Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
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.

2.
Cell Death Dis ; 9(3): 314, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29472706

ABSTRACT

Hereditary deficiencies in DNA damage signaling are invariably associated with cancer predisposition, immunodeficiency, radiation sensitivity, gonadal abnormalities, premature aging, and tissue degeneration. ATM kinase has been established as a central player in DNA double-strand break repair and its deficiency causes ataxia telangiectasia, a rare, multi-system disease with no cure. So ATM represents a highly attractive target for the development of novel types of gene therapy or transplantation strategies. Atm tamoxifen-inducible mouse models were generated to explore whether Atm reconstitution is able to restore Atm function in an Atm-deficient background. Body weight, immunodeficiency, spermatogenesis, and radioresistance were recovered in transgenic mice within 1 month from Atm induction. Notably, life span was doubled after Atm restoration, mice were protected from thymoma and no cerebellar defects were observed. Atm signaling was functional after DNA damage in vivo and in vitro. In summary, we propose a new Atm mouse model to investigate novel therapeutic strategies for ATM activation in ataxia telangiectasia disease.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia/enzymology , Disease Models, Animal , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/immunology , Ataxia Telangiectasia Mutated Proteins/genetics , DNA Damage , Enzyme Activation , Female , Humans , Male , Mice , Mice, Transgenic , Phenotype , Signal Transduction
3.
J Mol Cell Cardiol ; 98: 146-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27418252

ABSTRACT

Proper ß-adrenergic signaling is indispensable for modulating heart frequency. Studies on extremely-low-frequency pulsed electromagnetic field (ELF-PEMF) effects in the heart beat function are contradictory and no definitive conclusions were obtained so far. To investigate the interplay between ELF-PEMF exposure and ß-adrenergic signaling, cultures of primary murine neonatal cardiomyocytes and of sinoatrial node were exposed to ELF-PEMF and short and long-term effects were evaluated. The ELF-PEMF generated a variable magnetic induction field of 0-6mT at a frequency of 75Hz. Exposure to 3mT ELF-PEMF induced a decrease of contraction rate, Ca(2+) transients, contraction force, and energy consumption both under basal conditions and after ß-adrenergic stimulation in neonatal cardiomyocytes. ELF-PEMF exposure inhibited ß-adrenergic response in sinoatrial node (SAN) region. ELF-PEMF specifically modulated ß2 adrenergic receptor response and the exposure did not modify the increase of contraction rate after adenylate cyclase stimulation by forskolin. In HEK293T cells transfected with ß1 or ß2 adrenergic receptors, ELF-PEMF exposure induced a rapid and selective internalization of ß2 adrenergic receptor. The ß-adrenergic signaling, was reduced trough Gi protein by ELF-PEMF exposure since the phosphorylation level of phospholamban and the PI3K pathway were impaired after isoproterenol stimulation in neonatal cardiomyocytes. Long term effects of ELF-PEMF exposure were assessed in cultures of isolated cardiomyocytes. ELF-PEMF counteracts cell size increase, the generation of binucleated of cardiomyocytes and prevents the up-regulation of hypertrophic markers after ß-adrenergic stimulation, indicating an inhibition of cell growth and maturation. These data show that short and long term exposure to ELF-PEMF induces a reduction of cardiac ß-adrenergic response at molecular, functional and adaptative levels.


Subject(s)
Electromagnetic Fields , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/radiation effects , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Agonists/pharmacology , Algorithms , Animals , Calcium/metabolism , Calcium Signaling , Energy Metabolism/drug effects , Energy Metabolism/radiation effects , Mice , Models, Biological , Myocardial Contraction/drug effects , Myocardial Contraction/radiation effects , Myocytes, Cardiac/drug effects , Receptors, Adrenergic, beta/genetics , Signal Transduction/drug effects , Signal Transduction/radiation effects , Sinoatrial Node/drug effects , Sinoatrial Node/physiology , Sinoatrial Node/radiation effects
4.
Chromosoma ; 125(2): 189-203, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26440409

ABSTRACT

Meiosis is the biological process that, after a cycle of DNA replication, halves the cellular chromosome complement, leading to the formation of haploid gametes. Haploidization is achieved via two successive rounds of chromosome segregation, meiosis I and II. In mammals, during prophase of meiosis I, homologous chromosomes align and synapse through a recombination-mediated mechanism initiated by the introduction of DNA double-strand breaks (DSBs) by the SPO11 protein. In male mice, if SPO11 expression and DSB number are reduced below heterozygosity levels, chromosome synapsis is delayed, chromosome tangles form at pachynema, and defective cells are eliminated by apoptosis at epithelial stage IV at a spermatogenesis-specific endpoint. Whether DSB levels produced in Spo11 (+/-) spermatocytes represent, or approximate, the threshold level required to guarantee successful homologous chromosome pairing is unknown. Using a mouse model that expresses Spo11 from a bacterial artificial chromosome, within a Spo11 (-/-) background, we demonstrate that when SPO11 expression is reduced and DSBs at zygonema are decreased (approximately 40 % below wild-type level), meiotic chromosome pairing is normal. Conversely, DMC1 foci number is increased at pachynema, suggesting that under these experimental conditions, DSBs are likely made with delayed kinetics at zygonema. In addition, we provide evidences that when zygotene-like cells receive enough DSBs before chromosome tangles develop, chromosome synapsis can be completed in most cells, preventing their apoptotic elimination.


Subject(s)
Chromosome Pairing , DNA Breaks, Double-Stranded , Endodeoxyribonucleases/metabolism , Meiosis , Spermatocytes/cytology , Animals , Chromosomes/genetics , Chromosomes/metabolism , Endodeoxyribonucleases/genetics , Female , Male , Meiotic Prophase I , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatocytes/metabolism , Spermatogenesis
5.
Int J Dev Biol ; 57(2-4): 281-7, 2013.
Article in English | MEDLINE | ID: mdl-23784839

ABSTRACT

During meiosis, phosphorylation of H2AX is one of the earliest cellular responses to the generation of DNA double-strand breaks (DSBs) by the SPO11 topoisomerase. ATM is the kinase which mediates the formation of phosphorylated H2AX (H2AX) meiotic foci, while ATR is the kinase which signals chromosome asynapsis at the level of the XY bivalent. To investigate the possible role of ATR also in DNA damage signalling in meiotic cells, we studied the effect of UV radiation and chemotherapy drugs on H2AX phosphorylation and ATR relocalization in mouse pachytene spermatocytes. Here, we report that UV, a single strand break DNA-damaging agent, induces ATR relocalization from the XY sex body to nuclear foci and intense H2AX phosphorylation. Other DNA damage proteins such as MDC1, NBS1 and 53BP1 showed a similar relocalization following UVA microirradiation of spermatocytes. We found that DNA damage induced by UV increased the intensity and the number of H2AX foci also in Atm null spermatocytes. Inhibition of RNA synthesis was found to induce the formation of H2AX foci, but it did not influence the DNA damage response to UV irradiation. Finally, exposure of spermatocytes to double strand break DNA-damaging agents such as cisplatin, bleomycin or etoposide also induced ATR relocalization and intense H2AX phosphorylation and led to anomalies in synaptonemal assembly. Our results demonstrate that DNA damage induced by genotoxic stress can activate ATR and influence meiotic chromatin remodelling through H2AX phosphorylation, likely as part of a response which normally ensures germ cell genomic integrity.


Subject(s)
DNA Damage , Spermatocytes/metabolism , Ultraviolet Rays , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/physiology , Blotting, Western , Cell Differentiation , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Fluorescent Antibody Technique , Histones/genetics , Histones/metabolism , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Mice, Knockout , Spermatocytes/pathology , Spermatocytes/radiation effects
6.
PLoS One ; 8(1): e54271, 2013.
Article in English | MEDLINE | ID: mdl-23372698

ABSTRACT

Neural stem cells (NSCs) are self-renewing cells that can differentiate into multiple neural lineages and repopulate regions of the brain after injury. We have investigated the role of endocannabinoids (eCBs), endogenous cues that modulate neuronal functions including neurogenesis, and their receptors CB(1) and CB(2) in mouse NSCs. Real-time PCR and Western blot analyses indicated that CB(1) is present at higher levels than CB(2) in NSCs. The eCB anandamide (AEA) or the CB(1)-specific agonist ACEA enhanced NSC differentiation into neurons, but not astrocytes and oligodendrocytes, whereas the CB(2)-specific agonist JWH133 was ineffective. Conversely, the effect of AEA was inhibited by CB(1), but not CB(2), antagonist, corroborating the specificity of the response. CB(1) activation also enhanced maturation of neurons, as indicated by morphometric analysis of neurites. CB(1) stimulation caused long-term inhibition of the ERK1/2 pathway. Consistently, pharmacological inhibition of the ERK1/2 pathway recapitulated the effects exerted by CB(1) activation on neuronal differentiation and maturation. Lastly, gene array profiling showed that CB(1) activation augmented the expression of genes involved in neuronal differentiation while decreasing that of stemness genes. These results highlight the role of CB(1) in the regulation of NSC fate and suggest that its activation may represent a pro-neuronal differentiation signal.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Cell Differentiation/drug effects , Neural Stem Cells/cytology , Neurons/cytology , Receptor, Cannabinoid, CB1/genetics , Animals , Arachidonic Acids/pharmacology , Cannabinoids/pharmacology , Embryo, Mammalian , Endocannabinoids/pharmacology , Gene Expression/drug effects , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Microarray Analysis , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/metabolism , Polyunsaturated Alkamides/pharmacology , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Signal Transduction/drug effects
7.
Cell Mol Life Sci ; 69(24): 4177-90, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22802127

ABSTRACT

Estrogen (E(2)) regulates spermatogenesis, yet its direct target genes have not been identified in the testis. Here, we cloned the proximal 5' flanking region of the mouse fatty acid amide hydrolase (faah) gene upstream of the luciferase reporter gene, and demonstrated its promoter activity and E(2) inducibility in primary mouse Sertoli cells. Specific mutations in the E(2) response elements (ERE) of the faah gene showed that two proximal ERE sequences (ERE2/3) are essential for E(2)-induced transcription, and chromatin immunoprecipitation experiments showed that E(2) induced estrogen receptor ß binding at ERE2/3 sites in the faah promoter in vivo. Moreover, the histone demethylase LSD1 was found to be associated with ERE2/3 sites and to play a role in mediating E(2) induction of FAAH expression. E(2) induced epigenetic modifications at the faah proximal promoter compatible with transcriptional activation by remarkably decreasing methylation of both DNA at CpG site and histone H3 at lysine 9. Finally, FAAH silencing abolished E(2) protection against apoptosis induced by the FAAH substrate anandamide. Taken together, our results identify FAAH as the first direct target of E(2).


Subject(s)
Amidohydrolases/genetics , Estrogens/pharmacology , Gene Expression Regulation , Oxidoreductases, N-Demethylating/physiology , Sertoli Cells/metabolism , Amidohydrolases/chemistry , Amidohydrolases/physiology , Animals , Apoptosis , Base Sequence , DNA Methylation/drug effects , Estrogen Receptor beta/metabolism , Estrogen Receptor beta/physiology , Histone Demethylases , Histones/metabolism , Male , Methylation , Mice , Molecular Sequence Data , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Promoter Regions, Genetic , Sertoli Cells/drug effects
8.
PLoS One ; 5(2): e9064, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-20140225

ABSTRACT

A critical step of spermatogenesis is the entry of mitotic spermatogonia into meiosis. Progresses on these topics are hampered by the lack of an in vitro culture system allowing mouse spermatogonia differentiation and entry into meiosis. Previous studies have shown that mouse pachytene spermatocytes cultured in simulated microgravity (SM) undergo a spontaneous meiotic progression. Here we report that mouse mitotic spermatogonia cultured under SM with a rotary cell culture system (RCCS) enter into meiosis in the absence of any added exogenous factor or contact with somatic cells. We found that isolated Kit-positive spermatogonia under the RCCS condition enter into the prophase of the first meiotic division (leptotene stage), as monitored by chromosomal organization of the synaptonemal complex 3 protein (Scp3) and up-regulation of several pro-meiotic genes. SM was found to activate the phosphatidyl inositol 3 kinase (PI3K) pathway and to induce in Kit-positive spermatogonia the last round of DNA replication, typical of the preleptotene stage. A PI3K inhibitor abolished Scp3 induction and meiotic entry stimulated by RCCS conditions. A positive effect of SM on germ cell differentiation was also observed in undifferentiated (Kit-negative) spermatogonia, in which RCCS conditions stimulate the expression of Kit and Stra8. In conclusion, SM is an artificial environmental condition which promotes postnatal male germ cell differentiation and might provide a tool to study the molecular mechanisms underlying the switch from mitosis to meiosis in mammals.


Subject(s)
Cell Differentiation/physiology , Meiosis/physiology , Spermatogonia/metabolism , Weightlessness Simulation , Animals , Blotting, Western , Cell Culture Techniques , Cell Differentiation/genetics , Cells, Cultured , DNA/biosynthesis , Enzyme Activation , Male , Meiosis/genetics , Mice , Microscopy, Confocal , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Spermatocytes/cytology , Spermatocytes/metabolism , Spermatogonia/cytology
9.
J Cell Sci ; 123(Pt 6): 871-80, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20159962

ABSTRACT

In the mouse, three genes that are homologous to the Drosophila Nanos (Nos) gene have been identified. Deletion of one of these genes, Nanos2, results in male sterility, owing to loss of germ cells during fetal life. Before apoptosis, Nanos2-null gonocytes enter meiosis, suggesting that Nanos2 functions as a meiotic repressor. Here, we show that Nanos2 is continuously expressed in male germ cells from fetal gonocytes to postnatal spermatogonial stem cells. We observed that the promeiotic factor AtRA, an analog of retinoic acid (RA), downregulates NANOS2 levels, in both fetal and postnatal gonocytes, while promoting meiosis. Interestingly, FGF9, a growth factor crucial for sex differentiation and survival of fetal gonocytes, upregulates levels of NANOS2 in both male and female primordial germ cells (PGCs) and in premeiotic spermatogonia. This effect was paralleled by an impairment of meiotic entry, suggesting that FGF9 acts as an inhibitor of meiosis through the upregulation of Nanos2. We found that NANOS2 interacts with PUM2, and that these two proteins colocalize in the ribonucleoparticle and polysomal fractions on sucrose gradients, supporting the notion that they bind RNA. Finally, we found that recombinant NANOS2 binds to two spermatogonial mRNAs, Gata2 and Taf7l, which are involved in germ-cell differentiation.


Subject(s)
Carrier Proteins/genetics , Fibroblast Growth Factor 9/pharmacology , Germ Cells/cytology , Germ Cells/metabolism , Meiosis/drug effects , Tretinoin/pharmacology , Animals , Animals, Newborn , Carrier Proteins/metabolism , Down-Regulation/drug effects , Female , Fetus/cytology , Fetus/drug effects , Fetus/metabolism , Gene Expression Regulation, Developmental/drug effects , Germ Cells/drug effects , Male , Mice , Ovum/cytology , Ovum/drug effects , Ovum/metabolism , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Ribonucleoproteins/metabolism , Spermatozoa/cytology , Spermatozoa/drug effects , Spermatozoa/metabolism , Testis/cytology , Testis/drug effects , Testis/metabolism , Up-Regulation/drug effects
10.
Proc Natl Acad Sci U S A ; 106(27): 11131-6, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19541620

ABSTRACT

The exact role of the endocannabinoid system (ECS) during spermatogenesis has not been clarified. We used purified germ cell fractions representative of all phases of spermatogenesis and primary cultures of spermatogonia. This approach allowed the precise quantification of the cannabinoid receptor ligands, anandamide and 2-arachidonoylglycerol, and of the expression at transcriptional and transductional levels of their metabolic enzymes and receptors. Our data indicate that male mouse germ cells possess an active and complete ECS, which is modulated during meiosis, and suggest the presence of an autocrine endocannabinoid signal during spermatogenesis. Mitotic cells possess higher levels of 2-arachidonoylglycerol, which decrease in spermatocytes and spermatids. Accordingly, spermatogonia express higher and lower levels of 2-arachidonoylglycerol biosynthetic and degrading enzymes, respectively, as compared to meiotic and postmeiotic cells. This endocannabinoid likely plays a pivotal role in promoting the meiotic progression of germ cells by activating CB(2) receptors. In fact, we found that the selective CB(2) receptor agonist, JWH133, induced the Erk 1/2 MAPK phosphorylation cascade in spermatogonia and their progression toward meiosis, because it increased the number of cells positive for SCP3, a marker of meiotic prophase, and the expression of early meiotic prophase genes.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Receptor, Cannabinoid, CB2/metabolism , Spermatogenesis , Animals , Arachidonic Acids/biosynthesis , Cannabinoid Receptor Modulators/biosynthesis , Cannabinoids/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Fluorescent Antibody Technique , Glycerides/biosynthesis , MAP Kinase Signaling System/drug effects , Male , Meiotic Prophase I/drug effects , Mice , Polyunsaturated Alkamides , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/agonists , Spermatogenesis/drug effects , Spermatogonia/cytology , Spermatogonia/drug effects , Spermatogonia/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...