Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Lancet Haematol ; 10(8): e575-e576, 2023 08.
Article in English | MEDLINE | ID: mdl-37429305
2.
Lancet Haematol ; 9(2): e92-e93, 2022 02.
Article in English | MEDLINE | ID: mdl-34922648
5.
Lancet Haematol ; 6(11): e550, 2019 11.
Article in English | MEDLINE | ID: mdl-31645279
6.
Lancet Haematol ; 6(9): e445, 2019 09.
Article in English | MEDLINE | ID: mdl-31471005
8.
Lancet Oncol ; 20(7): 909-910, 2019 07.
Article in English | MEDLINE | ID: mdl-31178374
9.
10.
Brain Behav Immun ; 64: 65-70, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28012829

ABSTRACT

Somatic LINE-1 (L1) retrotransposition is a source of genomic mosaicism and potential phenotypic diversity among neurons during brain development. In the adult brain, L1 expression can be triggered by different environmental alterations, but its functional role in this context remains unknown. Here we demonstrate a neural activation-dependent increase in the number of L1 retrotransposon insertions in the hippocampus. Using both pharmacologic and genetic approaches in mice, we demonstrate that L1 expression in the adult hippocampus enables long-term memory formation. These results provide experimental evidence that L1 retrotransposition-induced genomic mosaicism is involved in cognitive processes such as memory formation.


Subject(s)
Hippocampus/metabolism , Long Interspersed Nucleotide Elements , Memory, Long-Term , Retroelements , Animals , Genomics , Male , Mice , Mosaicism
11.
Cell Rep ; 13(11): 2456-2469, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26670048

ABSTRACT

During metastatic colonization, tumor cells must establish a favorable microenvironment or niche that will sustain their growth. However, both the temporal and molecular details of this process remain poorly understood. Here, we found that metastatic initiating cells (MICs) exhibit a high capacity for lung fibroblast activation as a result of Thrombospondin 2 (THBS2) expression. Importantly, inhibiting the mesenchymal phenotype of MICs by blocking the epithelial-to-mesenchymal transition (EMT)-associated kinase AXL reduces THBS2 secretion, niche-activating ability, and, consequently, metastatic competence. Subsequently, disseminated metastatic cells revert to an AXL-negative, more epithelial phenotype to proliferate and decrease the phosphorylation levels of TGF-ß-dependent SMAD2-3 in favor of BMP/SMAD1-5 signaling. Remarkably, newly activated fibroblasts promote this transition. In summary, our data reveal a crosstalk between cancer cells and their microenvironment whereby the EMT status initially triggers and then is regulated by niche activation during metastatic colonization.


Subject(s)
Neoplastic Stem Cells/metabolism , Thrombospondins/metabolism , Animals , Benzocycloheptenes/toxicity , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD24 Antigen/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis , Neoplastic Stem Cells/cytology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Thrombospondins/antagonists & inhibitors , Thrombospondins/genetics , Transforming Growth Factor beta/metabolism , Transplantation, Heterologous , Triazoles/toxicity , Axl Receptor Tyrosine Kinase
12.
Nutrition ; 28(11-12): 1200-3, 2012.
Article in English | MEDLINE | ID: mdl-22898267

ABSTRACT

Fibromyalgia (FM) is a chronic pain syndrome with unknown etiology. Recent studies have shown evidence demonstrating that mitochondrial dysfunction and oxidative stress may have a role in the pathophysiology of FM. Coenzyme Q10 (CoQ10) is an essential electron carrier in the mitochondrial respiratory chain and a strong antioxidant. Low CoQ10 levels have been detected in patients with FM, and a significant decrease of clinical symptoms has been reported after oral CoQ10 supplementation. In this report, we show the effect of CoQ10 treatment on clinical symptoms, blood mononuclear cells, and mitochondrial and oxidative stress markers from a woman with FM. After CoQ10 treatment, the patient reported a significant improvement of clinical symptoms. At the cellular level, CoQ10 treatment restored mitochondrial dysfunction and the mtDNA copy number, decreased oxidative stress, and increased mitochondrial biogenesis. Our results suggest that CoQ10 could be an alternative therapeutic approach for FM.


Subject(s)
Ataxia/diet therapy , Dietary Supplements , Fibromyalgia/complications , Leukocytes, Mononuclear/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/diet therapy , Muscle Weakness/diet therapy , Ubiquinone/analogs & derivatives , Ataxia/complications , Female , Fibromyalgia/blood , Fibromyalgia/metabolism , Fibromyalgia/physiopathology , Humans , Middle Aged , Mitochondrial Diseases/complications , Mitochondrial Turnover , Muscle Weakness/complications , Oxidative Stress , Severity of Illness Index , Treatment Outcome , Ubiquinone/deficiency , Ubiquinone/therapeutic use
13.
J Neurosci ; 30(40): 13305-13, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20926656

ABSTRACT

Memory formation requires changes in gene expression, which are regulated by the activation of transcription factors and by changes in epigenetic factors. Poly[ADP]-ribosylation of nuclear proteins has been postulated as a chromatin modification involved in memory consolidation, although the mechanisms involved are not well characterized. Here we demonstrate that poly[ADP]-ribose polymerase 1 (PARP-1) activity and the poly[ADP]-ribosylation of proteins over a specific time course is required for the changes in synaptic plasticity related to memory stabilization in mice. At the molecular level, histone H1 poly[ADP]-ribosylation was evident in the hippocampus after the acquisition period, and it was selectively released in a PARP-1-dependent manner at the promoters of cAMP response element-binding protein and nuclear factor-κB dependent genes associated with learning and memory. These findings suggest that histone H1 poly[ADP]-ribosylation, and its loss at specific loci, is an epigenetic mechanism involved in the reprogramming of neuronal gene expression required for memory consolidation.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Learning/physiology , Poly Adenosine Diphosphate Ribose/metabolism , Proteins/metabolism , Animals , Chromatin/genetics , Epigenesis, Genetic/genetics , Exploratory Behavior/physiology , Gene Expression Regulation/physiology , Genetic Loci/genetics , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Histones/physiology , Male , Memory/physiology , Mice , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/physiology , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/physiology , Promoter Regions, Genetic/genetics , Proteins/physiology , Synaptic Transmission/genetics
14.
Curr Biol ; 19(1): 54-60, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19110430

ABSTRACT

Memory deficits in aging affect millions of people and are often disturbing to those concerned. Dissection of the molecular control of learning and memory is paramount to understand and possibly enhance cognitive functions. Old-age memory loss also has been recently linked to altered Ca(2+) homeostasis. We have previously identified DREAM (downstream regulatory element antagonistic modulator), a member of the neuronal Ca(2+) sensor superfamily of EF-hand proteins, with specific roles in different cell compartments. In the nucleus, DREAM is a Ca(2+)-dependent transcriptional repressor, binding to specific DNA signatures, or interacting with nucleoproteins regulating their transcriptional properties. Also, we and others have shown that dream mutant (dream(-/-)) mice exhibit marked analgesia. Here we report that dream(-/-) mice exhibit markedly enhanced learning and synaptic plasticity related to improved cognition. Mechanistically, DREAM functions as a negative regulator of the key memory factor CREB in a Ca(2+)-dependent manner, and loss of DREAM facilitates CREB-dependent transcription during learning. Intriguingly, 18-month-old dream(-/-) mice display learning and memory capacities similar to young mice. Moreover, loss of DREAM protects from brain degeneration in aging. These data identify the Ca(2+)-regulated "pain gene" DREAM as a novel key regulator of memory and brain aging.


Subject(s)
Aging/physiology , Kv Channel-Interacting Proteins/deficiency , Learning/physiology , Memory/physiology , Aging/genetics , Analysis of Variance , Animals , Blotting, Western , Calcium/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , DNA/metabolism , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Electrophysiology , Hippocampus/physiology , Immunohistochemistry , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Mice , Mice, Knockout , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...