Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38994563

ABSTRACT

SMCHD1 is an epigenetic regulatory protein known to modulate the targeted repression of large chromatin domains. Diminished SMCHD1 function in muscle fibers causes Facioscapulohumeral Muscular Dystrophy (FSHD2) through derepression of the D4Z4 chromatin domain, an event which permits the aberrant expression of the disease-causing gene DUX4. Given that SMCHD1 plays a broader role in establishing the cellular epigenome, we examined whether loss of SMCHD1 function might affect muscle homeostasis through additional mechanisms. Here we show that acute depletion of SMCHD1 results in a DUX4-independent defect in myoblast proliferation. Genomic and transcriptomic experiments determined that SMCHD1 associates with enhancers of genes controlling cell cycle to activate their expression. Amongst these cell cycle regulatory genes, we identified LAP2 as a key target of SMCHD1 required for the expansion of myoblasts, where the ectopic expression of LAP2 rescues the proliferation defect of SMCHD1-depleted cells. Thus, the epigenetic regulator SMCHD1 can play the role of a transcriptional co-activator for maintaining the expression of genes required for muscle progenitor expansion. This DUX4-independent role for SMCHD1 in myoblasts suggests that the pathology of FSHD2 may be a consequence of defective muscle regeneration in addition to the muscle wasting caused by spurious DUX4 expression.

2.
Int J Mol Sci ; 25(4)2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38396885

ABSTRACT

The mammalian myocardium grows rapidly during early development due to cardiomyocyte proliferation, which later transitions to cell hypertrophy to sustain the heart's postnatal growth. Although this cell transition in the postnatal heart is consistently preserved in mammalian biology, little is known about the regulatory mechanisms that link proliferation suppression with hypertrophy induction. We reasoned that the production of a micro-RNA(s) could serve as a key bridge to permit changes in gene expression that control the changed cell fate of postnatal cardiomyocytes. We used sequential expression analysis to identify miR205 as a micro-RNA that was uniquely expressed at the cessation of cardiomyocyte growth. Cardiomyocyte-specific miR205 deletion animals showed a 35% increase in heart mass by 3 months of age, with commensurate changes in cell cycle and Hippo pathway activity, confirming miR205's potential role in controlling cardiomyocyte proliferation. In contrast, overexpression of miR205 in newborn hearts had little effect on heart size or function, indicating a complex, probably redundant regulatory system. These findings highlight miR205's role in controlling the shift from cardiomyocyte proliferation to hypertrophic development in the postnatal period.


Subject(s)
Heart , MicroRNAs , Myocytes, Cardiac , Animals , Animals, Newborn , Cell Proliferation/genetics , Hypertrophy/metabolism , Mammals , Myocytes, Cardiac/metabolism , Mice
3.
Cells ; 12(18)2023 09 21.
Article in English | MEDLINE | ID: mdl-37759546

ABSTRACT

Heart disease remains a global leading cause of death and disability, necessitating a comprehensive understanding of the heart's development, repair, and dysfunction. This review surveys recent discoveries that explore the developmental transition of proliferative fetal cardiomyocytes into hypertrophic postnatal cardiomyocytes, a process yet to be well-defined. This transition is key to the heart's growth and has promising therapeutic potential, particularly for congenital or acquired heart damage, such as myocardial infarctions. Although significant progress has been made, much work is needed to unravel the complex interplay of signaling pathways that regulate cardiomyocyte proliferation and hypertrophy. This review provides a detailed perspective for future research directions aimed at the potential therapeutic harnessing of the perinatal heart transitions.


Subject(s)
Myocardial Infarction , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Signal Transduction , Myocardial Infarction/metabolism
4.
Microb Cell ; 10(8): 157-169, 2023 Aug 07.
Article in English | MEDLINE | ID: mdl-37545643

ABSTRACT

Caspase 3 activation is a hallmark of cell death and there is a strong correlation between elevated protease activity and evolving pathology in neurodegenerative disease, such as amyotrophic lateral sclerosis (ALS). At the cellular level, ALS is characterized by protein aggregates and inclusions, comprising the RNA binding protein TDP-43, which are hypothesized to trigger pathogenic activation of caspase 3. However, a growing body of evidence indicates this protease is essential for ensuring cell viability during growth, differentiation and adaptation to stress. Here, we explored whether caspase 3 acts to disperse toxic protein aggregates, a proteostasis activity first ascribed to the distantly related yeast metacaspase ScMCA1. We demonstrate that human caspase 3 can functionally substitute for the ScMCA1 and limit protein aggregation in yeast, including TDP-43 inclusions. Proteomic analysis revealed that disrupting caspase 3 in the same yeast substitution model resulted in detrimental TDP-43/mitochondrial protein associations. Similarly, suppression of caspase 3, in either murine or human skeletal muscle cells, led to accumulation of TDP-43 aggregates and impaired mitochondrial function. These results suggest that caspase 3 is not inherently pathogenic, but may act as a compensatory proteostasis factor, to limit TDP-43 protein inclusions and protect organelle function in aggregation related degenerative disease.

5.
Trends Cell Biol ; 33(10): 850-859, 2023 10.
Article in English | MEDLINE | ID: mdl-36997393

ABSTRACT

Self-inflicted DNA strand breaks are canonically linked with cell death pathways and the establishment of genetic diversity in immune and germline cells. Moreover, this form of DNA damage is an established source of genome instability in cancer development. However, recent studies indicate that nonlethal self-inflicted DNA strand breaks play an indispensable but underappreciated role in a variety of cell processes, including differentiation and cancer therapy responses. Mechanistically, these physiological DNA breaks originate from the activation of nucleases, which are best characterized for inducing DNA fragmentation in apoptotic cell death. In this review, we outline the emerging biology of one critical nuclease, caspase-activated DNase (CAD), and how directed activation or deployment of this enzyme can lead to divergent cell fate outcomes.


Subject(s)
Apoptosis , Neoplasms , Humans , DNA/metabolism , DNA Damage , Neoplasms/genetics , Cell Differentiation , DNA Breaks
6.
Science ; 377(6606): 666-669, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35926054

ABSTRACT

Muscle stem cells (MuSCs) reside in a specialized niche that ensures their regenerative capacity. Although we know that innate immune cells infiltrate the niche in response to injury, it remains unclear how MuSCs adapt to this altered environment for initiating repair. Here, we demonstrate that inflammatory cytokine signaling from the regenerative niche impairs the ability of quiescent MuSCs to reenter the cell cycle. The histone H3 lysine 27 (H3K27) demethylase JMJD3, but not UTX, allowed MuSCs to overcome inhibitory inflammation signaling by removing trimethylated H3K27 (H3K27me3) marks at the Has2 locus to initiate production of hyaluronic acid, which in turn established an extracellular matrix competent for integrating signals that direct MuSCs to exit quiescence. Thus, JMJD3-driven hyaluronic acid synthesis plays a proregenerative role that allows MuSC adaptation to inflammation and the initiation of muscle repair.


Subject(s)
Hyaluronic Acid , Inflammation , Jumonji Domain-Containing Histone Demethylases , Muscle, Skeletal , Myoblasts, Skeletal , Regeneration , Stem Cell Niche , Animals , Cell Cycle , Histones , Humans , Hyaluronic Acid/biosynthesis , Inflammation/metabolism , Interferon-gamma/metabolism , Interleukin-6 , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Myoblasts, Skeletal/metabolism
7.
Science ; 376(6592): 476-483, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35482866

ABSTRACT

Genotoxic therapy such as radiation serves as a frontline cancer treatment, yet acquired resistance that leads to tumor reoccurrence is frequent. We found that cancer cells maintain viability during irradiation by reversibly increasing genome-wide DNA breaks, thereby limiting premature mitotic progression. We identify caspase-activated DNase (CAD) as the nuclease inflicting these de novo DNA lesions at defined loci, which are in proximity to chromatin-modifying CCCTC-binding factor (CTCF) sites. CAD nuclease activity is governed through phosphorylation by DNA damage response kinases, independent of caspase activity. In turn, loss of CAD activity impairs cell fate decisions, rendering cancer cells vulnerable to radiation-induced DNA double-strand breaks. Our observations highlight a cancer-selective survival adaptation, whereby tumor cells deploy regulated DNA breaks to delimit the detrimental effects of therapy-evoked DNA damage.


Subject(s)
DNA Damage , Neoplasms , Chromatin , DNA/radiation effects , DNA Breaks, Double-Stranded , DNA Repair , Neoplasms/genetics
8.
Cells ; 11(6)2022 03 11.
Article in English | MEDLINE | ID: mdl-35326417

ABSTRACT

The induction of lineage-specific gene programs are strongly influenced by alterations in local chromatin architecture. However, key players that impact this genome reorganization remain largely unknown. Here, we report that the removal of the special AT-rich binding protein 2 (SATB2), a nuclear protein known to bind matrix attachment regions, is a key event in initiating myogenic differentiation. The deletion of myoblast SATB2 in vitro initiates chromatin remodeling and accelerates differentiation, which is dependent on the caspase 7-mediated cleavage of SATB2. A genome-wide analysis indicates that SATB2 binding within chromatin loops and near anchor points influences both loop and sub-TAD domain formation. Consequently, the chromatin changes that occur with the removal of SATB2 lead to the derepression of differentiation-inducing factors while also limiting the expression of genes that inhibit this cell fate change. Taken together, this study demonstrates that the temporal control of the SATB2 protein is critical in shaping the chromatin environment and coordinating the myogenic differentiation program.


Subject(s)
Matrix Attachment Region Binding Proteins , Caspases , Chromatin , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Myoblasts/metabolism , Transcription Factors/metabolism
9.
Brain ; 144(9): 2722-2731, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34581780

ABSTRACT

Striated muscle needs to maintain cellular homeostasis in adaptation to increases in physiological and metabolic demands. Failure to do so can result in rhabdomyolysis. The identification of novel genetic conditions associated with rhabdomyolysis helps to shed light on hitherto unrecognized homeostatic mechanisms. Here we report seven individuals in six families from different ethnic backgrounds with biallelic variants in MLIP, which encodes the muscular lamin A/C-interacting protein, MLIP. Patients presented with a consistent phenotype characterized by mild muscle weakness, exercise-induced muscle pain, variable susceptibility to episodes of rhabdomyolysis, and persistent basal elevated serum creatine kinase levels. The biallelic truncating variants were predicted to result in disruption of the nuclear localizing signal of MLIP. Additionally, reduced overall RNA expression levels of the predominant MLIP isoform were observed in patients' skeletal muscle. Collectively, our data increase the understanding of the genetic landscape of rhabdomyolysis to now include MLIP as a novel disease gene in humans and solidifies MLIP's role in normal and diseased skeletal muscle homeostasis.


Subject(s)
Co-Repressor Proteins/genetics , Creatine Kinase , Genetic Variation/genetics , Muscular Diseases/genetics , Myalgia/genetics , Nuclear Proteins/genetics , Rhabdomyolysis/genetics , Adolescent , Child , Child, Preschool , Creatine Kinase/blood , Female , Humans , Male , Muscular Diseases/blood , Muscular Diseases/diagnostic imaging , Myalgia/blood , Myalgia/diagnostic imaging , Rhabdomyolysis/blood , Rhabdomyolysis/diagnostic imaging , Young Adult
10.
Mol Cell ; 77(5): 927-929, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32142688
11.
J Am Heart Assoc ; 7(23): e010404, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30486716

ABSTRACT

Background Cardiac hypertrophy is an adaptive remodeling event that may improve or diminish contractile performance of the heart. Physiologic and pathologic hypertrophy yield distinct outcomes, yet both are dependent on caspase-directed proteolysis. This suggests that each form of myocardial growth may derive from a specific caspase cleavage event(s). We examined whether caspase 3 cleavage of the actin capping/severing protein gelsolin is essential for the development of pathologic hypertrophy. Methods and Results Caspase targeting of gelsolin was established through protein analysis of hypertrophic cardiomyocytes and mass spectrometry mapping of cleavage sites. Pathologic agonists induced late-stage caspase-mediated cleavage of gelsolin. The requirement of caspase-mediated gelsolin cleavage for hypertrophy induction was evaluated in primary cardiomyocytes by cell size analysis, monitoring of prohypertrophy markers, and measurement of hypertrophy-related transcription activity. The in vivo impact of caspase-mediated cleavage was investigated by echo-guided intramyocardial injection of adenoviral-expressed gelsolin. Expression of the N-terminal gelsolin caspase cleavage fragment was necessary and sufficient to cause pathologic remodeling in isolated cardiomyocytes and the intact heart, whereas expression of a noncleavable form prevents cardiac remodeling. Alterations in myocardium structure and function were determined by echocardiography and end-stage cardiomyocyte cell size analysis. Gelsolin secretion was also monitored for its impact on naïve cells using competitive antibody trapping, demonstrating that hypertrophic agonist stimulation of cardiomyocytes leads to gelsolin secretion, which induces hypertrophy in naïve cells. Conclusions These results suggest that cell autonomous caspase cleavage of gelsolin is essential for pathologic hypertrophy and that cardiomyocyte secretion of gelsolin may accelerate this negative remodeling response.


Subject(s)
Cardiomegaly/metabolism , Caspase 3/metabolism , Gelsolin/metabolism , Animals , Cardiomegaly/etiology , Fluorescent Antibody Technique , Gene Knockdown Techniques , Male , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley
12.
J Biol Chem ; 293(51): 19761-19770, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30389785

ABSTRACT

Muscle-enriched lamin-interacting protein (Mlip) is an alternatively spliced gene whose splicing specificity is dictated by tissue type. MLIP is most abundantly expressed in brain, cardiac, and skeletal muscle. In the present study, we systematically mapped the transcriptional start and stop sites of murine Mlip Rapid amplification of cDNA ends (RACE) of Mlip transcripts from the brain, heart, and skeletal muscle revealed two transcriptional start sites (TSSs), exon 1a and exon 1b, and only one transcriptional termination site. RT-PCR analysis of the usage of the two identified TSSs revealed that the heart utilizes only exon 1a for MLIP expression, whereas the brain exclusively uses exon 1b TSS. Loss of Mlip exon 1a in mice resulted in a 7-fold increase in the prevalence of centralized nuclei in muscle fibers with the Mlip exon1a-deficient satellite cells on single fibers exhibiting a significant delay in commitment to a MYOD-positive phenotype. Furthermore, we demonstrate that the A-type lamin-binding domain in MLIP is encoded in exon 1a, indicating that MLIP isoforms generated with exon 1b TSS lack the A-type lamin-binding domain. Collectively these findings suggest that Mlip tissue-specific expression and alternative splicing play a critical role in determining MLIP's functions in mice.


Subject(s)
Alternative Splicing/genetics , Carrier Proteins/genetics , Gene Expression Regulation/genetics , Nuclear Proteins/genetics , Transcription Initiation Site , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Line , Co-Repressor Proteins , Exons/genetics , Humans , Introns/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Organ Specificity , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism
13.
Can J Physiol Pharmacol ; 96(5): 535-539, 2018 May.
Article in English | MEDLINE | ID: mdl-29533724

ABSTRACT

The capacity to isolate and study single cardiomyocytes has dramatically enhanced our understanding of the fundamental mechanisms of the heart. Currently, 2 primary methods for the isolation of cardiomyocytes are employed: (i) the neonatal isolation protocol and (ii) the Langendorff isolation method. A major limiting feature of both procedures is the inability to isolate cardiomyocytes between 3 days and 3 weeks after birth. Herein, we report the establishment and validation of a new method for the rapid and efficient isolation of mouse cardiomyocytes, regardless of age. This novel procedure utilizes whole heart perfusion of a trypsin-collagenase Krebs-based buffer through the left ventricle at a high flow rate. Cardiomyocytes can be isolated in significantly less time with a simple, syringe-pump-based apparatus. Typically, we can digest 10-15 hearts per hour. Altogether, we have established an efficient and reproducible method for the rapid isolation of fresh cardiomyocytes from postnatal mouse hearts of any age.


Subject(s)
Cell Separation/methods , Fibroblasts/cytology , Myocytes, Cardiac/cytology , Animals , Mice , Time Factors
14.
Microb Cell ; 5(1): 4-31, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29354647

ABSTRACT

Elucidating the biology of yeast in its full complexity has major implications for science, medicine and industry. One of the most critical processes determining yeast life and physiology is cel-lular demise. However, the investigation of yeast cell death is a relatively young field, and a widely accepted set of concepts and terms is still missing. Here, we propose unified criteria for the defi-nition of accidental, regulated, and programmed forms of cell death in yeast based on a series of morphological and biochemical criteria. Specifically, we provide consensus guidelines on the differ-ential definition of terms including apoptosis, regulated necrosis, and autophagic cell death, as we refer to additional cell death rou-tines that are relevant for the biology of (at least some species of) yeast. As this area of investigation advances rapidly, changes and extensions to this set of recommendations will be implemented in the years to come. Nonetheless, we strongly encourage the au-thors, reviewers and editors of scientific articles to adopt these collective standards in order to establish an accurate framework for yeast cell death research and, ultimately, to accelerate the pro-gress of this vibrant field of research.

15.
Semin Cell Dev Biol ; 82: 96-104, 2018 10.
Article in English | MEDLINE | ID: mdl-29129746

ABSTRACT

Caspase signaling pathways were originally discovered as conveyors of programmed cell death, yet a compendium of research over the past two decades have demonstrated that these same conduits have a plethora of physiologic functions. Arguably the most extensive non-death activity that has been attributed to this protease clade is the capacity to induce cell differentiation. Caspase control of differentiation is conserved across diverse metazoan organisms from flies to humans, suggesting an ancient origin for this form of cell fate control. Here we discuss the mechanisms by which caspase enzymes manage differentiation, the targeted substrates that may be common across cell lineages, and the countervailing signals that may be essential for these proteases to 'execute' this non-death cell fate.


Subject(s)
Caspases/metabolism , Animals , Cell Differentiation , Humans
16.
Cell Res ; 27(10): 1195-1215, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28785017

ABSTRACT

The post-natal heart adapts to stress and overload through hypertrophic growth, a process that may be pathologic or beneficial (physiologic hypertrophy). Physiologic hypertrophy improves cardiac performance in both healthy and diseased individuals, yet the mechanisms that propagate this favorable adaptation remain poorly defined. We identify the cytokine cardiotrophin 1 (CT1) as a factor capable of recapitulating the key features of physiologic growth of the heart including transient and reversible hypertrophy of the myocardium, and stimulation of cardiomyocyte-derived angiogenic signals leading to increased vascularity. The capacity of CT1 to induce physiologic hypertrophy originates from a CK2-mediated restraining of caspase activation, preventing the transition to unrestrained pathologic growth. Exogenous CT1 protein delivery attenuated pathology and restored contractile function in a severe model of right heart failure, suggesting a novel treatment option for this intractable cardiac disease.


Subject(s)
Cytokines/genetics , Heart Failure/genetics , Heart/growth & development , Vascular Remodeling/genetics , Animals , Cytokines/administration & dosage , Heart/physiopathology , Heart Failure/physiopathology , Humans , Mice , Muscle Development/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Rats , Signal Transduction
17.
Cell Death Differ ; 24(8): 1359-1368, 2017 08.
Article in English | MEDLINE | ID: mdl-28338655

ABSTRACT

The phenotypic and biochemical similarities between caspase-mediated apoptosis and cellular differentiation are striking. They include such diverse phenomenon as mitochondrial membrane perturbations, cytoskeletal rearrangements and DNA fragmentation. The parallels between the two disparate processes suggest some common ancestry and highlight the paradoxical nature of the death-centric view of caspases. That is, what is the driving selective pressure that sustains death-inducing proteins throughout eukaryotic evolution? Plausibly, caspase function may be rooted in a primordial non-death function, such as cell differentiation, and was co-opted for its role in programmed cell death. This review will delve into the links between caspase-mediated apoptosis and cell differentiation and examine the distinguishing features of these events. More critically, we chronicle the evolutionary origins of caspases and propose that caspases may have held an ancient role in mediating the fidelity of cell division/differentiation through its effects on proteostasis and protein quality control.


Subject(s)
Apoptosis/genetics , Caspases/genetics , Cell Differentiation/genetics , Eukaryotic Cells/enzymology , Proteostasis/genetics , Animals , Biological Evolution , Caspases/metabolism , Cytoskeleton/enzymology , Cytoskeleton/ultrastructure , DNA Fragmentation , Eukaryotic Cells/cytology , Gene Expression Regulation , Humans , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/ultrastructure , Signal Transduction
18.
Cell Discov ; 2: 15041, 2016.
Article in English | MEDLINE | ID: mdl-27462438

ABSTRACT

Transient DNA strand break formation has been identified as an effective means to enhance gene expression in living cells. In the muscle lineage, cell differentiation is contingent upon the induction of caspase-mediated DNA strand breaks, which act to establish the terminal gene expression program. This coordinated DNA nicking is rapidly resolved, suggesting that myoblasts may deploy DNA repair machinery to stabilize the genome and entrench the differentiated phenotype. Here, we identify the base excision repair pathway component XRCC1 as an indispensable mediator of muscle differentiation. Caspase-triggered XRCC1 repair foci form rapidly within differentiating myonuclei, and then dissipate as the maturation program proceeds. Skeletal myoblast deletion of Xrcc1 does not have an impact on cell growth, yet leads to perinatal lethality, with sustained DNA damage and impaired myofiber development. Together, these results demonstrate that XRCC1 manages a temporally responsive DNA repair process to advance the muscle differentiation program.

19.
Skelet Muscle ; 6: 19, 2016.
Article in English | MEDLINE | ID: mdl-27148436

ABSTRACT

[This corrects the article DOI: 10.1186/s13395-016-0086-6.].

20.
Skelet Muscle ; 6: 16, 2016.
Article in English | MEDLINE | ID: mdl-27054028

ABSTRACT

Muscle atrophy derived from excessive proteolysis is a hallmark of numerous disease conditions. Accordingly, the negative consequences of skeletal muscle protein breakdown often overshadow the critical nature of proteolytic systems in maintaining normal cellular function. Here, we discuss the major cellular proteolysis machinery-the ubiquitin/proteosome system, the autophagy/lysosomal system, and caspase-mediated protein cleavage-and the critical role of these protein machines in establishing and preserving muscle health. We examine how ordered degradation modifies (1) the spatiotemporal expression of myogenic regulatory factors during myoblast differentiation, (2) membrane fusion during myotube formation, (3) sarcomere remodeling and muscle growth following physical stress, and (4) energy homeostasis during nutrient deprivation. Finally, we review the origin and etiology of a number of myopathies and how these devastating conditions arise from inborn errors in proteolysis.


Subject(s)
Muscle Development , Muscle, Skeletal/enzymology , Muscular Atrophy/enzymology , Peptide Hydrolases/metabolism , Proteolysis , Regeneration , Stress, Physiological , Adaptation, Physiological , Animals , Autophagy , Caspases/metabolism , Humans , Lysosomes/enzymology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...