Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965447

ABSTRACT

TFEB, a bHLH-leucine zipper transcription factor belonging to the MiT/TFE family, globally modulates cell metabolism by regulating autophagy and lysosomal functions. Remarkably, loss of TFEB in mice causes embryonic lethality due to severe defects in placentation associated with aberrant vascularization and resulting hypoxia. However, the molecular mechanism underlying this phenotype has remained elusive. By integrating in vivo analyses with multi-omics approaches and functional assays, we have uncovered an unprecedented function for TFEB in promoting the formation of a functional syncytiotrophoblast in the placenta. Our findings demonstrate that constitutive loss of TFEB in knock-out mice is associated with defective formation of the syncytiotrophoblast layer. Indeed, using in vitro models of syncytialization, we demonstrated that TFEB translocates into the nucleus during syncytiotrophoblast formation and binds to the promoters of crucial placental genes, including genes encoding fusogenic proteins (Syncytin-1 and Syncytin-2) and enzymes involved in steroidogenic pathways, such as CYP19A1, the rate-limiting enzyme for the synthesis of 17ß-Estradiol (E2). Conversely, TFEB depletion impairs both syncytial fusion and endocrine properties of syncytiotrophoblast, as demonstrated by a significant decrease in the secretion of placental hormones and E2 production. Notably, restoration of TFEB expression resets syncytiotrophoblast identity. Our findings identify that TFEB controls placental development and function by orchestrating both the transcriptional program underlying trophoblast fusion and the acquisition of endocrine function, which are crucial for the bioenergetic requirements of embryonic development.

2.
EMBO Mol Med ; 14(9): e15377, 2022 09 07.
Article in English | MEDLINE | ID: mdl-35929194

ABSTRACT

Lysosomes are cell organelles that degrade macromolecules to recycle their components. If lysosomal degradative function is impaired, e.g., due to mutations in lysosomal enzymes or membrane proteins, lysosomal storage diseases (LSDs) can develop. LSDs manifest often with neurodegenerative symptoms, typically starting in early childhood, and going along with a strongly reduced life expectancy and quality of life. We show here that small molecule activation of the Ca2+ -permeable endolysosomal two-pore channel 2 (TPC2) results in an amelioration of cellular phenotypes associated with LSDs such as cholesterol or lipofuscin accumulation, or the formation of abnormal vacuoles seen by electron microscopy. Rescue effects by TPC2 activation, which promotes lysosomal exocytosis and autophagy, were assessed in mucolipidosis type IV (MLIV), Niemann-Pick type C1, and Batten disease patient fibroblasts, and in neurons derived from newly generated isogenic human iPSC models for MLIV and Batten disease. For in vivo proof of concept, we tested TPC2 activation in the MLIV mouse model. In sum, our data suggest that TPC2 is a promising target for the treatment of different types of LSDs, both in vitro and in-vivo.


Subject(s)
Lysosomal Storage Diseases , Mucolipidoses , Neuronal Ceroid-Lipofuscinoses , Animals , Child, Preschool , Humans , Lysosomes/metabolism , Mice , Mucolipidoses/genetics , Mucolipidoses/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Quality of Life
3.
Traffic ; 23(5): 238-269, 2022 05.
Article in English | MEDLINE | ID: mdl-35343629

ABSTRACT

Since the discovery of lysosomes more than 70 years ago, much has been learned about the functions of these organelles. Lysosomes were regarded as exclusively degradative organelles, but more recent research has shown that they play essential roles in several other cellular functions, such as nutrient sensing, intracellular signalling and metabolism. Methodological advances played a key part in generating our current knowledge about the biology of this multifaceted organelle. In this review, we cover current methods used to analyze lysosome morphology, positioning, motility and function. We highlight the principles behind these methods, the methodological strategies and their advantages and limitations. To extract accurate information and avoid misinterpretations, we discuss the best strategies to identify lysosomes and assess their characteristics and functions. With this review, we aim to stimulate an increase in the quantity and quality of research on lysosomes and further ground-breaking discoveries on an organelle that continues to surprise and excite cell biologists.


Subject(s)
Lysosomes , Metabolic Networks and Pathways , Lysosomes/metabolism , Signal Transduction
4.
EMBO Mol Med ; 13(10): e13742, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34411438

ABSTRACT

Batten diseases (BDs) are a group of lysosomal storage disorders characterized by seizure, visual loss, and cognitive and motor deterioration. We discovered increased levels of globotriaosylceramide (Gb3) in cellular and murine models of CLN3 and CLN7 diseases and used fluorescent-conjugated bacterial toxins to label Gb3 to develop a cell-based high content imaging (HCI) screening assay for the repurposing of FDA-approved compounds able to reduce this accumulation within BD cells. We found that tamoxifen reduced the lysosomal accumulation of Gb3 in CLN3 and CLN7 cell models, including neuronal progenitor cells (NPCs) from CLN7 patient-derived induced pluripotent stem cells (iPSC). Here, tamoxifen exerts its action through a mechanism that involves activation of the transcription factor EB (TFEB), a master gene of lysosomal function and autophagy. In vivo administration of tamoxifen to the CLN7Δex2 mouse model reduced the accumulation of Gb3 and SCMAS, decreased neuroinflammation, and improved motor coordination. These data strongly suggest that tamoxifen may be a suitable drug to treat some types of Batten disease.


Subject(s)
Neuronal Ceroid-Lipofuscinoses , Animals , Drug Repositioning , Humans , Lysosomes , Membrane Glycoproteins/genetics , Mice , Molecular Chaperones/genetics , Neuronal Ceroid-Lipofuscinoses/drug therapy , Phenotype , Tamoxifen/pharmacology
5.
iScience ; 23(11): 101691, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33163944

ABSTRACT

The transcription factor EB (TFEB) has emerged as a master regulator of lysosomal biogenesis, exocytosis, and autophagy, promoting the clearance of substrates stored in cells. c-Abl is a tyrosine kinase that participates in cellular signaling in physiological and pathophysiological conditions. In this study, we explored the connection between c-Abl and TFEB. Here, we show that under pharmacological and genetic c-Abl inhibition, TFEB translocates into the nucleus promoting the expression of its target genes independently of its well-known regulator, mammalian target of rapamycin complex 1. Active c-Abl induces TFEB phosphorylation on tyrosine and the inhibition of this kinase promotes lysosomal biogenesis, autophagy, and exocytosis. c-Abl inhibition in Niemann-Pick type C (NPC) models, a neurodegenerative disease characterized by cholesterol accumulation in lysosomes, promotes a cholesterol-lowering effect in a TFEB-dependent manner. Thus, c-Abl is a TFEB regulator that mediates its tyrosine phosphorylation, and the inhibition of c-Abl activates TFEB promoting cholesterol clearance in NPC models.

6.
Cells ; 9(5)2020 05 10.
Article in English | MEDLINE | ID: mdl-32397616

ABSTRACT

The MiT/TFE family of transcription factors (MITF, TFE3, and TFEB), which control transcriptional programs for autophagy and lysosome biogenesis have emerged as regulators of energy metabolism in cancer. Thus, their activation increases lysosomal catabolic function to sustain cancer cell growth and survival in stress conditions. Here, we found that TFEB depletion dramatically reduces basal expression levels of the cyclin-dependent kinase (CDK) inhibitor p21/WAF1 in various cell types. Conversely, TFEB overexpression increases p21 in a p53-dependent manner. Furthermore, induction of DNA damage using doxorubicin induces TFEB-mediated activation of p21, delays G2/M phase arrest, and promotes cell survival. Pharmacological inhibition of p21, instead, abrogates TFEB-mediated protection during the DNA damage response. Together, our findings uncover a novel and direct role of TFEB in the regulation of p21 expression in both steady-state conditions and during the induction of DNA-damage response (DDR). Our observations might open novel therapeutic strategies to promote cancer cell death by targeting the TFEB-p21 pathway in the presence of genotoxic agents.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Cell Survival , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage/genetics , G2 Phase , Gene Expression Regulation , HeLa Cells , Humans , Mitosis , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
7.
EMBO J ; 39(8): e102468, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32154600

ABSTRACT

Vertebrate vision relies on the daily phagocytosis and lysosomal degradation of photoreceptor outer segments (POS) within the retinal pigment epithelium (RPE). However, how these events are controlled by light is largely unknown. Here, we show that the light-responsive miR-211 controls lysosomal biogenesis at the beginning of light-dark transitions in the RPE by targeting Ezrin, a cytoskeleton-associated protein essential for the regulation of calcium homeostasis. miR-211-mediated down-regulation of Ezrin leads to Ca2+ influx resulting in the activation of calcineurin, which in turn activates TFEB, the master regulator of lysosomal biogenesis. Light-mediated induction of lysosomal biogenesis and function is impaired in the RPE from miR-211-/- mice that show severely compromised vision. Pharmacological restoration of lysosomal biogenesis through Ezrin inhibition rescued the miR-211-/- phenotype, pointing to a new therapeutic target to counteract retinal degeneration associated with lysosomal dysfunction.


Subject(s)
Calcium/metabolism , Cytoskeletal Proteins/metabolism , Gene Expression Regulation , Lysosomes/metabolism , MicroRNAs/metabolism , Animals , Autophagy , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Down-Regulation , Light , Lysosomes/ultrastructure , Mice , Mice, Knockout , MicroRNAs/genetics , Phagocytosis , Phagosomes/metabolism , Phagosomes/ultrastructure , Retinal Pigment Epithelium/metabolism
8.
Stem Cell Reports ; 10(6): 1766-1781, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29681545

ABSTRACT

Human neural development begins at embryonic day 19 and marks the beginning of organogenesis. Neural stem cells in the neural tube undergo profound functional, morphological, and metabolic changes during neural specification, coordinated by a combination of exogenous and endogenous cues. The temporal cell signaling activities that mediate this process, during development and in the postnatal brain, are incompletely understood. We have applied gene expression studies and transcription factor-activated reporter lentiviruses during in vitro neural specification of human pluripotent stem cells. We show that nuclear factor κB orchestrates a multi-faceted metabolic program necessary for the maturation of neural progenitor cells during neurogenesis.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Energy Metabolism , NF-kappa B/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Autophagy , Biomarkers , Cell Cycle , Cell Differentiation/genetics , Cells, Cultured , Computational Biology/methods , Gene Expression Profiling , Gene Ontology , Humans , Immunohistochemistry , Models, Biological , Neurogenesis/genetics , Phenotype , Signal Transduction
9.
Stem Cells ; 34(1): 124-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26507573

ABSTRACT

The transcription factor REST is a key suppressor of neuronal genes in non-neuronal tissues. REST has been shown to suppress proneuronal microRNAs in neural progenitors indicating that REST-mediated neurogenic suppression may act in part via microRNAs. We used neural differentiation of Rest-null mouse ESC to identify dozens of microRNAs regulated by REST during neural development. One of the identified microRNAs, miR-375, was upregulated during human spinal motor neuron development. We found that miR-375 facilitates spinal motor neurogenesis by targeting the cyclin kinase CCND2 and the transcription factor PAX6. Additionally, miR-375 inhibits the tumor suppressor p53 and protects neurons from apoptosis in response to DNA damage. Interestingly, motor neurons derived from a spinal muscular atrophy patient displayed depressed miR-375 expression and elevated p53 protein levels. Importantly, SMA motor neurons were significantly more susceptible to DNA damage induced apoptosis suggesting that miR-375 may play a protective role in motor neurons.


Subject(s)
MicroRNAs/genetics , Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration/pathology , Animals , Apoptosis/genetics , Base Sequence , Humans , Mice , MicroRNAs/metabolism , Molecular Sequence Data , Muscular Atrophy, Spinal/genetics , Nerve Degeneration/genetics , Neurogenesis/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism
10.
J Neurosci Res ; 93(8): 1203-14, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25691247

ABSTRACT

Adult neural stem cell (aNSC) activity is tuned by external stimuli through the recruitment of transcription factors. This study examines the RE1 silencing transcription factor (REST) in neural stem/progenitor cells isolated from the subventricular zone of adult mouse brain and provides the first extensive characterization of REST-mediated control of the cellular and molecular properties. This study shows that REST knockdown affects the capacity of progenitor cells to generate neurospheres, reduces cell proliferation, and triggers cell differentiation despite the presence of growth factors. Genome- and transcriptome-wide analyses show that REST binding sites are significantly enriched in genes associated with synaptic transmission and nervous system development and function. Seeking candidate regulators of aNSC function, this study identifies a member of the bone morphogenetic protein (BMP) family, BMP6, the mRNA and protein of which increased after REST knockdown. The results of this study extend previous findings, demonstrating a reciprocal control of REST expression by BMPs. Administration of exogenous BMP6 inhibits aNSC proliferation and induces the expression of the astrocytic marker glial fibrillary acidic protein, highlighting its antimitogenic and prodifferentiative effects. This study suggests that BMP6 produced in a REST-regulated manner together with other signals can contribute to regulation of NSC maintenance and fate.


Subject(s)
Adult Stem Cells/physiology , Gene Silencing/physiology , Lateral Ventricles/cytology , Lateral Ventricles/physiology , Neural Stem Cells/physiology , Repressor Proteins/physiology , Animals , Bone Morphogenetic Protein 6/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Humans , Male , Mice , Transcription Factors/physiology
11.
J Neurochem ; 124(3): 418-30, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23145961

ABSTRACT

Huntingtin (Htt) protein interacts with many transcriptional regulators, with widespread disruption to the transcriptome in Huntington's disease (HD) brought about by altered interactions with the mutant Htt (muHtt) protein. Repressor Element-1 Silencing Transcription Factor (REST) is a repressor whose association with Htt in the cytoplasm is disrupted in HD, leading to increased nuclear REST and concomitant repression of several neuronal-specific genes, including brain-derived neurotrophic factor (Bdnf). Here, we explored a wide set of HD dysregulated genes to identify direct REST targets whose expression is altered in a cellular model of HD but that can be rescued by knock-down of REST activity. We found many direct REST target genes encoding proteins important for nervous system development, including a cohort involved in synaptic transmission, at least two of which can be rescued at the protein level by REST knock-down. We also identified several microRNAs (miRNAs) whose aberrant repression is directly mediated by REST, including miR-137, which has not previously been shown to be a direct REST target in mouse. These data provide evidence of the contribution of inappropriate REST-mediated transcriptional repression to the widespread changes in coding and non-coding gene expression in a cellular model of HD that may affect normal neuronal function and survival.


Subject(s)
Huntington Disease/genetics , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Neurons/pathology , Nuclear Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/physiology , Animals , Cells, Cultured , Corpus Striatum/cytology , Gene Expression Regulation/physiology , Gene Knock-In Techniques , Gene Knockdown Techniques , Humans , Huntingtin Protein , Huntington Disease/pathology , Mice , Nerve Tissue Proteins/genetics , Neurons/cytology , Nuclear Proteins/genetics , RNA, Small Interfering/genetics , Repressor Proteins/genetics
12.
PLoS One ; 7(12): e51798, 2012.
Article in English | MEDLINE | ID: mdl-23240065

ABSTRACT

During neural development, spatially regulated expression of specific transcription factors is crucial for central nervous system (CNS) regionalization, generation of neural precursors (NPs) and subsequent differentiation of specific cell types within defined regions. A critical role in dopaminergic differentiation in the midbrain (MB) has been assigned to the transcription factor Nurr1. Nurr1 controls the expression of key genes involved in dopamine (DA) neurotransmission, e.g. tyrosine hydroxylase (TH) and the DA transporter (DAT), and promotes the dopaminergic phenotype in embryonic stem cells. We investigated whether cells derived from different areas of the mouse CNS could be directed to differentiate into dopaminergic neurons in vitro by forced expression of the transcription factor Nurr1. We show that Nurr1 overexpression can promote dopaminergic cell fate specification only in NPs obtained from E13.5 ganglionic eminence (GE) and MB, but not in NPs isolated from E13.5 cortex (CTX) and spinal cord (SC) or from the adult subventricular zone (SVZ). Confirming previous studies, we also show that Nurr1 overexpression can increase the generation of TH-positive neurons in mouse embryonic stem cells. These data show that Nurr1 ability to induce a dopaminergic phenotype becomes restricted during CNS development and is critically dependent on the region of NPs derivation. Our results suggest that the plasticity of NPs and their ability to activate a dopaminergic differentiation program in response to Nurr1 is regulated during early stages of neurogenesis, possibly through mechanisms controlling CNS regionalization.


Subject(s)
Central Nervous System , Mesencephalon , Neurogenesis , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Animals , Cell Differentiation , Central Nervous System/growth & development , Central Nervous System/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Mesencephalon/cytology , Mesencephalon/growth & development , Mesencephalon/metabolism , Mice , Neurons/cytology , Neurons/metabolism , Synaptic Transmission
13.
Stem Cells ; 30(3): 425-34, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22162260

ABSTRACT

Neural differentiation of embryonic stem cells (ESCs) requires coordinated repression of the pluripotency regulatory program and reciprocal activation of the neurogenic regulatory program. Upon neural induction, ESCs rapidly repress expression of pluripotency genes followed by staged activation of neural progenitor and differentiated neuronal and glial genes. The transcriptional factors that underlie maintenance of pluripotency are partially characterized whereas those underlying neural induction are much less explored, and the factors that coordinate these two developmental programs are completely unknown. One transcription factor, REST (repressor element 1 silencing transcription factor), has been linked with terminal differentiation of neural progenitors and more recently, and controversially, with control of pluripotency. Here, we show that in the absence of REST, coordination of pluripotency and neural induction is lost and there is a resultant delay in repression of pluripotency genes and a precocious activation of both neural progenitor and differentiated neuronal and glial genes. Furthermore, we show that REST is not required for production of radial glia-like progenitors but is required for their subsequent maintenance and differentiation into neurons, oligodendrocytes, and astrocytes. We propose that REST acts as a regulatory hub that coordinates timely repression of pluripotency with neural induction and neural differentiation.


Subject(s)
Embryonic Stem Cells/physiology , Neurogenesis , Pluripotent Stem Cells/physiology , Repressor Proteins/physiology , Animals , Benzamides/pharmacology , Cell Differentiation , Dioxoles/pharmacology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Mice , Mice, Knockout , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Pluripotent Stem Cells/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism
14.
J Neurochem ; 116(3): 415-25, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21105876

ABSTRACT

Transcriptional dysfunction is a prominent hallmark of Huntington's disease (HD). Several transcription factors have been implicated in the aetiology of HD progression and one of the most prominent is repressor element 1 (RE1) silencing transcription factor (REST). REST is a global repressor of neuronal gene expression and in the presence of mutant Huntingtin increased nuclear REST levels lead to elevated RE1 occupancy and a concomitant increase in target gene repression, including brain-derived neurotrophic factor. It is of great interest to devise strategies to reverse transcriptional dysregulation caused by increased nuclear REST and determine the consequences in HD. Thus far, such strategies have involved RNAi or mutant REST constructs. Decoys are double-stranded oligodeoxynucleotides corresponding to the DNA-binding element of a transcription factor and act to sequester it, thereby abrogating its transcriptional activity. Here, we report the use of a novel decoy strategy to rescue REST target gene expression in a cellular model of HD. We show that delivery of the decoy in cells expressing mutant Huntingtin leads to its specific interaction with REST, a reduction in REST occupancy of RE1s and rescue of target gene expression, including Bdnf. These data point to an alternative strategy for rebalancing the transcriptional dysregulation in HD.


Subject(s)
Genetic Therapy/methods , Huntington Disease/genetics , Huntington Disease/therapy , Oligonucleotides/genetics , Oligonucleotides/pharmacology , Repressor Proteins/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Knock-In Techniques , Gene Silencing , Humans , Huntingtin Protein , Huntington Disease/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotides/therapeutic use , Repressor Proteins/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
15.
PLoS One ; 4(3): e4715, 2009.
Article in English | MEDLINE | ID: mdl-19262748

ABSTRACT

BACKGROUND: Oncogene activation plays a role in metabolic reprogramming of cancer cells. We have previously shown that K-ras transformed fibroblasts have a stronger dependence on glycolysis and a reduced oxidative phosphorylation ability as compared to their normal counterparts. Another metabolic adaptation of cancer cells, that has long been established, is their propensity to exhibit increased glutamine consumption, although the effects induced by glutamine deprivation on cancer cells are still controversial. METHODOLOGY AND PRINCIPAL FINDINGS: Here, by using nutritional perturbations and molecular physiology, we show that reduction or complete depletion of glutamine availability in K-ras transformed fibroblasts causes a strong decrease of proliferation ability and a slower re-entry of synchronized cells into the cell cycle. The reduced proliferation is accompanied by sustained expression of cyclin D and E, abortive S phase entrance and is dependent on Ras signalling deregulation, since it is rescued by expression of a dominant negative guanine nucleotide exchange factor. The growth potential of transformed cells as well as the ability to execute the G(1) to S transition is restored by adding the four deoxyribonucleotides, indicating that the arrest of proliferation of K-ras transformed cells induced by glutamine depletion is largely due to a reduced supply of DNA in the presence of signalling pathways promoting G(1) to S transition. CONCLUSIONS AND SIGNIFICANCE: Our results suggest that the differential effects of glutamine and glucose on cell viability are not a property of the transformed phenotype per se, but rather depend on the specific pathway being activated in transformation. For instance, myc-overexpressing cells have been reported to die under glutamine depletion and not under glucose shortage, while the opposite holds for ras-transformed fibroblasts as shown in this paper. These different responses of transformed cells to nutritional stress should be taken into account when designing anti-cancer therapies that aim to exploit metabolic differences between normal and transformed cells.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Deoxyribonucleotides/pharmacology , Fibroblasts/pathology , Genes, ras , Glutamine/deficiency , S Phase/drug effects , 3T3 Cells , Animals , Cell Cycle , Cell Proliferation , Fibroblasts/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...