Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 5856, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37730692

ABSTRACT

The impact of genetic ablation of SOS1 or SOS2 is evaluated in a murine model of KRASG12D-driven lung adenocarcinoma (LUAD). SOS2 ablation shows some protection during early stages but only SOS1 ablation causes significant, specific long term increase of survival/lifespan of the KRASG12D mice associated to markedly reduced tumor burden and reduced populations of cancer-associated fibroblasts, macrophages and T-lymphocytes in the lung tumor microenvironment (TME). SOS1 ablation also causes specific shrinkage and regression of LUAD tumoral masses and components of the TME in pre-established KRASG12D LUAD tumors. The critical requirement of SOS1 for KRASG12D-driven LUAD is further confirmed by means of intravenous tail injection of KRASG12D tumor cells into SOS1KO/KRASWT mice, or of SOS1-less, KRASG12D tumor cells into wildtype mice. In silico analyses of human lung cancer databases support also the dominant role of SOS1 regarding tumor development and survival in LUAD patients. Our data indicate that SOS1 is critically required for development of KRASG12D-driven LUAD and confirm the validity of this RAS-GEF activator as an actionable therapeutic target in KRAS mutant LUAD.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Cancer-Associated Fibroblasts , Lung Neoplasms , Animals , Humans , Mice , Adenocarcinoma/genetics , Adenocarcinoma of Lung/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Microenvironment/genetics
2.
Int J Mol Sci ; 24(8)2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37108381

ABSTRACT

The isolation of the first human oncogene (HRAS), a critical breakthrough in cancer research, has occurred over forty years ago, and the identification of new pathogenic oncogenes has continuously grown since [...].


Subject(s)
Oncogenes , Humans
3.
Cancers (Basel) ; 14(16)2022 Aug 11.
Article in English | MEDLINE | ID: mdl-36010887

ABSTRACT

We showed previously that the ABL-mediated phosphorylation of SOS1 promotes RAC activation and contributes to BCR-ABL leukemogenesis, suggesting the relevant role of SOS1 in the pathogenesis of CML. To try and obtain direct experimental evidence of the specific mechanistic implication of SOS1 in CML development, here, we combined a murine model of CML driven by a p210BCR/ABL transgene with our tamoxifen-inducible SOS1/2-KO system in order to investigate the phenotypic impact of the direct genetic ablation of SOS1 or SOS2 on the pathogenesis of CML. Our observations showed that, in contrast to control animals expressing normal levels of SOS1 and SOS2 or to single SOS2-KO mice, p210BCR/ABL transgenic mice devoid of SOS1 presented significantly extended survival curves and also displayed an almost complete disappearance of the typical hematological alterations and splenomegaly constituting the hallmarks of CML. SOS1 ablation also resulted in a specific reduction in the proliferation and the total number of colony-forming units arising from the population of bone marrow stem/progenitor cells from p210BCR/ABL transgenic mice. The specific blockade of CML development caused by SOS1 ablation in p210BCR/ABL mice indicates that SOS1 is critically required for CML pathogenesis and supports the consideration of this cellular GEF as a novel, alternative bona fide therapeutic target for CML treatment in the clinic.

4.
Biomedicines ; 9(9)2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34572343

ABSTRACT

Recent reports have identified rare, biallelic damaging variants of the AGTPBP1 gene that cause a novel and documented human disease known as childhood-onset neurodegeneration with cerebellar atrophy (CONDCA), linking loss of function of the AGTPBP1 protein to human neurodegenerative diseases. CONDCA patients exhibit progressive cognitive decline, ataxia, hypotonia or muscle weakness among other clinical features that may be fatal. Loss of AGTPBP1 in humans recapitulates the neurodegenerative course reported in a well-characterised murine animal model harbouring loss-of-function mutations in the AGTPBP1 gene. In particular, in the Purkinje cell degeneration (pcd) mouse model, mutations in AGTPBP1 lead to early cerebellar ataxia, which correlates with the massive loss of cerebellar Purkinje cells. In addition, neurodegeneration in the olfactory bulb, retina, thalamus and spinal cord were also reported. In addition to neurodegeneration, pcd mice show behavioural deficits such as cognitive decline. Here, we provide an overview of what is currently known about the structure and functional role of AGTPBP1 and discuss the various alterations in AGTPBP1 that cause neurodegeneration in the pcd mutant mouse and humans with CONDCA. The sequence of neuropathological events that occur in pcd mice and the mechanisms governing these neurodegenerative processes are also reported. Finally, we describe the therapeutic strategies that were applied in pcd mice and focus on the potential usefulness of pcd mice as a promising model for the development of new therapeutic strategies for clinical trials in humans, which may offer potential beneficial options for patients with AGTPBP1 mutation-related CONDCA.

5.
Int J Mol Sci ; 22(12)2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34205562

ABSTRACT

The SOS family of Ras-GEFs encompasses two highly homologous and widely expressed members, SOS1 and SOS2. Despite their similar structures and expression patterns, early studies of constitutive KO mice showing that SOS1-KO mutants were embryonic lethal while SOS2-KO mice were viable led to initially viewing SOS1 as the main Ras-GEF linking external stimuli to downstream RAS signaling, while obviating the functional significance of SOS2. Subsequently, different genetic and/or pharmacological ablation tools defined more precisely the functional specificity/redundancy of the SOS1/2 GEFs. Interestingly, the defective phenotypes observed in concomitantly ablated SOS1/2-DKO contexts are frequently much stronger than in single SOS1-KO scenarios and undetectable in single SOS2-KO cells, demonstrating functional redundancy between them and suggesting an ancillary role of SOS2 in the absence of SOS1. Preferential SOS1 role was also demonstrated in different RASopathies and tumors. Conversely, specific SOS2 functions, including a critical role in regulation of the RAS-PI3K/AKT signaling axis in keratinocytes and KRAS-driven tumor lines or in control of epidermal stem cell homeostasis, were also reported. Specific SOS2 mutations were also identified in some RASopathies and cancer forms. The relevance/specificity of the newly uncovered functional roles suggests that SOS2 should join SOS1 for consideration as a relevant biomarker/therapy target.


Subject(s)
SOS1 Protein/physiology , Son of Sevenless Proteins/physiology , Animals , Humans , Neoplasms/metabolism
6.
Oncogene ; 40(27): 4538-4551, 2021 07.
Article in English | MEDLINE | ID: mdl-34120142

ABSTRACT

SOS1 ablation causes specific defective phenotypes in MEFs including increased levels of intracellular ROS. We showed that the mitochondria-targeted antioxidant MitoTEMPO restores normal endogenous ROS levels, suggesting predominant involvement of mitochondria in generation of this defective SOS1-dependent phenotype. The absence of SOS1 caused specific alterations of mitochondrial shape, mass, and dynamics accompanied by higher percentage of dysfunctional mitochondria and lower rates of electron transport in comparison to WT or SOS2-KO counterparts. SOS1-deficient MEFs also exhibited specific alterations of respiratory complexes and their assembly into mitochondrial supercomplexes and consistently reduced rates of respiration, glycolysis, and ATP production, together with distinctive patterns of substrate preference for oxidative energy metabolism and dependence on glucose for survival. RASless cells showed defective respiratory/metabolic phenotypes reminiscent of those of SOS1-deficient MEFs, suggesting that the mitochondrial defects of these cells are mechanistically linked to the absence of SOS1-GEF activity on cellular RAS targets. Our observations provide a direct mechanistic link between SOS1 and control of cellular oxidative stress and suggest that SOS1-mediated RAS activation is required for correct mitochondrial dynamics and function.


Subject(s)
Mitochondrial Dynamics , Homeostasis , ras Guanine Nucleotide Exchange Factors
7.
Cancers (Basel) ; 13(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946974

ABSTRACT

Prior reports showed the critical requirement of Sos1 for epithelial carcinogenesis, but the specific functionalities of the homologous Sos1 and Sos2 GEFs in skin homeostasis and tumorigenesis remain unclear. Here, we characterize specific mechanistic roles played by Sos1 or Sos2 in primary mouse keratinocytes (a prevalent skin cell lineage) under different experimental conditions. Functional analyses of actively growing primary keratinocytes of relevant genotypes-WT, Sos1-KO, Sos2-KO, and Sos1/2-DKO-revealed a prevalent role of Sos1 regarding transcriptional regulation and control of RAS activation and mechanistic overlapping of Sos1 and Sos2 regarding cell proliferation and survival, with dominant contribution of Sos1 to the RAS-ERK axis and Sos2 to the RAS-PI3K/AKT axis. Sos1/2-DKO keratinocytes could not grow under 3D culture conditions, but single Sos1-KO and Sos2-KO keratinocytes were able to form pseudoepidermis structures that showed disorganized layer structure, reduced proliferation, and increased apoptosis in comparison with WT 3D cultures. Remarkably, analysis of the skin of both newborn and adult Sos2-KO mice uncovered a significant reduction of the population of stem cells located in hair follicles. These data confirm that Sos1 and Sos2 play specific, cell-autonomous functions in primary keratinocytes and reveal a novel, essential role of Sos2 in control of epidermal stem cell homeostasis.

8.
Biochim Biophys Acta Rev Cancer ; 1874(2): 188445, 2020 12.
Article in English | MEDLINE | ID: mdl-33035641

ABSTRACT

SOS1 and SOS2 are the most universal and widely expressed family of guanine exchange factors (GEFs) capable or activating RAS or RAC1 proteins in metazoan cells. SOS proteins contain a sequence of modular domains that are responsible for different intramolecular and intermolecular interactions modulating mechanisms of self-inhibition, allosteric activation and intracellular homeostasis. Despite their homology, analyses of SOS1/2-KO mice demonstrate functional prevalence of SOS1 over SOS2 in cellular processes including proliferation, migration, inflammation or maintenance of intracellular redox homeostasis, although some functional redundancy cannot be excluded, particularly at the organismal level. Specific SOS1 gain-of-function mutations have been identified in inherited RASopathies and various sporadic human cancers. SOS1 depletion reduces tumorigenesis mediated by RAS or RAC1 in mouse models and is associated with increased intracellular oxidative stress and mitochondrial dysfunction. Since WT RAS is essential for development of RAS-mutant tumors, the SOS GEFs may be considered as relevant biomarkers or therapy targets in RAS-dependent cancers. Inhibitors blocking SOS expression, intrinsic GEF activity, or productive SOS protein-protein interactions with cellular regulators and/or RAS/RAC targets have been recently developed and shown preclinical and clinical effectiveness blocking aberrant RAS signaling in RAS-driven and RTK-driven tumors.


Subject(s)
Mutation , Neoplasms/genetics , Son of Sevenless Proteins/genetics , Son of Sevenless Proteins/metabolism , Allosteric Regulation , Animals , Homeostasis , Humans , Mice , Neoplasms/metabolism , rac1 GTP-Binding Protein/metabolism , ras Proteins/metabolism
9.
Hum Gene Ther ; 31(7-8): 415-422, 2020 04.
Article in English | MEDLINE | ID: mdl-32126838

ABSTRACT

Nerve growth factor (NGF) gene therapy rescues and stimulates cholinergic neurons, which degenerate in Alzheimer's disease (AD). In a recent clinical trial for AD, intraparenchymal adeno-associated virus serotype 2 (AAV2)-NGF delivery was safe but did not improve cognition. Before concluding that NGF gene therapy is ineffective, it must be shown that AAV2-NGF successfully engaged the target cholinergic neurons of the basal forebrain. In this study, patients with clinically diagnosed early- to middle-stage AD received a total dose of 2 × 1011 vector genomes of AAV2-NGF by stereotactic injection of the nucleus basalis of Meynert. After a mean survival of 4.0 years, AAV2-NGF targeting, spread, and expression were assessed by immunolabeling of NGF and the low-affinity NGF receptor p75 at 15 delivery sites in 3 autopsied patients. NGF gene expression persisted for at least 7 years at sites of AAV2-NGF injection. However, the mean distance of AAV2-NGF spread was only 0.96 ± 0.34 mm. NGF did not directly reach cholinergic neurons at any of the 15 injection sites due to limited spread and inaccurate stereotactic targeting. Because AAV2-NGF did not directly engage the target cholinergic neurons, we cannot conclude that growth factor gene therapy is ineffective for AD. Upcoming clinical trials for AD will utilize real-time magnetic resonance imaging guidance and convection-enhanced delivery to improve the targeting and spread of growth factor gene delivery.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/therapy , Dependovirus , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Nerve Growth Factor/genetics , Aged , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Autopsy , Basal Forebrain/pathology , Cholinergic Neurons/metabolism , Female , Humans , Male , Middle Aged , Neuropsychological Tests
10.
Neurobiol Dis ; 127: 312-322, 2019 07.
Article in English | MEDLINE | ID: mdl-30905767

ABSTRACT

The Purkinje cell (PC) degeneration (pcd) mouse harbors a mutation in Agtpbp1 gene that encodes for the cytosolic carboxypeptidase, CCP1. The mutation causes degeneration and death of PCs during the postnatal life, resulting in clinical and pathological manifestation of cerebellar ataxia. Monogenic biallelic damaging variants in the Agtpbp1 gene cause infantile-onset neurodegeneration and cerebellar atrophy, linking loss of functional CCP1 with human neurodegeneration. Although CCP1 plays a key role in the regulation of tubulin stabilization, its loss of function in PCs leads to a severe nuclear phenotype with heterochromatinization and accumulation of DNA damage. Therefore, the pcd mice provides a useful neuronal model to investigate nuclear mechanisms involved in neurodegeneration, particularly the nucleolar stress. In this study, we demonstrated that the Agtpbp1 gene mutation induces a p53-dependent nucleolar stress response in PCs, which is characterized by nucleolar fragmentation, nucleoplasmic and cytoplasmic mislocalization of nucleolin, and dysfunction of both pre-rRNA processing and mRNA translation. RT-qPCR analysis revealed reduction of mature 18S rRNA, with a parallel increase of its intermediate 18S-5'-ETS precursor, that correlates with a reduced expression of Fbl mRNA, which encodes an essential factor for rRNA processing. Moreover, nucleolar alterations were accompanied by a reduction of PTEN mRNA and protein levels, which appears to be related to the chromosome instability and accumulation of DNA damage in degenerating PCs. Our results highlight the essential contribution of nucleolar stress to PC degeneration and also underscore the nucleoplasmic mislocalization of nucleolin as a potential indicator of neurodegenerative processes.


Subject(s)
Cell Nucleolus/metabolism , GTP-Binding Proteins/metabolism , Phosphoproteins/metabolism , Purkinje Cells/metabolism , RNA-Binding Proteins/metabolism , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , Animals , GTP-Binding Proteins/genetics , Mice , Mutation , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Purkinje Cells/pathology , Serine-Type D-Ala-D-Ala Carboxypeptidase/genetics , Nucleolin
11.
J Leukoc Biol ; 106(4): 815-822, 2019 10.
Article in English | MEDLINE | ID: mdl-30720883

ABSTRACT

Circulating neutrophils are, by necessity, quiescent and relatively unresponsive to acute stimuli. In regions of inflammation, mediators can prime neutrophils to react to acute stimuli with stronger proinflammatory, pathogen-killing responses. In neutrophils G protein-coupled receptor (GPCR)-driven proinflammatory responses, such as reactive oxygen species (ROS) formation and accumulation of the key intracellular messenger phosphatidylinositol (3,4,5)-trisphosphate (PIP3 ), are highly dependent on PI3K-γ, a Ras-GTP, and Gßγ coincidence detector. In unprimed cells, the major GPCR-triggered activator of Ras is the Ras guanine nucleotide exchange factor (GEF), Ras guanine nucleotide releasing protein 4 (RasGRP4). Although priming is known to increase GPCR-PIP3 signaling, the mechanisms underlying this augmentation remain unclear. We used genetically modified mice to address the role of the 2 RasGEFs, RasGRP4 and son of sevenless (SOS)1/2, in neutrophil priming. We found that following GM-CSF/TNFα priming, RasGRP4 had only a minor role in the enhanced responses. In contrast, SOS1/2 acquired a substantial role in ROS formation, PIP3 accumulation, and ERK activation in primed cells. These results suggest that SOS1/2 signaling plays a key role in determining the responsiveness of neutrophils in regions of inflammation.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation/pathology , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/metabolism , SOS1 Protein/metabolism , Son of Sevenless Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , ras Proteins/metabolism , Animals , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Phosphatidylinositol Phosphates/metabolism , Phospholipase C beta/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , ras Guanine Nucleotide Exchange Factors/metabolism
12.
Mol Cell Biol ; 38(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29844066

ABSTRACT

Using Sos1 knockout (Sos1-KO), Sos2-KO, and Sos1/2 double-knockout (Sos1/2-DKO) mice, we assessed the functional role of Sos1 and Sos2 in skin homeostasis under physiological and/or pathological conditions. Sos1 depletion resulted in significant alterations of skin homeostasis, including reduced keratinocyte proliferation, altered hair follicle and blood vessel integrity in dermis, and reduced adipose tissue in hypodermis. These defects worsened significantly when both Sos1 and Sos2 were absent. Simultaneous Sos1/2 disruption led to severe impairment of the ability to repair skin wounds, as well as to almost complete ablation of the neutrophil-mediated inflammatory response in the injury site. Furthermore, Sos1 disruption delayed the onset of tumor initiation, decreased tumor growth, and prevented malignant progression of papillomas in a DMBA (7,12-dimethylbenz[α]anthracene)/TPA (12-O-tetradecanoylphorbol-13-acetate)-induced skin carcinogenesis model. Finally, Sos1 depletion in preexisting chemically induced papillomas resulted also in decreased tumor growth, probably linked to significantly reduced underlying keratinocyte proliferation. Our data unveil novel, distinctive mechanistic roles of Sos 1 and Sos2 in physiological control of skin homeostasis and wound repair, as well as in pathological development of chemically induced skin tumors. These observations underscore the essential role of Sos proteins in cellular proliferation and migration and support the consideration of these RasGEFs as potential biomarkers/therapy targets in Ras-driven epidermal tumors.


Subject(s)
SOS1 Protein/metabolism , Skin Neoplasms/etiology , Skin/metabolism , Son of Sevenless Proteins/metabolism , Animals , Carcinogenesis , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Homeostasis , Mice , Mice, Knockout , Neovascularization, Physiologic , Papilloma/metabolism , Papilloma/pathology , SOS1 Protein/deficiency , SOS1 Protein/genetics , Skin/blood supply , Skin/cytology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Son of Sevenless Proteins/deficiency , Son of Sevenless Proteins/genetics , Wound Healing
13.
Methods Mol Biol ; 1254: 43-54, 2015.
Article in English | MEDLINE | ID: mdl-25431056

ABSTRACT

Nuclear architecture is highly concerted including the organization of chromosome territories and distinct nuclear bodies, such as nucleoli, Cajal bodies, nuclear speckles of splicing factors, and promyelocytic leukemia nuclear bodies, among others. The organization of such nuclear compartments is very dynamic and may represent a sensitive indicator of the functional status of the cell. Here, we describe methodologies that allow isolating discrete cell populations from the brain and the fine observation of nuclear signs that could be insightful predictors of an early neuronal injury in a wide range of neurodegenerative disorders. The tools here described may be of use for the early detection of pre-degenerative processes in neurodegenerative diseases and for validating novel rescue strategies.


Subject(s)
Cell Compartmentation/genetics , Cell Nucleolus/pathology , Coiled Bodies/pathology , Neurodegenerative Diseases/pathology , Cell Nucleolus/genetics , Cell Nucleolus/ultrastructure , Coiled Bodies/genetics , Coiled Bodies/ultrastructure , Humans , Molecular Biology/methods , Neurodegenerative Diseases/genetics , RNA Splicing/genetics
14.
Mol Cell Biol ; 33(22): 4562-78, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24043312

ABSTRACT

Sos1 and Sos2 are ubiquitously expressed, universal Ras guanine nucleotide exchange factors (Ras-GEFs) acting in multiple signal transduction pathways activated by upstream cellular kinases. The embryonic lethality of Sos1 null mutants has hampered ascertaining the specific in vivo contributions of Sos1 and Sos2 to processes controlling adult organism survival or development of hematopoietic and nonhematopoietic organs, tissues, and cell lineages. Here, we generated a tamoxifen-inducible Sos1-null mouse strain allowing analysis of the combined disruption of Sos1 and Sos2 (Sos1/2) during adulthood. Sos1/2 double-knockout (DKO) animals died precipitously, whereas individual Sos1 and Sos2 knockout (KO) mice were perfectly viable. A reduced percentage of total bone marrow precursors occurred in single-KO animals, but a dramatic depletion of B-cell progenitors was specifically detected in Sos1/2 DKO mice. We also confirmed a dominant role of Sos1 over Sos2 in early thymocyte maturation, with almost complete thymus disappearance and dramatically higher reduction of absolute thymocyte counts in Sos1/2 DKO animals. Absolute counts of mature B and T cells in spleen and peripheral blood were unchanged in single-KO mutants, while significantly reduced in Sos1/2 DKO mice. Our data demonstrate functional redundancy between Sos1 and Sos2 for homeostasis and survival of the full organism and for development and maturation of T and B lymphocytes.


Subject(s)
Lymphopoiesis , SOS1 Protein/metabolism , Son of Sevenless Proteins/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Count , Female , Homeostasis , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Male , Mice , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , SOS1 Protein/genetics , Son of Sevenless Proteins/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
15.
Glia ; 61(2): 254-72, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23047288

ABSTRACT

Purkinje Cell Degeneration (PCD) mice harbor a nna1 gene mutation which leads to an early and rapid degeneration of Purkinje cells (PC) between the third and fourth week of age. This mutation also underlies the death of mitral cells (MC) in the olfactory bulb (OB), but this process is slower and longer than in PC. No clear interpretations supporting the marked differences in these neurodegenerative processes exist. Growing evidence suggests that either beneficial or detrimental effects of gliosis in damaged regions would underlie these divergences. Here, we examined the gliosis occurring during PC and MC death in the PCD mouse. Our results demonstrated different glial reactions in both affected regions. PC disappearance stimulated a severe gliosis characterized by strong morphological changes, enhanced glial proliferation, as well as the release of pro-inflammatory mediators. By contrast, MC degeneration seems to promote a more attenuated glial response in the PCD OB compared with that of the cerebellum. Strikingly, cerebellar oligodendrocytes died by apoptosis in the PCD, whereas bulbar ones were not affected. Interestingly, the level of nna1 mRNA under normal conditions was higher in the cerebellum than in the OB, probably related to a faster neurodegeneration and stronger glial reaction in its absence. The glial responses may thus influence the neurodegenerative course in the cerebellum and OB of the mutant mouse brain, providing harmful and beneficial microenvironments, respectively.


Subject(s)
GTP-Binding Proteins/genetics , Mutation/genetics , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neuroglia/physiology , Purkinje Cells/pathology , Serine-Type D-Ala-D-Ala Carboxypeptidase/genetics , Age Factors , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Calcium-Binding Proteins/metabolism , Cell Death/genetics , Cell Proliferation , Cerebellum/pathology , GTP-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Glial Fibrillary Acidic Protein/metabolism , Gliosis/genetics , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microscopy, Electron, Transmission , Nerve Tissue Proteins/metabolism , Olfactory Bulb/pathology , Oligonucleotide Array Sequence Analysis , Purkinje Cells/ultrastructure , RNA, Messenger , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism
16.
J Biol Chem ; 286(32): 28287-302, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21700704

ABSTRACT

DNA repair protects neurons against spontaneous or disease-associated DNA damage. Dysfunctions of this mechanism underlie a growing list of neurodegenerative disorders. The Purkinje cell (PC) degeneration mutation causes the loss of nna1 expression and is associated with the postnatal degeneration of PCs. This PC degeneration dramatically affects nuclear architecture and provides an excellent model to elucidate the nuclear mechanisms involved in a whole array of neurodegenerative disorders. We used immunocytochemistry for histone variants and components of the DNA damage response, an in situ transcription assay, and in situ hybridization for telomeres to analyze changes in chromatin architecture and function. We demonstrate that the phosphorylation of H2AX, a DNA damage signal, and the trimethylation of the histone H4K20, a repressive mark, in extensive domains of genome are epigenetic hallmarks of chromatin in degenerating PCs. These histone modifications are associated with a large scale reorganization of chromatin, telomere clustering, and heterochromatin-induced gene silencing, all of them key factors in PC degeneration. Furthermore, ataxia telangiectasia mutated and 53BP1, two components of the DNA repair pathway, fail to be concentrated in the damaged chromatin compartments, even though the expression levels of their coding genes were slightly up-regulated. Although the mechanism by which Nna1 loss of function leads to PC neurodegeneration is undefined, the progressive accumulation of DNA damage in chromosome territories irreversibly compromises global gene transcription and seems to trigger PC degeneration and death.


Subject(s)
Chromatin Assembly and Disassembly , DNA Repair , Gene Silencing , Neurodegenerative Diseases/metabolism , Purkinje Cells/metabolism , Signal Transduction , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Damage/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Histones/genetics , Histones/metabolism , Male , Mice , Mice, Mutant Strains , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Purkinje Cells/pathology , Serine-Type D-Ala-D-Ala Carboxypeptidase/genetics , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1
17.
Cell Transplant ; 20(8): 1179-92, 2011.
Article in English | MEDLINE | ID: mdl-21294954

ABSTRACT

Many studies have reported the contribution of bone marrow-derived cells (BMDC) to the CNS, raising the possibility of using them as a new source to repair damaged brain tissue or restore neuronal function. This process has mainly been investigated in the cerebellum, in which a degenerative microenvironment has been suggested to be responsible for its modulation. The present study further analyzes the contribution of BMDC to different neural types in other adult brain areas, under both physiological and neurodegenerative conditions, together with the mechanisms of plasticity involved. We grafted genetically marked green fluorescent protein/Cre bone marrow in irradiated recipients: a) the PCD (Purkinje Cell Degeneration) mutant mice, suffering a degeneration of specific neuronal populations at different ages, and b) their corresponding healthy controls. These mice carried the conditional lacZ reporter gene to allow the identification of cell fusion events. Our results demonstrate that BMDC mainly generate microglial cells, although to a lesser extent a clear formation of neuronal types also exists. This neuronal recruitment was not increased by the neurodegenerative processes occurring in PCD mice, where BMDC did not contribute to rescuing the degenerated neuronal populations either. However, an increase in the number of bone marrow-derived microglia was found along the life span in both experimental groups. Six weeks after transplantation more bone marrow-derived microglial cells were observed in the olfactory bulb of the PCD mice compared to the control animals, where the degeneration of mitral cells was in process. In contrast, this difference was not observed in the cerebellum, where Purkinje cell degeneration had been completed. These findings demonstrated that the degree of neurodegenerative environment can foster the recruitment of neural elements derived from bone marrow, but also provide the first evidence that BMDC can contribute simultaneously to different encephalic areas through different mechanisms of plasticity: cell fusion for Purkinje cells and differentiation for olfactory bulb interneurons.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Transplantation , Central Nervous System/pathology , Neuronal Plasticity/physiology , Neurons/pathology , Animals , Central Nervous System/physiopathology , Female , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/pathology , Microscopy, Fluorescence , Nerve Degeneration/pathology , Nerve Degeneration/therapy
18.
Brain Pathol ; 21(4): 374-88, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21054627

ABSTRACT

The Purkinje cell (PC) degeneration (pcd) phenotype results from mutation in nna1 gene and is associated with the degeneration and death of PCs during the postnatal life. Although the pcd mutation is a model of the ataxic mouse, it shares clinical and pathological characteristics of inherited human spinocerebellar ataxias. PC degeneration in pcd mice provides a useful neuronal system to study nuclear mechanisms involved in DNA damage-dependent neurodegeneration, particularly the contribution of nucleoli and Cajal bodies (CBs). Both nuclear structures are engaged in housekeeping functions for neuronal survival, the biogenesis of ribosomes and the maturation of snRNPs and snoRNPs required for pre-mRNA and pre-rRNA processing, respectively. In this study, we use ultrastructural analysis, in situ transcription assay and molecular markers for DNA damage, nucleoli and CB components to demonstrate that PC degeneration involves the progressive accumulation of nuclear DNA damage associated with disruption of nucleoli and CBs, disassembly of polyribosomes into monoribosomes, ribophagy and shut down of nucleolar and extranucleolar transcription. Microarray analysis reveals that four genes encoding repressors of nucleolar rRNA synthesis (p53, Rb, PTEN and SNF2) are upregulated in the cerebellum of pcd mice. Collectively, these data support that nucleolar and CB alterations are hallmarks of DNA damage-induced neurodegeneration.


Subject(s)
Cell Nucleolus/pathology , Coiled Bodies/pathology , DNA Damage , Nerve Degeneration/pathology , Purkinje Cells/pathology , Animals , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Coiled Bodies/genetics , Coiled Bodies/metabolism , Female , Fluorescent Antibody Technique , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Mutant Strains , Microscopy, Electron, Transmission , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Oligonucleotide Array Sequence Analysis , Purkinje Cells/metabolism
19.
J Neurosci Res ; 89(1): 35-43, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21046560

ABSTRACT

The periglomerular cells (PG) of the olfactory bulb (OB) are involved in the primary processing and the refinement of sensory information from the olfactory epithelium. The neurochemical composition of these neurons has been studied in depth in many species, and over the last decades such studies have focused mainly on the rat. The increasing use of genetic models for research into olfactory function demands a profound characterization of the mouse olfactory bulb, including the chemical composition of bulbar interneurons. Regarding both their connectivity with the olfactory nerve and their neurochemical fate, recently, two different types of PG have been identified in the mouse. In the present report, we analyze both the synaptology and the chemical composition of specific PG populations in the murine olfactory bulb, in particular, those containing the neuropeptide cholecystokinin. Our results demonstrate the existence in the mouse of non-GABAergic PG and that these establish synaptic contacts with the olfactory nerve within the glomeruli. Based on previous classifications, we propose that this population would constitute a new subtype of type 1 mouse PG. In addition, we demonstrate the partial coexistence of cholecystokinin with the calcium-binding proteins neurocalcin and parvalbumin. All these findings add further data to our knowledge of the synaptology and neurochemistry of mouse PG. The differences observed from other rodents reflect the neurochemical heterogeneity of PG in the mammalian OB.


Subject(s)
Cholecystokinin/metabolism , Interneurons/chemistry , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Olfactory Nerve/cytology , Olfactory Nerve/physiology , Synapses/physiology , Animals , Cholecystokinin/classification , Interneurons/classification , Interneurons/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Olfactory Nerve/metabolism , Synapses/chemistry , Synapses/metabolism
20.
Cell Mol Neurobiol ; 29(8): 1081-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19399607

ABSTRACT

The Pax6 transcription factor is a key element along brain development in both the visual and olfactory systems. The involvement of Pax6 in neural fate is well documented in the visual system, whereas in the olfactory system, and in particular in the olfactory bulb (OB), its expression during adulthood has only begun to be elucidated. In the OB, the modulation of primary sensory information is first performed by periglomerular cells (PG). A considerable body of information has unveiled the neurochemical heterogeneity of these neurons. Thus it is well known that Pax6 coexists with dopaminergic/GABAergic mouse PG. However, the presence of this transcription factor in other mouse PG subpopulations has not been studied. Here, we analyzed whether Pax6 is expressed in PG containing the calcium-binding proteins neurocalcin and parvalbumin, and the neuropeptide cholecystokinin. Our results show that Pax6 is not expressed by these PG subpopulations, suggesting that it is mainly restricted to GABAergic PG populations. These findings provide new data in the chemical characterization of mouse Pax6-positive PG.


Subject(s)
Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Neurons/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Cholecystokinin/metabolism , Male , Mice , Neurocalcin/metabolism , Neurons/cytology , PAX6 Transcription Factor , Parvalbumins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...