Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
2.
Trends Mol Med ; 30(3): 278-294, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38408879

ABSTRACT

Earlier evidence that targeting the balance between histone acetyltransferases (HATs) and deacetylases (HDACs), through exposure to HDAC inhibitors (HDACis), could enhance skeletal myogenesis, prompted interest in using HDACis to promote muscle regeneration. Further identification of constitutive HDAC activation in dystrophin-deficient muscles, caused by dysregulated nitric oxide (NO) signaling, provided the rationale for HDACi-based therapeutic interventions for Duchenne muscular dystrophy (DMD). In this review, we describe the molecular, preclinical, and clinical evidence supporting the efficacy of HDACis in countering disease progression by targeting pathogenic networks of gene expression in multiple muscle-resident cell types of patients with DMD. Given that givinostat is paving the way for HDACi-based interventions in DMD, next-generation HDACis with optimized therapeutic profiles and efficacy could be also explored for synergistic combinations with other therapeutic strategies.


Subject(s)
Muscular Dystrophy, Duchenne , Mice , Animals , Humans , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Muscle, Skeletal/metabolism , Mice, Inbred mdx , Dystrophin/metabolism , Signal Transduction
3.
Nat Commun ; 14(1): 1898, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37019933

ABSTRACT

N6-Methyladenosine (m6A) is well-known for controlling different processes of linear RNA metabolism. Conversely, its role in the biogenesis and function of circular RNAs (circRNAs) is still poorly understood. Here, we characterize circRNA expression in the pathological context of rhabdomyosarcoma (RMS), observing a global increase when compared to wild-type myoblasts. For a set of circRNAs, such an increase is due to the raised expression of the m6A machinery, which we also find to control the proliferation activity of RMS cells. Furthermore, we identify the RNA helicase DDX5 as a mediator of the back-splicing reaction and as a co-factor of the m6A regulatory network. DDX5 and the m6A reader YTHDC1 are shown to interact and to promote the production of a common subset of circRNAs in RMS. In line with the observation that YTHDC1/DDX5 depletion reduces RMS proliferation, our results provide proteins and RNA candidates for the study of rhabdomyosarcoma tumorigenicity.


Subject(s)
RNA, Circular , Rhabdomyosarcoma , Humans , RNA, Circular/metabolism , RNA/metabolism , RNA Splicing , RNA Splicing Factors/metabolism , Nerve Tissue Proteins/metabolism , DEAD-box RNA Helicases/metabolism
4.
Cell Rep ; 40(9): 111267, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36044855

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma of childhood characterized by the inability to exit the proliferative myoblast-like stage. The alveolar fusion positive subtype (FP-RMS) is the most aggressive and is mainly caused by the expression of PAX3/7-FOXO1 oncoproteins, which are challenging pharmacological targets. Here, we show that the DEAD box RNA helicase 5 (DDX5) is overexpressed in alveolar RMS cells and that its depletion and pharmacological inhibition decrease FP-RMS viability and slow tumor growth in xenograft models. Mechanistically, we provide evidence that DDX5 functions upstream of the EHMT2/AKT survival signaling pathway, by directly interacting with EHMT2 mRNA, modulating its stability and consequent protein expression. We show that EHMT2 in turns regulates PAX3-FOXO1 activity in a methylation-dependent manner, thus sustaining FP-RMS myoblastic state. Together, our findings identify another survival-promoting loop in FP-RMS and highlight DDX5 as a potential therapeutic target to arrest RMS growth.


Subject(s)
DEAD-box RNA Helicases , Rhabdomyosarcoma, Alveolar , Rhabdomyosarcoma, Embryonal , Rhabdomyosarcoma , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens , Histone-Lysine N-Methyltransferase/metabolism , Humans , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/genetics , RNA Helicases/metabolism , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/metabolism , Rhabdomyosarcoma, Alveolar/pathology
5.
Bioorg Med Chem Lett ; 72: 128858, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35718104

ABSTRACT

A new series of in vitro potent and highly selective histone methyl transferase enzyme G9a inhibitors was obtained. In particular, compound 2a, one the most potent G9a inhibitor identified, was endowed with >130-fold selectivity over GLP and excellent ligand efficiency. Therefore, it may represent a valuable tool compound to validate the role of highly selective G9a inhibitors in different pathological conditions. When 2a was characterized in vitro in cellular models of skeletal muscle differentiation, a relevant increase of myofibers' size and reduction of the fibroadipogenic infiltration were observed, further confirming the therapeutic potential of selective G9a inhibitors for the treatment of Duchenne muscle dystrophy.


Subject(s)
Histone-Lysine N-Methyltransferase , Histones , Enzyme Inhibitors/pharmacology
6.
Life (Basel) ; 12(4)2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35455028

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked disease, caused by a mutant dystrophin gene, leading to muscle membrane instability, followed by muscle inflammation, infiltration of pro-inflammatory macrophages and fibrosis. The calcium-activated potassium channel type 3.1 (KCa3.1) plays key roles in controlling both macrophage phenotype and fibroblast proliferation, two critical contributors to muscle damage. In this work, we demonstrate that pharmacological blockade of the channel in the mdx mouse model during the early degenerative phase favors the acquisition of an anti-inflammatory phenotype by tissue macrophages and reduces collagen deposition in muscles, with a concomitant reduction of muscle damage. As already observed with other treatments, no improvement in muscle performance was observed in vivo. In conclusion, this work supports the idea that KCa3.1 channels play a contributing role in controlling damage-causing cells in DMD. A more complete understanding of their function could lead to the identification of novel therapeutic approaches.

7.
Neurobiol Dis ; 168: 105718, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35390481

ABSTRACT

Dystrophinopaties, e.g., Duchenne muscular dystrophy (DMD), Becker muscular dystrophy and X-linked dilated cardiomyopathy are inherited neuromuscular diseases, characterized by progressive muscular degeneration, which however associate with a significant impact on general system physiology. The more severe is the pathology and its diversified manifestations, the heavier are its effects on organs, systems, and tissues other than muscles (skeletal, cardiac and smooth muscles). All dystrophinopaties are characterized by mutations in a single gene located on the X chromosome encoding dystrophin (Dp427) and its shorter isoforms, but DMD is the most devasting: muscular degenerations manifests within the first 4 years of life, progressively affecting motility and other muscular functions, and leads to a fatal outcome between the 20s and 40s. To date, after years of studies on both DMD patients and animal models of the disease, it has been clearly demonstrated that a significant percentage of DMD patients are also afflicted by cognitive, neurological, and autonomic disorders, of varying degree of severity. The anatomical correlates underlying neural functional damages are established during embryonic development and the early stages of postnatal life, when brain circuits, sensory and motor connections are still maturing. The impact of the absence of Dp427 on the development, differentiation, and consolidation of specific cerebral circuits (hippocampus, cerebellum, prefrontal cortex, amygdala) is significant, and amplified by the frequent lack of one or more of its lower molecular mass isoforms. The most relevant aspect, which characterizes DMD-associated neurological disorders, is based on morpho-functional alterations of selective synaptic connections within the affected brain areas. This pathological feature correlates neurological conditions of DMD to other severe neurological disorders, such as schizophrenia, epilepsy and autistic spectrum disorders, among others. This review discusses the organization and the role of the dystrophin-dystroglycan complex in muscles and neurons, focusing on the neurological aspect of DMD and on the most relevant morphological and functional synaptic alterations, in both central and autonomic nervous systems, described in the pathology and its animal models.


Subject(s)
Cardiomyopathy, Dilated , Muscular Dystrophy, Duchenne , Animals , Dystrophin/genetics , Humans , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Neurons/pathology , Protein Isoforms
8.
Trends Genet ; 38(5): 501-513, 2022 05.
Article in English | MEDLINE | ID: mdl-35078651

ABSTRACT

Muscle stem cells (MuSCs) are responsible for skeletal muscle homeostasis and repair. In response to extracellular cues, MuSCs activate from quiescence, expand, differentiate into mature myofibers, and self-renew within their regenerative niche. These steps are accomplished by the dynamic action of different chromatin-modifying enzymes that, cooperating with myogenic transcription factors, coordinately regulate defined transcriptional programs. Here, we review the current knowledge on the epigenetic dynamics that allow MuSCs' fate decisions. We describe the emerging mechanisms showing how chromatin topology impacts the 3D genome architecture of MuSCs during myogenesis. Because these processes contribute to shape and maintain cell identity, we highlight how defects in proper epigenetic control of MuSCs' fate decisions underlie the pathogenesis of muscle diseases, causing the acquisition of derailed cell fates and the incapacity to properly self-renew.


Subject(s)
Muscle, Skeletal , Stem Cells , Cell Differentiation/genetics , Chromatin/genetics , Epigenesis, Genetic , Muscle, Skeletal/physiology
9.
Bio Protoc ; 11(21): e4209, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34859124

ABSTRACT

LncRNAs have been recently implicated in the epigenetic control of muscle differentiation and their functional characterization has traditionally relied upon in vitro models of myogenic differentiation. However, the use of experimental paradigms to specifically target lncRNAs expression in muscle stem cells (MuSCs), also known as satellite cells, represents an important requisite to interrogate their function in more physiological contexts. Since isolation and culture of single myofibers preserves satellite cells within their physiological niche underneath the surrounding basal lamina, this procedure represents the optimal approach to follow satellite cell dynamics ex-vivo, such as activation from quiescence, expansion of committed progenitors, differentiation, and self-renewal. Here, we detail an optimized protocol to isolate viable single myofibers from the extensor digitorum longus (EDL) skeletal muscle of adult mice and to manipulate the expression of lncRNAs by antisense LNA GapmeRs-mediated knock-down (KD). Furthermore, we describe a method of EdU incorporation that, coupled to lncRNA KD and subsequent immunofluorescence analysis of proliferating, differentiating, and satellite cell-specific markers, permits the inference of lncRNAs function on muscle stem cells dynamics. Graphic abstract: Graphical representation of the single myofiber isolation method. Experimental workflow showing the main steps of the protocol procedure: EDL muscle harvesting from the mouse hindlimb; EDL digestion into single myofibers; transfection with antisense oligos and culture for 96h; immunofluorescence protocol and image outcome.

10.
Sci Adv ; 7(23)2021 06.
Article in English | MEDLINE | ID: mdl-34078594

ABSTRACT

H3K9 methylation maintains cell identity orchestrating stable silencing and anchoring of alternate fate genes within the heterochromatic compartment underneath the nuclear lamina (NL). However, how cell type-specific genomic regions are specifically targeted to the NL is still elusive. Using fibro-adipogenic progenitors (FAPs) as a model, we identified Prdm16 as a nuclear envelope protein that anchors H3K9-methylated chromatin in a cell-specific manner. We show that Prdm16 mediates FAP developmental capacities by orchestrating lamina-associated domain organization and heterochromatin sequestration at the nuclear periphery. We found that Prdm16 localizes at the NL where it cooperates with the H3K9 methyltransferases G9a/GLP to mediate tethering and silencing of myogenic genes, thus repressing an alternative myogenic fate in FAPs. Genetic and pharmacological disruption of this repressive pathway confers to FAP myogenic competence, preventing fibro-adipogenic degeneration of dystrophic muscles. In summary, we reveal a druggable mechanism of heterochromatin perinuclear sequestration exploitable to reprogram FAPs in vivo.

11.
Elife ; 102021 01 12.
Article in English | MEDLINE | ID: mdl-33432928

ABSTRACT

Skeletal muscle possesses an outstanding capacity to regenerate upon injury due to the adult muscle stem cell (MuSC) activity. This ability requires the proper balance between MuSC expansion and differentiation, which is critical for muscle homeostasis and contributes, if deregulated, to muscle diseases. Here, we functionally characterize a novel chromatin-associated long noncoding RNA (lncRNA), Lnc-Rewind, which is expressed in murine MuSCs and conserved in human. We find that, in mouse, Lnc-Rewind acts as an epigenetic regulator of MuSC proliferation and expansion by influencing the expression of skeletal muscle genes and several components of the WNT (Wingless-INT) signalling pathway. Among them, we identified the nearby Wnt7b gene as a direct Lnc-Rewind target. We show that Lnc-Rewind interacts with the G9a histone lysine methyltransferase and mediates the in cis repression of Wnt7b by H3K9me2 deposition. Overall, these findings provide novel insights into the epigenetic regulation of adult muscle stem cells fate by lncRNAs.


Subject(s)
Epigenesis, Genetic , Muscle Development/genetics , Muscle, Skeletal/metabolism , Myoblasts/metabolism , RNA, Long Noncoding/genetics , Animals , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/growth & development , RNA, Long Noncoding/metabolism
12.
J Vis Exp ; (161)2020 07 01.
Article in English | MEDLINE | ID: mdl-32716379

ABSTRACT

Autosomal dominant Emery-Dreifuss muscular dystrophy (EDMD) is caused by mutations in the LMNA gene, which encodes the A-type nuclear lamins, intermediate filament proteins that sustain the nuclear envelope and the components of the nucleoplasm. We recently reported that muscle wasting in EDMD can be ascribed to intrinsic epigenetic dysfunctions affecting muscle (satellite) stem cells regenerative capacity. Isolation and culture of single myofibers is one of the most physiological ex-vivo approaches to monitor satellite cells behavior within their niche, as they remain between the basal lamina surrounding the fiber and the sarcolemma. Therefore, it represents an invaluable experimental paradigm to study satellite cells from a variety of murine models. Here, we describe a re-adapted method to isolate intact and viable single myofibers from post-natal hindlimb muscles (Tibialis Anterior, Extensor Digitorum Longus, Gastrocnemius and Soleus). Following this protocol, we were able to study satellite cells from Lamin Δ8-11 -/- mice, a severe EDMD murine model, at only 19 days after birth. We detail the isolation procedure, as well as the culture conditions for obtaining a good amount of myofibers and their associated satellite-cells-derived progeny. When cultured in growth-factors rich medium, satellite cells derived from wild type mice activate, proliferate, and eventually differentiate or undergo self-renewal. In homozygous Lamin Δ8-11 -/- mutant mice these capabilities are severely impaired. This technique, if strictly followed, allows to study all processes linked to the myofiber-associated satellite cell even in early post-natal developmental stages and in fragile muscles.


Subject(s)
Embryonic Development/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/embryology , Muscular Dystrophy, Emery-Dreifuss/genetics , Myoblasts, Skeletal/metabolism , Animals , Disease Models, Animal , Humans , Mice
14.
J Clin Invest ; 130(5): 2408-2421, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31999646

ABSTRACT

Lamin A is a component of the inner nuclear membrane that, together with epigenetic factors, organizes the genome in higher order structures required for transcriptional control. Mutations in the lamin A/C gene cause several diseases belonging to the class of laminopathies, including muscular dystrophies. Nevertheless, molecular mechanisms involved in the pathogenesis of lamin A-dependent dystrophies are still largely unknown. The polycomb group (PcG) of proteins are epigenetic repressors and lamin A interactors, primarily involved in the maintenance of cell identity. Using a murine model of Emery-Dreifuss muscular dystrophy (EDMD), we show here that lamin A loss deregulated PcG positioning in muscle satellite stem cells, leading to derepression of non-muscle-specific genes and p16INK4a, a senescence driver encoded in the Cdkn2a locus. This aberrant transcriptional program caused impairment in self-renewal, loss of cell identity, and premature exhaustion of the quiescent satellite cell pool. Genetic ablation of the Cdkn2a locus restored muscle stem cell properties in lamin A/C-null dystrophic mice. Our findings establish a direct link between lamin A and PcG epigenetic silencing and indicate that lamin A-dependent muscular dystrophy can be ascribed to intrinsic epigenetic dysfunctions of muscle stem cells.


Subject(s)
Epigenesis, Genetic , Lamin Type A/biosynthesis , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Emery-Dreifuss/metabolism , Polycomb-Group Proteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Lamin Type A/genetics , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/pathology , Polycomb-Group Proteins/genetics , Repressor Proteins/genetics
15.
J Enzyme Inhib Med Chem ; 35(1): 129-137, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31694426

ABSTRACT

The 3-hydroxy-3-methylglutaryl-CoA reductase, a key enzyme of the mevalonate pathway for the synthesis of cholesterol in mammals (ergosterol in fungi), is inhibited by statins, a class of cholesterol lowering drugs. Indeed, statins are in a wide medical use, yet statins treatment could induce side effects as hepatotoxicity and myopathy in patients. We used Saccharomyces cerevisiae as a model to investigate the effects of statins on mitochondria. We demonstrate that statins are active in S.cerevisiae by lowering the ergosterol content in cells and interfering with the attachment of mitochondrial DNA to the inner mitochondrial membrane. Experiments on murine myoblasts confirmed these results in mammals. We propose that the instability of mitochondrial DNA is an early indirect target of statins.


Subject(s)
DNA, Mitochondrial/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Saccharomyces cerevisiae/chemistry , DNA, Mitochondrial/chemistry , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Mitochondrial Membranes/chemistry
16.
Front Physiol ; 10: 1074, 2019.
Article in English | MEDLINE | ID: mdl-31496956

ABSTRACT

Skeletal muscle is composed of a large and heterogeneous assortment of cell populations that interact with each other to maintain muscle homeostasis and orchestrate regeneration. Although satellite cells (SCs) - which are muscle stem cells - are the protagonists of functional muscle repair following damage, several other cells such as inflammatory, vascular, and mesenchymal cells coordinate muscle regeneration in a finely tuned process. Fibro-adipogenic progenitors (FAPs) are a muscle interstitial mesenchymal cell population, which supports SCs differentiation during tissue regeneration. During the first days following muscle injury FAPs undergo massive expansion, which is followed by their macrophage-mediated clearance and the re-establishment of their steady-state pool. It is during this critical time window that FAPs, together with the other cellular components of the muscle stem cell niche, establish a dynamic network of interactions that culminate in muscle repair. A number of different molecules have been recently identified as important mediators of this cross-talk, and its alteration has been associated with different muscle pathologies. In this review, we will focus on the soluble factors that regulate FAPs activity, highlighting their roles in orchestrating the inter-cellular interactions between FAPs and the other cell populations that participate in muscle regeneration.

17.
J Cell Biol ; 218(9): 2826-2828, 2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31427369

ABSTRACT

Lamins A and C are intermediate filaments that provide structural support to the nuclear envelope and regulate gene expression. In this issue, Bertero et al. (2019. J. Cell Biol. https://doi.org/10.1083/jcb.201902117) report that although lamin A/C haploinsufficient cardiomyocytes show disease-associated phenotypes, those changes cannot be explained by alterations in chromatin compartmentalization.


Subject(s)
Chromatin , Induced Pluripotent Stem Cells , Haploinsufficiency , Lamin Type A/genetics , Nuclear Envelope
18.
Methods Mol Biol ; 1480: 311-22, 2016.
Article in English | MEDLINE | ID: mdl-27659996

ABSTRACT

Polycomb group (PcG) proteins are key epigenetic factors responsible for the proper spatiotemporal repression of defined transcriptional programs along the process of cell differentiation, including myogenesis. The discovery of the pivotal role played by PcG factors during myogenic differentiation relied on the possibility to culture myogenic cells in vitro. We describe here the methods currently used to isolate muscle stem cells (MuSCs) both from single myofibers and from bulk muscles by fluorescence-activated cell sorting (FACS), highlighting experimental details and critical steps. Through these techniques MuSCs can be efficiently isolated and cultured in vitro to recapitulate the different phases of myogenesis: activation, expansion, differentiation, and self-renewal.


Subject(s)
Cell Separation/methods , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Stem Cells/cytology , Cell Differentiation/genetics , Cells, Cultured , Flow Cytometry/methods , Muscle Development/genetics
19.
J Cell Biol ; 211(3): 533-51, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26553927

ABSTRACT

Beyond its role in providing structure to the nuclear envelope, lamin A/C is involved in transcriptional regulation. However, its cross talk with epigenetic factors--and how this cross talk influences physiological processes--is still unexplored. Key epigenetic regulators of development and differentiation are the Polycomb group (PcG) of proteins, organized in the nucleus as microscopically visible foci. Here, we show that lamin A/C is evolutionarily required for correct PcG protein nuclear compartmentalization. Confocal microscopy supported by new algorithms for image analysis reveals that lamin A/C knock-down leads to PcG protein foci disassembly and PcG protein dispersion. This causes detachment from chromatin and defects in PcG protein-mediated higher-order structures, thereby leading to impaired PcG protein repressive functions. Using myogenic differentiation as a model, we found that reduced levels of lamin A/C at the onset of differentiation led to an anticipation of the myogenic program because of an alteration of PcG protein-mediated transcriptional repression. Collectively, our results indicate that lamin A/C can modulate transcription through the regulation of PcG protein epigenetic factors.


Subject(s)
Lamin Type A/metabolism , Polycomb-Group Proteins/metabolism , Transcription, Genetic/genetics , Animals , Cell Differentiation/genetics , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Drosophila , Epigenesis, Genetic/genetics , Humans , Lamin Type A/genetics , Mice , Mice, Inbred C57BL , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Polycomb-Group Proteins/genetics
20.
Nat Rev Mol Cell Biol ; 16(8): 499-513, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26204160

ABSTRACT

The methylation of histone Lys residues by Lys methyltransferases (KMTs) regulates chromatin organization and either activates or represses gene expression, depending on the residue that is targeted. KMTs are emerging as key components in several cellular processes, and their deregulation is often associated with pathogenesis. Here, we review the current knowledge on the main KMTs that are associated with gene silencing: namely, those responsible for methylating histone H3 Lys 9 (H3K9), H3K27 and H4K20. We discuss their biochemical properties and the various mechanisms by which they are targeted to the chromatin and regulate gene expression, as well as new data on the interplay between them and other chromatin modifiers.


Subject(s)
Gene Silencing , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Protein Processing, Post-Translational , Animals , Chromatin Assembly and Disassembly , Humans , Methylation
SELECTION OF CITATIONS
SEARCH DETAIL
...