Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Biomolecules ; 13(6)2023 06 03.
Article in English | MEDLINE | ID: mdl-37371518

ABSTRACT

The highly specialized structure and function of neurons depend on a sophisticated organization of the cytoskeleton, which supports a similarly sophisticated system to traffic organelles and cargo vesicles. Mitochondria sustain crucial functions by providing energy and buffering calcium where it is needed. Accordingly, the distribution of mitochondria is not even in neurons and is regulated by a dynamic balance between active transport and stable docking events. This system is finely tuned to respond to changes in environmental conditions and neuronal activity. In this review, we summarize the mechanisms by which mitochondria are selectively transported in different compartments, taking into account the structure of the cytoskeleton, the molecular motors and the metabolism of neurons. Remarkably, the motor proteins driving the mitochondrial transport in axons have been shown to also mediate their transfer between cells. This so-named intercellular transport of mitochondria is opening new exciting perspectives in the treatment of multiple diseases.


Subject(s)
Axons , Neurons , Neurons/metabolism , Axons/metabolism , Mitochondria/metabolism , Cytoskeleton/metabolism , Microtubules/metabolism
3.
Biomedicines ; 11(4)2023 Mar 24.
Article in English | MEDLINE | ID: mdl-37189624

ABSTRACT

Lipoaspirates represent a source of adult stem cells, cytokines, and growth factors of adipocyte origin with immunomodulation and regenerative medicine potential. However, rapid and simple protocols for their purification using self-contained devices that can be deployed at the points of care are lacking. Here, we characterize and benchmark a straightforward mechanical dissociation procedure to collect mesenchymal stem cells (MSCs) and soluble fractions from lipoaspirates. IStemRewind, a benchtop self-contained cell purification device, allowed a one-procedure purification of cells and soluble material from lipoaspirates with minimal manipulation. The recovered cellular fraction contained CD73+, CD90+, CD105+, CD10+ and CD13+ MSCs. These markers were comparably expressed on MSCs isolated using IstemRewind or classic enzymatic dissociation procedures, apart from CD73+ MSCs, which were even more abundant in IStemRewind isolates. IstemRewind-purified MSCs retained viability and differentiation into adipocytes and osteocytes, even after a freezing-thawing cycle. Levels of IL4, IL10, bFGF and VEGF were higher compared to the pro-inflammatory cytokines TNFα, IL1ß and IL6 in the IStemRewind-isolated liquid fraction. In sum, IStemRewind can be useful for straightforward, rapid, and efficient isolation of MSCs and immunomodulatory soluble factors from lipoaspirates, opening the possibility to directly isolate and employ them at the point-of-care.

4.
Cells ; 12(7)2023 04 05.
Article in English | MEDLINE | ID: mdl-37048162

ABSTRACT

Recent proteomic, metabolomic, and transcriptomic studies have highlighted a connection between changes in mitochondria physiology and cellular pathophysiological mechanisms. Secondary assays to assess the function of these organelles appear fundamental to validate these -omics findings. Although mitochondrial membrane potential is widely recognized as an indicator of mitochondrial activity, high-content imaging-based approaches coupled to multiparametric to measure it have not been established yet. In this paper, we describe a methodology for the unbiased high-throughput quantification of mitochondrial membrane potential in vitro, which is suitable for 2D to 3D models. We successfully used our method to analyze mitochondrial membrane potential in monolayers of human fibroblasts, neural stem cells, spheroids, and isolated muscle fibers. Moreover, by combining automated image analysis and machine learning, we were able to discriminate melanoma cells from macrophages in co-culture and to analyze the subpopulations separately. Our data demonstrated that our method is a widely applicable strategy for large-scale profiling of mitochondrial activity.


Subject(s)
Microscopy , Proteomics , Humans , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Fibroblasts/metabolism
5.
Cell Death Dis ; 14(2): 162, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36849544

ABSTRACT

The approved gene therapies for spinal muscular atrophy (SMA), caused by loss of survival motor neuron 1 (SMN1), greatly ameliorate SMA natural history but are not curative. These therapies primarily target motor neurons, but SMN1 loss has detrimental effects beyond motor neurons and especially in muscle. Here we show that SMN loss in mouse skeletal muscle leads to accumulation of dysfunctional mitochondria. Expression profiling of single myofibers from a muscle specific Smn1 knockout mouse model revealed down-regulation of mitochondrial and lysosomal genes. Albeit levels of proteins that mark mitochondria for mitophagy were increased, morphologically deranged mitochondria with impaired complex I and IV activity and respiration and that produced excess reactive oxygen species accumulated in Smn1 knockout muscles, because of the lysosomal dysfunction highlighted by the transcriptional profiling. Amniotic fluid stem cells transplantation that corrects the SMN knockout mouse myopathic phenotype restored mitochondrial morphology and expression of mitochondrial genes. Thus, targeting muscle mitochondrial dysfunction in SMA may complement the current gene therapy.


Subject(s)
Muscle, Skeletal , Muscular Atrophy, Spinal , Animals , Mice , Muscular Atrophy, Spinal/genetics , Motor Neurons , Mice, Knockout , Mitochondria/genetics
7.
Nat Metab ; 3(12): 1633-1647, 2021 12.
Article in English | MEDLINE | ID: mdl-34873337

ABSTRACT

White to brown/beige adipocytes conversion is a possible therapeutic strategy to tackle the current obesity epidemics. While mitochondria are key for energy dissipation in brown fat, it is unknown if they can drive adipocyte browning. Here, we show that the mitochondrial cristae biogenesis protein optic atrophy 1 (Opa1) facilitates cell-autonomous adipocyte browning. In two cohorts of patients with obesity, including weight discordant monozygotic twin pairs, adipose tissue OPA1 levels are reduced. In the mouse, Opa1 overexpression favours white adipose tissue expandability as well as browning, ultimately improving glucose tolerance and insulin sensitivity. Transcriptomics and metabolomics analyses identify the Jumanji family chromatin remodelling protein Kdm3a and urea cycle metabolites, including fumarate, as effectors of Opa1-dependent browning. Mechanistically, the higher cyclic adenosine monophosphate (cAMP) levels in Opa1 pre-adipocytes activate cAMP-responsive element binding protein (CREB), which transcribes urea cycle enzymes. Flux analyses in pre-adipocytes indicate that Opa1-dependent fumarate accumulation depends on the urea cycle. Conversely, adipocyte-specific Opa1 deletion curtails urea cycle and beige differentiation of pre-adipocytes, and is rescued by fumarate supplementation. Thus, the urea cycle links the mitochondrial dynamics protein Opa1 to white adipocyte browning.


Subject(s)
Adipocytes, Brown/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Metabolic Networks and Pathways , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Urea/metabolism , Adipocytes, Beige/metabolism , Adipocytes, White/metabolism , Adipose Tissue/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Diet, High-Fat , Gene Expression Regulation , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Obesity/genetics , Obesity/metabolism , Thermogenesis , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
8.
J Cell Biol ; 220(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33566069

ABSTRACT

The commitment of mesenchymal stem cells to preadipocytes is stimulated by hormonal induction. Preadipocytes induced to differentiate repress protein synthesis, remodel their cytoskeleton, and increase mitochondrial function to support anabolic pathways. These changes enable differentiation into mature adipocytes. Our understanding of the factors that coordinately regulate the early events of adipocyte differentiation remains incomplete. Here, by using multipronged approaches, we have identified zinc finger CCCH-type containing 10 (Zc3h10) as a critical regulator of the early stages of adipogenesis. Zc3h10 depletion in preadipocytes resulted in increased protein translation and impaired filamentous (F)-actin remodeling, with the latter detrimental effect leading to mitochondrial and metabolic dysfunction. These defects negatively affected differentiation to mature adipocytes. In contrast, Zc3h10 overexpression yielded mature adipocytes with remarkably increased lipid droplet size. Overall, our study establishes Zc3h10 as a fundamental proadipogenic transcription factor that represses protein synthesis and promotes F-actin/mitochondria dynamics to ensure proper energy metabolism and favor lipid accumulation.


Subject(s)
Actins/metabolism , Adipogenesis , Mitochondria/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue, White/metabolism , Animals , Cell Line , Citric Acid Cycle , Cyclic AMP-Dependent Protein Kinases/metabolism , Energy Metabolism/genetics , Gene Expression Regulation , Lipid Metabolism/genetics , Male , Metabolome , Mice, Inbred C57BL , Mitochondrial Dynamics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Transcriptome/genetics , rho GTP-Binding Proteins/metabolism
9.
Cell Metab ; 31(5): 987-1003.e8, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32315597

ABSTRACT

While endothelial cell (EC) function is influenced by mitochondrial metabolism, the role of mitochondrial dynamics in angiogenesis, the formation of new blood vessels from existing vasculature, is unknown. Here we show that the inner mitochondrial membrane mitochondrial fusion protein optic atrophy 1 (OPA1) is required for angiogenesis. In response to angiogenic stimuli, OPA1 levels rapidly increase to limit nuclear factor kappa-light-chain-enhancer of activated B cell (NFκB) signaling, ultimately allowing angiogenic genes expression and angiogenesis. Endothelial Opa1 is indeed required in an NFκB-dependent pathway essential for developmental and tumor angiogenesis, impacting tumor growth and metastatization. A first-in-class small molecule-specific OPA1 inhibitor confirms that EC Opa1 can be pharmacologically targeted to curtail tumor growth. Our data identify Opa1 as a crucial component of physiological and tumor angiogenesis.


Subject(s)
GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Signal Transduction , Zebrafish
10.
Cell Rep ; 26(13): 3784-3797.e8, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30917329

ABSTRACT

Skeletal muscle is composed of different myofiber types that preferentially use glucose or lipids for ATP production. How fuel preference is regulated in these post-mitotic cells is largely unknown, making this issue a key question in the fields of muscle and whole-body metabolism. Here, we show that microRNAs (miRNAs) play a role in defining myofiber metabolic profiles. mRNA and miRNA signatures of all myofiber types obtained at the single-cell level unveiled fiber-specific regulatory networks and identified two master miRNAs that coordinately control myofiber fuel preference and mitochondrial morphology. Our work provides a complete and integrated mouse myofiber type-specific catalog of gene and miRNA expression and establishes miR-27a-3p and miR-142-3p as regulators of lipid use in skeletal muscle.


Subject(s)
MicroRNAs/genetics , Muscle Fibers, Skeletal/metabolism , Transcriptome , Animals , Cell Line , Cells, Cultured , Gene Regulatory Networks , Glycogen/metabolism , Glycolysis , Humans , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Oxidative Phosphorylation
11.
Front Mol Neurosci ; 11: 351, 2018.
Article in English | MEDLINE | ID: mdl-30319356

ABSTRACT

Epilepsies are a group of common neurological diseases exerting a strong burden on patients and society, often lacking clear etiology and effective therapeutical strategies. Early intervention during the development of epilepsy (epileptogenesis) is of great medical interest, though hampered by poorly characterized epileptogenetic processes. Using the intrahippocampal kainic acid mouse model of temporal lobe epilepsy, we investigated the functional role of the endogenous opioid enkephalin during epileptogenesis. We addressed three sequential questions: (1) How does enkephalin affect seizure threshold and how is it regulated during epileptogenesis? (2) Does enkephalin influence detrimental effects during epileptogenesis? (3) How is enkephalin linked to mitochondrial function during epileptogenesis?. In contrast to other neuropeptides, the expression of enkephalin is not regulated in a seizure dependent manner. The pattern of regulation, and enkephalin's proconvulsive effects suggested it as a potential driving force in epileptogenesis. Surprisingly, enkephalin deficiency aggravated progressive granule cell dispersion in kainic acid induced epileptogenesis. Based on reported beneficial effects of enkephalin on mitochondrial function in hypoxic/ischemic states, we hypothesized that enkephalin may be involved in the adaptation of mitochondrial respiration during epileptogenesis. Using high-resolution respirometry, we observed dynamic improvement of hippocampal mitochondrial respiration after kainic acid-injections in wild-type, but not in enkephalin-deficient mice. Thus, wild-type mice displayed higher efficiency in the use of mitochondrial capacity as compared to enkephalin-deficient mice. Our data demonstrate a Janus-headed role of enkephalin in epileptogenesis. In naive mice, enkephalin facilitates seizures, but in subsequent stages it contributes to neuronal survival through improved mitochondrial respiration.

12.
Cell Metab ; 27(4): 886-897.e4, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617646

ABSTRACT

How intracellular pathogens acquire essential non-diffusible host metabolites and whether the host cell counteracts the siphoning of these nutrients by its invaders are open questions. Here we show that host mitochondria fuse during infection by the intracellular parasite Toxoplasma gondii to limit its uptake of fatty acids (FAs). A combination of genetics and imaging of FA trafficking indicates that Toxoplasma infection triggers lipophagy, the autophagy of host lipid droplets (LDs), to secure cellular FAs essential for its proliferation. Indeed, Toxoplasma FA siphoning and growth are reduced in host cells genetically deficient for autophagy or triglyceride depots. Conversely, Toxoplasma FA uptake and proliferation are increased in host cells lacking mitochondrial fusion, required for efficient mitochondrial FA oxidation, or where mitochondrial FA oxidation is pharmacologically inhibited. Thus, mitochondrial fusion can be regarded as a cellular defense mechanism against intracellular parasites, by limiting Toxoplasma access to host nutrients liberated by lipophagy.


Subject(s)
Fatty Acids/metabolism , Lipid Droplets/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Toxoplasma/growth & development , Toxoplasma/metabolism , Toxoplasmosis/parasitology , Animals , Autophagy , Caco-2 Cells , Fibroblasts , Humans , Mice , Toxoplasmosis/immunology
13.
Cell Physiol Biochem ; 42(1): 169-184, 2017.
Article in English | MEDLINE | ID: mdl-28531892

ABSTRACT

BACKGROUND: Ankrd2 is a stress responsive protein mainly expressed in muscle cells. Upon the application of oxidative stress, Ankrd2 translocates into the nucleus where it regulates the activity of genes involved in cellular response to stress. Emery-Dreifuss Muscular Dystrophy 2 (EDMD2) is a muscular disorder caused by mutations of the gene encoding lamin A, LMNA. As well as many phenotypic abnormalities, EDMD2 muscle cells also feature a permanent basal stress state, the underlying molecular mechanisms of which are currently unclear. METHODS: Experiments were performed in EDMD2-lamin A overexpressing cell lines and EDMD2-affected human myotubes. Oxidative stress was produced by H2O2 treatment. Co-immunoprecipitation, cellular subfractionation and immunofluorescence analysis were used to validate the relation between Ankrd2 and forms of lamin A; cellular sensibility to stress was monitored by the analysis of Reactive Oxygen Species (ROS) release and cell viability. RESULTS: Our data demonstrate that oxidative stress induces the formation of a complex between Ankrd2 and lamin A. However, EDMD2-lamin A mutants were able to bind and mislocalize Ankrd2 in the nucleus even under basal conditions. Nonetheless, cells co-expressing Ankrd2 and EDMD2-lamin A mutants were more sensitive to oxidative stress than the Ankrd2-wild type lamin A counterpart. CONCLUSIONS: For the first time, we present evidence that in muscle fibers from patients affected by EDMD2, Ankrd2 has an unusual nuclear localization. By introducing a plausible mechanism ruling this accumulation, our data hint at a novel function of Ankrd2 in the pathogenesis of EDMD2-affected cells.


Subject(s)
Cell Nucleus/metabolism , Lamin Type A/metabolism , Muscle Proteins/metabolism , Muscular Dystrophy, Emery-Dreifuss/pathology , Nuclear Proteins/metabolism , Oxidative Stress , Repressor Proteins/metabolism , Cell Survival/drug effects , Cells, Cultured , HEK293 Cells , Humans , Hydrogen Peroxide/toxicity , Immunoprecipitation , Lamin Type A/chemistry , Lamin Type A/genetics , Microscopy, Fluorescence , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Oxidative Stress/drug effects , Plasmids/genetics , Plasmids/metabolism , Protein Binding , Protein Prenylation/drug effects , Reactive Oxygen Species/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics
14.
PLoS One ; 6(2): e16807, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21364935

ABSTRACT

BACKGROUND: Skeletal muscle is a complex, versatile tissue composed of a variety of functionally diverse fiber types. Although the biochemical, structural and functional properties of myofibers have been the subject of intense investigation for the last decades, understanding molecular processes regulating fiber type diversity is still complicated by the heterogeneity of cell types present in the whole muscle organ. METHODOLOGY/PRINCIPAL FINDINGS: We have produced a first catalogue of genes expressed in mouse slow-oxidative (type 1) and fast-glycolytic (type 2B) fibers through transcriptome analysis at the single fiber level (microgenomics). Individual fibers were obtained from murine soleus and EDL muscles and initially classified by myosin heavy chain isoform content. Gene expression profiling on high density DNA oligonucleotide microarrays showed that both qualitative and quantitative improvements were achieved, compared to results with standard muscle homogenate. First, myofiber profiles were virtually free from non-muscle transcriptional activity. Second, thousands of muscle-specific genes were identified, leading to a better definition of gene signatures in the two fiber types as well as the detection of metabolic and signaling pathways that are differentially activated in specific fiber types. Several regulatory proteins showed preferential expression in slow myofibers. Discriminant analysis revealed novel genes that could be useful for fiber type functional classification. CONCLUSIONS/SIGNIFICANCE: As gene expression analyses at the single fiber level significantly increased the resolution power, this innovative approach would allow a better understanding of the adaptive transcriptomic transitions occurring in myofibers under physiological and pathological conditions.


Subject(s)
Gene Expression Profiling , Genomics/methods , Microchemistry/methods , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/metabolism , Animals , Cluster Analysis , Feasibility Studies , Gene Expression Profiling/methods , Male , Mice , Miniaturization/methods , Muscle Fibers, Fast-Twitch/chemistry , Muscle Fibers, Slow-Twitch/chemistry , Muscle, Skeletal/chemistry , Oligonucleotide Array Sequence Analysis/methods , Protein Isoforms/genetics , Protein Isoforms/metabolism
15.
Biochim Biophys Acta ; 1783(6): 1023-35, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18302940

ABSTRACT

Ankrd2 is a member of the Muscle Ankyrin Repeat Protein family (MARPs), consisting of sarcomere-associated proteins that can also localize in the nucleus. There are indications that MARPs might function as shuttle proteins between the cytoplasm and nucleus, likely sending information to the nucleus concerning the changes in the structure or function of the contractile machinery. Even though recent findings suggest that the MARP gene family is not essential for the basal functioning of skeletal muscle, its influence on the gene expression program of skeletal muscle cells was highlighted. To investigate this regulatory role we produced and examined both morphological and functional features of myocytes stable overexpressing or silencing the Ankrd2 protein. The transcriptional profiles of the myocytes revealed that the molecular pathways perturbed by changes in Ankrd2 protein level are congruent with the morpho-physiological and biochemical data obtained in Ankrd2-modified myoblasts induced to differentiate. Our results suggest that Ankrd2 gives an important contribution to the coordination of proliferation and apoptosis during myogenic differentiation in vitro, mainly through the p53 network.


Subject(s)
Cell Cycle/physiology , Muscle Proteins/physiology , Muscle, Skeletal/cytology , Myoblasts/metabolism , Animals , Apoptosis/physiology , Caspase 3/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Flow Cytometry , Gene Expression/physiology , Gene Expression Profiling , Mice , Muscle Cells/cytology , Muscle Cells/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Transfection
16.
Physiol Genomics ; 25(1): 60-74, 2006 Mar 13.
Article in English | MEDLINE | ID: mdl-16380408

ABSTRACT

Denervation deeply affects muscle structure and function, the alterations being different in slow and fast muscles. Because the effects of denervation on fast muscles are still controversial, and high-throughput studies on gene expression in denervated muscles are lacking, we studied gene expression during atrophy progression following denervation in mouse tibialis anterior (TA). The sciatic nerve was cut close to trochanter in adult CD1 mice. One, three, seven, and fourteen days after denervation, animals were killed and TA muscles were dissected out and utilized for physiological experiments and gene expression studies. Target cDNAs from TA muscles were hybridized on a dedicated cDNA microarray of muscle genes. Seventy-one genes were found differentially expressed. Microarray results were validated, and the expression of relevant genes not probed on our array was monitored by real-time quantitative PCR (RQ-PCR). Nuclear- and mitochondrial-encoded genes implicated in energy metabolism were consistently downregulated. Among genes implicated in muscle contraction (myofibrillar and sarcoplasmic reticulum), genes typical of fast fibers were downregulated, whereas those typical of slow fibers were upregulated. Electrophoresis and Western blot showed less pronounced changes in myofibrillar protein expression, partially confirming changes in gene expression. Isometric tension of skinned fibers was little affected by denervation, whereas calcium sensitivity decreased. Functional studies in mouse extensor digitorum longus muscle showed prolongation in twitch time parameters and shift to the left in force-frequency curves after denervation. We conclude that, if studied at the mRNA level, fast muscles appear not less responsive than slow muscles to the interruption of neural stimulation.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Muscle Fibers, Fast-Twitch/metabolism , Muscle, Skeletal/metabolism , Animals , Mice , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle Denervation , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/innervation , Muscular Atrophy/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reproducibility of Results , Time Factors
17.
J Mol Biol ; 349(2): 349-66, 2005 Jun 03.
Article in English | MEDLINE | ID: mdl-15890200

ABSTRACT

Skeletal muscle development requires the coordinated expression of numerous transcription factors to control the specification of the muscle fate in mesodermal cells and the differentiation of the committed myoblasts into functional contractile fibers. The bHLH transcription factor MyoD plays a key role in these processes, since its forced expression is sufficient to induce the myogenesis in a variety of non-muscle cells in culture. Consistent with this observation, the majority of skeletal muscle genes require MyoD to activate their own transcription. In order to identify novel MyoD-target genes we generated C2C12 MyoD-silenced clones, and used a muscle-specific cDNA microarray to study the induced modifications of the transcriptional profile. Gene expression was analyzed at three different stages in differentiating MyoD(-)C2C12 myoblasts. These microarray data sets identified many additional uncharacterized downstream MyoD transcripts that may play important functions in muscle cell differentiation. Among these genes, we concentrated our study on the cell cycle regulators Cdkn1c and calcyclin and on the muscle-specific putative myogenic regulator Ankrd2. Bioinformatic and functional studies on the promoters of these genes clarified their dependence on MyoD activity. Clues of other regulatory mechanisms that might interact with the principal bHLH transcription factor have been revealed by the unexpected up-regulation in MyoD(-) cells of these novel (and other) target transcripts, at the differentiation stage in which MyoD became normally down-regulated.


Subject(s)
Cell Cycle Proteins/genetics , Gene Expression Regulation , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Proteins/genetics , MyoD Protein/metabolism , Nuclear Proteins/genetics , S100 Proteins/genetics , Animals , Base Sequence , Cell Differentiation , Cell Line , Cyclin-Dependent Kinase Inhibitor p57 , Down-Regulation , Gene Silencing , Genes, Reporter/genetics , Mice , Molecular Sequence Data , MyoD Protein/genetics , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , S100 Calcium Binding Protein A6 , Transcription, Genetic/genetics
18.
J Mol Biol ; 326(1): 137-49, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12547197

ABSTRACT

We have characterized a novel unconventional myosin heavy chain, named MYO18B, that appears to be expressed mainly in human cardiac and skeletal muscles and, at lower levels, in testis. MYO18B transcript is detected in all types of striated muscles but at much lower levels compared to class II sarcomeric myosins, and it is up regulated after in vitro differentiation of myoblasts into myotubes. Phylogenetic analysis shows that this myosin belongs to the recently identified class XVIII, however, unlike the other member of this class, it seems to be unique to Vertebrate since it contains two large amino acid domains of unknown function at the N and C-termini. Immunolocalization of MYO18B protein in skeletal muscle cells shows that this myosin heavy chain is located in the cytoplasm of undifferentiated myoblasts. After in vitro differentiation into myotubes, a fraction of this protein is accumulated in a subset of myonuclei. This nuclear localization was confirmed by immunofluorescence experiments on primary cardiomyocytes and adult muscle sections. In the cytoplasm MYO18B shows a punctate staining, both in cardiac and skeletal fibers. In some cases, cardiomyocytes show a partial sarcomeric pattern of MYO18B alternating that of alpha-actinin-2. In skeletal muscle the cytoplasmic MYO18B results much more evident in the fast type fibers.


Subject(s)
Cell Differentiation , Cell Nucleus/metabolism , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myosin Heavy Chains/metabolism , Animals , Cells, Cultured , Cytoplasm/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , In Vitro Techniques , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/classification , Myosin Heavy Chains/genetics , Phylogeny , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...