAs (miRNAs) must assemble with any of the four mammalian Argonaute (Ago) family of proteins, Ago1-4, into an effector complex known as the RNA-induced silencing complex (RISC). While the mature miRNA guides the RISC complex to its target mRNA, the Ago protein represses mRNA translation. The specific roles of the various Ago members in mediating miRNAs activity, however, haven't been clearly established. In this study, we investigated the contribution of Ago2, the only human Ago protein endowed with nuclease activity, to the function of tumor-suppressor miR-145-5p in breast cancer (BC). We show that miR-145-5p and Ago2 protein are concomitantly downregulated in BC tissues and that restoration of miR-145-5p expression in BC cells leads to Ago2 protein induction through the loosening of Ago2 mRNA translational repression. Functionally, miR-145-5p exerts its inhibitory activity on cell migration only in presence of Ago2, while, upon Ago2 depletion, we observed increased miR-145/Ago1 complex and enhanced cell motility. Profiling by microarray of miR-145-5p target mRNAs, in BC cells depleted or not of Ago2, revealed that miR-145-5p drives Ago2-dependent and -independent activities. Our results highlight that the Ago2 protein in cancer cells strictly dictates miR-145-5p tumor suppressor activity.


Subject(s)
Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression , MicroRNAs/genetics , MicroRNAs/metabolism , A549 Cells , Breast Neoplasms/pathology , Cell Cycle , Cell Movement , Eukaryotic Initiation Factors/metabolism , Female , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Polyribosomes/metabolism , Protein Biosynthesis , Transfection
13.
Mol Cancer ; 16(1): 88, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28486946

ABSTRACT

BACKGROUND: Thymoma and thymic carcinoma are the most frequent subtypes of thymic epithelial tumors (TETs). A relevant advance in TET management could derive from a deeper molecular characterization of these neoplasms. We previously identified a set of microRNA (miRNAs) differentially expressed in TETs and normal thymic tissues and among the most significantly deregulated we described the down-regulation of miR-145-5p in TET. Here we describe the mRNAs diversely regulated in TETs and analyze the correlation between these and the miRNAs previously identified, focusing in particular on miR-145-5p. Then, we examine the functional role of miR-145-5p in TETs and its epigenetic transcriptional regulation. METHODS: mRNAs expression profiling of a cohort of fresh frozen TETs and normal tissues was performed by microarray analysis. MiR-145-5p role in TETs was evaluated in vitro, modulating its expression in a Thymic Carcinoma (TC1889) cell line. Epigenetic transcriptional regulation of miR-145-5p was examined by treating the TC1889 cell line with the HDAC inhibitor Valproic Acid (VPA). RESULTS: Starting from the identification of a 69-gene signature of miR-145-5p putative target mRNAs, whose expression was inversely correlated to that of miR-145-5p, we followed the expression of some of them in vitro upon overexpression of miR-145-5p; we observed that this resulted in the down-regulation of the target genes, impacting on TETs cancerous phenotype. We also found that VPA treatment of TC1889 cells led to miR-145-5p up-regulation and concomitant down-regulation of miR-145-5p target genes and exhibited antitumor effects, as indicated by the induction of cell cycle arrest and by the reduction of cell viability, colony forming ability and migration capability. The importance of miR-145-5p up-regulation mediated by VPA is evidenced by the fact that hampering miR-145-5p activity by a LNA inhibitor reduced the impact of VPA treatment on cell viability and colony forming ability of TET cells. Finally, we observed that VPA was also able to enhance the response of TET cells to cisplatin and erlotinib. CONCLUSIONS: Altogether our results suggest that the epigenetic regulation of miR-145-5p expression, as well as the modulation of its functional targets, could be relevant players in tumor progression and treatment response in TETs.


Subject(s)
Epigenesis, Genetic , MicroRNAs/genetics , Neoplasms, Glandular and Epithelial/genetics , Thymoma/genetics , Thymus Neoplasms/genetics , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Erlotinib Hydrochloride/administration & dosage , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/pathology , RNA, Messenger/genetics , Thymoma/drug therapy , Thymoma/pathology , Thymus Neoplasms/drug therapy , Thymus Neoplasms/pathology
14.
Skelet Muscle ; 5: 18, 2015.
Article in English | MEDLINE | ID: mdl-26146542

ABSTRACT

BACKGROUND: The Y-box protein MSY3/Csda represses myogenin transcription in skeletal muscle by binding a highly conserved cis-acting DNA element located just upstream of the myogenin minimal promoter (myogHCE). It is not known how this MSY3 activity is controlled in skeletal muscle. In this study, we provide multiple lines of evidence showing that the post-translational phosphorylation of MSY3 by Akt kinase modulates the MSY3 repression of myogenin. METHODS: Skeletal muscle and myogenic C2C12 cells were used to study the effects of MSY3 phosphorylation in vivo and in vitro on its sub-cellular localization and activity, by blocking the IGF1/PI3K/Akt pathway, by Akt depletion and over-expression, and by mutating potential MSY3 phosphorylation sites. RESULTS: We observed that, as skeletal muscle progressed from perinatal to postnatal and adult developmental stages, MSY3 protein became gradually dephosphorylated and accumulated in the nucleus. This correlated well with the reduction of phosphorylated active Akt. In C2C12 myogenic cells, blocking the IGF1/PI3K/Akt pathway using LY294002 inhibitor reduced MSY3 phosphorylation levels resulting in its accumulation in the nuclei. Knocking down Akt expression increased the amount of dephosphorylated MSY3 and reduced myogenin expression and muscle differentiation. MSY3 phosphorylation by Akt in vitro impaired its binding at the MyogHCE element, while blocking Akt increased MSY3 binding activity. While Akt over-expression rescued myogenin expression in MSY3 overexpressing myogenic cells, ablation of the Akt substrate, (Ser126 located in the MSY3 cold shock domain) promoted MSY3 accumulation in the nucleus and abolished this rescue. Furthermore, forced expression of Akt in adult skeletal muscle induced MSY3 phosphorylation and myogenin derepression. CONCLUSIONS: These results support the hypothesis that MSY3 phosphorylation by Akt interferes with MSY3 repression of myogenin circuit activity during muscle development. This study highlights a previously undescribed Akt-mediated signaling pathway involved in the repression of myogenin expression in myogenic cells and in mature muscle. Given the significance of myogenin regulation in adult muscle, the Akt/MSY3/myogenin regulatory circuit is a potential therapeutic target to counteract muscle degenerative disease.

15.
J Cachexia Sarcopenia Muscle ; 4(2): 125-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23314905

ABSTRACT

Skeletal muscle regeneration is the process that ensures tissue repair after damage by injury or in degenerative diseases such as muscular dystrophy. Satellite cells, the adult skeletal muscle progenitor cells, are commonly considered to be the main cell type involved in skeletal muscle regeneration. Their mechanism of action in this process is extensively characterized. However, evidence accumulated in the last decade suggests that other cell types may participate in skeletal muscle regeneration. Although their actual contribution to muscle formation and regeneration is still not clear; if properly manipulated, these cells may become new suitable and powerful sources for cell therapy of skeletal muscle degenerative diseases. Mesoangioblasts, vessel associated stem/progenitor cells with high proliferative, migratory and myogenic potential, are very good candidates for clinical applications and are already in clinical experimentation. In addition, pluripotent stem cells are very promising sources for regeneration of most tissues, including skeletal muscle. Conditions such as muscle cachexia or aging that severely alter homeostasis may be counteracted by transplantation of donor and/or recruitment and activation of resident muscle stem/progenitor cells. Advantages and limitations of different cell therapy approaches will be discussed.

16.
Stem Cells ; 27(1): 157-64, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18845762

ABSTRACT

Mesoangioblasts have been characterized as a population of vessel-associated stem cells able to differentiate into several mesodermal cell types, including skeletal muscle. Here, we report that the paired box transcription factor Pax3 plays a crucial role in directing mouse mesoangioblasts toward skeletal myogenesis in vitro and in vivo. Mesoangioblasts isolated from the aorta of Pax3 null embryos are severely impaired in skeletal muscle differentiation, whereas most other differentiation programs are not affected by the absence of Pax3. Moreover, Pax3(-/-) null mesoangioblasts failed to rescue the myopathic phenotype of the alpha-sarcoglycan mutant mouse. In contrast, mesoangioblasts from Pax3 gain of function, Pax3(PAX3-FKHR/+), mice display enhanced myogenesis in vitro and are more efficient in regenerating new muscle fibers in this model of muscular dystrophy. These data demonstrate that Pax3 is required for the differentiation of mesoangioblast stem cells into skeletal muscle, in keeping with its role in orchestrating entry into the myogenic program.


Subject(s)
Blood Vessels/cytology , Cell Differentiation , Embryo, Mammalian/cytology , Mesoderm/cytology , Muscle, Skeletal/cytology , Paired Box Transcription Factors/metabolism , Alkaline Phosphatase/metabolism , Animals , Blood Vessels/enzymology , Bone and Bones/cytology , Cell Proliferation , Cell Shape , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Mice , Muscle Development , Muscular Dystrophy, Animal/metabolism , Myocytes, Smooth Muscle/cytology , PAX3 Transcription Factor , Paired Box Transcription Factors/deficiency , Phenotype , Sarcoglycans/biosynthesis
17.
Genes Dev ; 22(15): 2125-38, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676817

ABSTRACT

Myogenin is the dominant transcriptional regulator of embryonic and fetal muscle differentiation and during maturation is profoundly down-regulated. We show that a highly conserved 17-bp DNA cis-acting sequence element located upstream of the myogenin promoter (myogHCE) is essential for postnatal repression of myogenin in transgenic animals. We present multiple lines of evidence supporting the idea that repression is mediated by the Y-box protein MSY-3. Electroporation in vivo shows that myogHCE and MSY-3 are required for postnatal repression. We further show that, in the C2C12 cell culture system, ectopic MSY-3 can repress differentiation, while reduced MSY-3 promotes premature differentiation. MSY-3 binds myogHCE simultaneously with the homeodomain protein Pbx in postnatal innervated muscle. We therefore propose a model in which the myogHCE motif operates as a switch by specifying opposing functions; one that was shown previously is regulated by MyoD and Pbx and it specifies a chromatin opening, gene-activating function at the time myoblasts begin to differentiate; the other includes MYS-3 and Pbx, and it specifies a repression function that operates during and after postnatal muscle maturation in vivo and in myoblasts before they begin to differentiate.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Muscle, Skeletal/growth & development , Myogenin/genetics , RNA-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Electroporation , Genetic Vectors , Homeodomain Proteins/metabolism , Lentivirus/genetics , Mice , MyoD Protein/genetics , MyoD Protein/physiology , Myoblasts/physiology , Myogenin/physiology , Pre-B-Cell Leukemia Transcription Factor 1 , Transcription Factors/metabolism
18.
Proc Natl Acad Sci U S A ; 103(45): 16995-7000, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-17077152

ABSTRACT

Inflammatory myopathies (IM) are acquired diseases of skeletal muscle comprising dermatomyositis (DM), polymyositis (PM), and inclusion-body myositis (IBM). Immunosuppressive therapies, usually beneficial for DM and PM, are poorly effective in IBM. We report the isolation and characterization of mesoangioblasts, vessel-associated stem cells, from diagnostic muscle biopsies of IM. The number of cells isolated, proliferation rate and lifespan, markers expression, and ability to differentiate into smooth muscle do not differ among normal and IM mesoangioblasts. At variance with normal, DM and PM mesoangioblasts, cells isolated from IBM, fail to differentiate into skeletal myotubes. These data correlate with lack in connective tissue of IBM muscle of alkaline phosphatase (ALP)-positive cells, conversely dramatically increased in PM and DM. A myogenic inhibitory basic helix-loop-helix factor B3 is highly expressed in IBM mesoangioblasts. Indeed, silencing this gene or overexpressing MyoD rescues the myogenic defect of IBM mesoangioblasts, opening novel cell-based therapeutic strategies for this crippling disorder.


Subject(s)
Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , MyoD Protein/genetics , MyoD Protein/metabolism , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/pathology , Alkaline Phosphatase/metabolism , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cells, Cultured , Gene Expression , Gene Silencing , Humans , Muscle Development , Muscle, Skeletal/blood supply , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Myositis, Inclusion Body/therapy , RNA, Small Interfering/genetics
19.
Dev Biol ; 283(1): 171-9, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15890335

ABSTRACT

Biochemical and cell culture studies have characterized the myocyte enhancer factor 2 (MEF2) transcriptional regulatory proteins as obligatory partners for the myogenic regulatory factors (MRFs) in the differentiation of myogenic cells in culture. However, the role of MEF2 activation in somitic myogenesis has not been fully characterized. Here, we report a critical interaction between the p38 mitogen-activated protein kinase (p38 MAPK) and MEF2 in the developing somite myotome. We document expression of MEF2A and p38 MAPK proteins in the somite of 9.5 dpc mouse embryos concurrent with Myf 5 protein expression. We also observed that abrogation of p38 MAPK signaling blocks MEF2 activation using a MEF2 transgenic 'sensor' mouse. Inhibition of p38 MAPK signaling concurrently inhibited myogenic differentiation in somite cultures and in embryos in vivo using transplacental injection of a p38 inhibitor (SB203580). Finally, we document that commitment to the myogenic lineage is not appreciably affected by p38 MAPK inhibition since the activation of an early marker of myogenic commitment (Myf 5) occurs normally when p38 MAPK signaling is inhibited. Thus, we present novel evidence indicating a crucial role for p38 MAPK signaling to the MEF2 transcriptional regulators during early mammalian somite development and myotome formation.


Subject(s)
DNA-Binding Proteins/genetics , Embryonic Development , Gene Expression Regulation, Developmental , Muscle, Skeletal/embryology , Signal Transduction , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Crosses, Genetic , DNA-Binding Proteins/metabolism , Female , MEF2 Transcription Factors , Male , Mice , Mice, Transgenic , Myogenic Regulatory Factors , Myogenin/genetics , Myosin Light Chains/genetics , Transcription Factors/metabolism , beta-Galactosidase/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
20.
Circ Res ; 95(9): 911-21, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15472116

ABSTRACT

Cardiac myocytes have been traditionally regarded as terminally differentiated cells that adapt to increased work and compensate for disease exclusively through hypertrophy. However, in the past few years, compelling evidence has accumulated suggesting that the heart has regenerative potential. Recent studies have even surmised the existence of resident cardiac stem cells, endothelial cells generating cardiomyocytes by cell contact or extracardiac progenitors for cardiomyocytes, but these findings are still controversial. We describe the isolation of undifferentiated cells that grow as self-adherent clusters (that we have termed "cardiospheres") from subcultures of postnatal atrial or ventricular human biopsy specimens and from murine hearts. These cells are clonogenic, express stem and endothelial progenitor cell antigens/markers, and appear to have the properties of adult cardiac stem cells. They are capable of long-term self-renewal and can differentiate in vitro and after ectopic (dorsal subcutaneous connective tissue) or orthotopic (myocardial infarction) transplantation in SCID beige mouse to yield the major specialized cell types of the heart: myocytes (ie, cells demonstrating contractile activity and/or showing cardiomyocyte markers) and vascular cells (ie, cells with endothelial or smooth muscle markers).


Subject(s)
Cell Separation/methods , Myocardium/cytology , Myocytes, Cardiac/cytology , Stem Cells/cytology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Cell Aggregation , Cell Differentiation , Cell Division , Cells, Cultured/cytology , Child , Child, Preschool , Clone Cells/cytology , Coculture Techniques , Flow Cytometry , Genes, Reporter , Humans , Immunophenotyping , Infant , Infant, Newborn , Mice , Mice, Mutant Strains , Mice, SCID , Mice, Transgenic , Middle Aged , Myocardial Contraction , Myocardial Infarction/therapy , Organoids/cytology , Rats , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...