Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 12(1): 107-119, 2024 01 03.
Article in English | MEDLINE | ID: mdl-37922405

ABSTRACT

γδ T cells are a rare but potent subset of T cells with pleiotropic functions. They commonly reside within tumors but the response of γδ T cells to tyrosine kinase inhibition is unknown. To address this, we studied a genetically engineered mouse model of gastrointestinal stromal tumor (GIST) driven by oncogenic Kit signaling that responds to the Kit inhibitor imatinib. At baseline, γδ T cells were antitumoral, as blockade of either γδ T-cell receptor or IL17A increased tumor weight and decreased antitumor immunity. However, imatinib therapy further stimulated intratumoral γδ T cells, as determined by flow cytometry and single-cell RNA sequencing (scRNA-seq). Imatinib expanded a highly activated γδ T-cell subset with increased IL17A production and higher expression of immune checkpoints and cytolytic effector molecules. Consistent with the mouse model, γδ T cells produced IL17A in fresh human GIST specimens, and imatinib treatment increased γδ T-cell gene signatures, as measured by bulk tumor RNA-seq. Furthermore, tumor γδ T cells correlated with survival in patients with GIST. Our findings highlight the interplay between tumor cell oncogene signaling and antitumor immune responses and identify γδ T cells as targets for immunotherapy in GIST.


Subject(s)
Antineoplastic Agents , Gastrointestinal Stromal Tumors , Mice , Animals , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Gastrointestinal Stromal Tumors/pathology , Proto-Oncogene Proteins c-kit/genetics , Enzyme Inhibitors/therapeutic use , Signal Transduction , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology
2.
Oncogene ; 42(34): 2578-2588, 2023 08.
Article in English | MEDLINE | ID: mdl-37468679

ABSTRACT

Gastrointestinal stromal tumor (GIST) is the most common human sarcoma and is typically driven by a single mutation in the Kit or PDGFRA receptor. While highly effective, tyrosine kinase inhibitors (TKIs) are not curative. The natural ligand for the Kit receptor is Kit ligand (KitL), which exists in both soluble and membrane-bound forms. While KitL is known to stimulate human GIST cell lines in vitro, we used a genetically engineered mouse model of GIST containing a common human KIT mutation to investigate the intratumoral sources of KitL, importance of KitL during GIST oncogenesis, and contribution of soluble KitL to tumor growth in vivo. We discovered that in addition to tumor cells, endothelia and smooth muscle cells produced KitL in KitV558Δ/+ tumors, even after imatinib therapy. Genetic reduction of total KitL in tumor cells of KitV558Δ/+ mice impaired tumor growth in vivo. Similarly, genetic reduction of tumor cell soluble KitL in KitV558Δ/+ mice decreased tumor size. By RNA sequencing, quantitative PCR, and immunohistochemistry, KitL expression was heterogeneous in human GIST specimens. In particular, PDGFRA-mutant tumors had much higher KitL expression than Kit-mutant tumors, suggesting the benefit of Kit activation in the absence of mutant KIT. Serum KitL was higher in GIST patients with tumors resistant to imatinib and in those with tumors expressing more KitL RNA. Overall, KitL supports the growth of GIST at baseline and after imatinib therapy and remains a potential biomarker and therapeutic target.


Subject(s)
Antineoplastic Agents , Gastrointestinal Stromal Tumors , Humans , Mice , Animals , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Stem Cell Factor/genetics , Stem Cell Factor/pharmacology , Stem Cell Factor/therapeutic use , Pyrimidines/pharmacology , Piperazines/pharmacology , Piperazines/therapeutic use , Benzamides/pharmacology , Drug Resistance, Neoplasm/genetics , Proto-Oncogene Proteins c-kit , Mutation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
3.
Cancer Immunol Res ; 10(10): 1210-1223, 2022 10 04.
Article in English | MEDLINE | ID: mdl-35917579

ABSTRACT

Targeted therapy with a tyrosine kinase inhibitor (TKI) such as imatinib is effective in treating gastrointestinal stromal tumor (GIST), but it is rarely curative. Despite the presence of a robust immune CD8+ T-cell infiltrate, combining a TKI with immune-checkpoint blockade (ICB) in advanced GIST has achieved only modest effects. To identify limitations imposed by imatinib on the antitumor immune response, we performed bulk RNA sequencing (RNA-seq), single-cell RNA-seq, and flow cytometry to phenotype CD8+ T-cell subsets in a genetically engineered mouse model of GIST. Imatinib reduced the frequency of effector CD8+ T cells and increased the frequency of naïve CD8+ T cells within mouse GIST, which coincided with altered tumor chemokine production, CD8+ T-cell recruitment, and reduced CD8+ T-cell intracellular PI3K signaling. Imatinib also failed to induce intratumoral T-cell receptor (TCR) clonal expansion. Consistent with these findings, human GISTs sensitive to imatinib harbored fewer effector CD8+ T cells but more naïve CD8+ T cells. Combining an IL15 superagonist (IL15SA) with imatinib restored intratumoral effector CD8+ T-cell function and CD8+ T-cell intracellular PI3K signaling, resulting in greater tumor destruction. Combination therapy with IL15SA and ICB resulted in the greatest tumor killing and maintained an effector CD8+ T-cell population in the presence of imatinib. Our findings highlight the impact of oncogene inhibition on intratumoral CD8+ T cells and support the use of agonistic T-cell therapy during TKI and/or ICB administration.


Subject(s)
Gastrointestinal Stromal Tumors , Animals , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Chemokines , Gastrointestinal Stromal Tumors/drug therapy , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Immune Checkpoint Inhibitors , Interleukin-15/pharmacology , Mice , Phosphatidylinositol 3-Kinases , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/pharmacology
4.
J Vis Exp ; (183)2022 05 02.
Article in English | MEDLINE | ID: mdl-35575516

ABSTRACT

Gastrointestinal stromal tumor (GIST) is the most common human sarcoma and is typically driven by a single mutation in the KIT receptor. Across tumor types, numerous mouse models have been developed in order to investigate the next generation of cancer therapies. However, in GIST, most in vivo studies use xenograft mouse models which have inherent limitations. Here, we describe an immunocompetent, genetically engineered mouse model of gastrointestinal stromal tumor harboring a KitV558Δ/+ mutation. In this model, mutant KIT, the oncogene responsible for most GISTs, is driven by its endogenous promoter leading to a GIST which mimics the histological appearance and immune infiltrate seen in human GISTs. Furthermore, this model has been used successfully to investigate both targeted molecular and immune therapies. Here, we describe the breeding and maintenance of a KitV558Δ/+ mouse colony. Additionally, this paper details the treatment and procurement of GIST, draining mesenteric lymph node, and adjacent cecum in KitV558Δ/+ mice, as well as sample preparation for molecular and immunologic analyses.


Subject(s)
Gastrointestinal Stromal Tumors , Animals , Disease Models, Animal , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Humans , Immunologic Techniques , Mice , Mutation , Proto-Oncogene Proteins c-kit/genetics
5.
Neurobiol Dis ; 102: 49-59, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28237314

ABSTRACT

Treatment options for degenerative cerebellar ataxias are currently very limited. A large fraction of such disorders is represented by hereditary cerebellar ataxias, whose familiar transmission facilitates an early diagnosis and may possibly allow to start preventive treatments before the onset of the neurodegeneration and appearance of first symptoms. In spite of the heterogeneous aetiology, histological alterations of ataxias often include the primary degeneration of the cerebellar cortex caused by Purkinje cells (PCs) loss. Thus, approaches aimed at replacing or preserving PCs could represent promising ways of disease management. In the present study, we compared the efficacy of two different preventive strategies, namely cell replacement and motor training. We used tambaleante (tbl) mice as a model for progressive ataxia caused by selective loss of PCs and evaluated the effectiveness of the preventive transplantation of healthy PCs into early postnatal tbl cerebella, in terms of PC replacement and functional preservation. On the other hand, we investigated the effects of motor training on PC survival, cerebellar circuitry and their behavioral correlates. Our results demonstrate that, despite a good survival rate and integration of grafted PCs, the adopted grafting protocol could not alleviate the ataxic symptoms in tbl mice. Conversely, preventive motor training increases PCs survival with a moderate positive impact on the motor phenotype.


Subject(s)
Autophagy , Cerebellar Ataxia/pathology , Cerebellar Ataxia/prevention & control , Exercise Therapy , Neural Stem Cells/transplantation , Purkinje Cells/transplantation , Animals , Autophagy/physiology , Cell Survival , Cerebellar Ataxia/physiopathology , Cerebellum/pathology , Cerebellum/physiopathology , Cerebellum/surgery , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice, Transgenic , Motor Activity/physiology , Neural Stem Cells/pathology , Neural Stem Cells/physiology , Neuroprotection , Purkinje Cells/pathology , Purkinje Cells/physiology , Synapses/pathology , Synapses/physiology
6.
Neuropsychopharmacology ; 41(6): 1457-66, 2016 May.
Article in English | MEDLINE | ID: mdl-26538449

ABSTRACT

Autism spectrum disorders (ASDs) are neurodevelopmental disorders characterized by impaired social interaction, isolated areas of interest, and insistence on sameness. Mutations in Phosphatase and tensin homolog missing on chromosome 10 (PTEN) have been reported in individuals with ASDs. Recent evidence highlights a crucial role of the cerebellum in the etiopathogenesis of ASDs. In the present study we analyzed the specific contribution of cerebellar Purkinje cell (PC) PTEN loss to these disorders. Using the Cre-loxP recombination system, we generated conditional knockout mice in which PTEN inactivation was induced specifically in PCs. We investigated PC morphology and physiology as well as sociability, repetitive behavior, motor learning, and cognitive inflexibility of adult PC PTEN-mutant mice. Loss of PTEN in PCs results in autistic-like traits, including impaired sociability, repetitive behavior and deficits in motor learning. Mutant PCs appear hypertrophic and show structural abnormalities in dendrites and axons, decreased excitability, disrupted parallel fiber and climbing fiber synapses and late-onset cell death. Our results unveil new roles of PTEN in PC function and provide the first evidence of a link between the loss of PTEN in PCs and the genesis of ASD-like traits.


Subject(s)
Autistic Disorder/physiopathology , Cerebellum/physiopathology , PTEN Phosphohydrolase/physiology , Purkinje Cells/physiology , Animals , Female , Male , Mice , Mice, Knockout , Social Behavior , Spatial Learning/physiology , Stereotyped Behavior/physiology
8.
Brain Struct Funct ; 221(6): 3193-209, 2016 07.
Article in English | MEDLINE | ID: mdl-26264050

ABSTRACT

Perineuronal nets (PNNs) are aggregates of extracellular matrix molecules surrounding several types of neurons in the adult CNS, which contribute to stabilising neuronal connections. Interestingly, a reduction of PNN number and staining intensity has been observed in conditions associated with plasticity in the adult brain. However, it is not known whether spontaneous PNN changes are functional to plasticity and repair after injury. To address this issue, we investigated PNN expression in the vestibular nuclei of the adult mouse during vestibular compensation, namely the resolution of motor deficits resulting from a unilateral peripheral vestibular lesion. After unilateral labyrinthectomy, we found that PNN number and staining intensity were strongly attenuated in the lateral vestibular nucleus on both sides, in parallel with remodelling of excitatory and inhibitory afferents. Moreover, PNNs were completely restored when vestibular deficits of the mice were abated. Interestingly, in mice with genetically reduced PNNs, vestibular compensation was accelerated. Overall, these results strongly suggest that temporal tuning of PNN expression may be crucial for vestibular compensation.


Subject(s)
Axons/physiology , Extracellular Matrix/physiology , Neuronal Plasticity , Recovery of Function , Vestibular Nuclei/physiology , Animals , Axons/metabolism , Ear, Inner/injuries , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Postural Balance , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Vestibular Nuclei/metabolism
9.
Psychopharmacology (Berl) ; 232(24): 4455-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26482898

ABSTRACT

RATIONALE: Prior research has accumulated a substantial amount of evidence on the ability of cocaine to produce short- and long-lasting molecular and structural plasticity in the corticostriatal-limbic circuitry. However, traditionally, the cerebellum has not been included in the addiction circuitry, even though growing evidence supports its involvement in the behavioural changes observed after repeated drug experiences. OBJECTIVES: In the present study, we explored the ability of seven cocaine administrations to alter plasticity in the cerebellar vermis. METHODS: After six cocaine injections, one injection every 48 h, mice remained undisturbed for 1 month in their home cages. Following this withdrawal period, they received a new cocaine injection of a lower dose. Locomotion, behavioural stereotypes and several molecular and structural cerebellar parameters were evaluated. RESULTS: Cerebellar proBDNF and mature BDNF levels were both enhanced by cocaine. The high BDNF expression was associated with dendritic sprouting and increased terminal size in Purkinje neurons. Additionally, we found a reduction in extracellular matrix components that might facilitate the subsequent remodelling of Purkinje-nuclear neuron synapses. CONCLUSIONS: Although speculative, it is possible that these cocaine-dependent cerebellar changes were incubated during withdrawal and manifested by the last drug injection. Importantly, the present findings indicate that cocaine is able to promote plasticity modifications in the cerebellum of sensitised animals similar to those in the basal ganglia.


Subject(s)
Central Nervous System Stimulants/administration & dosage , Cerebellum/drug effects , Cocaine/administration & dosage , Locomotion/drug effects , Neuronal Plasticity/drug effects , Synapses/drug effects , Animals , Behavior, Addictive/metabolism , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cerebellum/metabolism , Male , Mice , Mice, Inbred BALB C , Protein Precursors/metabolism , Synapses/metabolism
10.
J Neurosci ; 35(19): 7388-402, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25972168

ABSTRACT

Cerebellar GABAergic interneurons in mouse comprise multiple subsets of morphologically and neurochemically distinct phenotypes located at strategic nodes of cerebellar local circuits. These cells are produced by common progenitors deriving from the ventricular epithelium during embryogenesis and from the prospective white matter (PWM) during postnatal development. However, it is not clear whether these progenitors are also shared by other cerebellar lineages and whether germinative sites different from the PWM originate inhibitory interneurons. Indeed, the postnatal cerebellum hosts another germinal site along the Purkinje cell layer (PCL), in which Bergmann glia are generated up to first the postnatal weeks, which was proposed to be neurogenic. Both PCL and PWM comprise precursors displaying traits of juvenile astroglia and neural stem cell markers. First, we examine the proliferative and fate potential of these niches, showing that different proliferative dynamics regulate progenitor amplification at these sites. In addition, PCL and PWM differ in the generated progeny. GABAergic interneurons are produced exclusively by PWM astroglial-like progenitors, whereas PCL precursors produce only astrocytes. Finally, through in vitro, ex vivo, and in vivo clonal analyses we provide evidence that the postnatal PWM hosts a bipotent progenitor that gives rise to both interneurons and white matter astrocytes.


Subject(s)
Cell Proliferation/physiology , Cerebellum/cytology , Gene Expression Regulation, Developmental/physiology , Interneurons/physiology , Neuroglia/physiology , Stem Cells/physiology , Actins/genetics , Actins/metabolism , Animals , Animals, Newborn , CD2 Antigens/genetics , CD2 Antigens/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Cerebellum/embryology , Cerebellum/growth & development , Embryo, Mammalian , Estrogen Antagonists/pharmacology , Excitatory Amino Acid Transporter 1/genetics , Female , GABAergic Neurons/physiology , Gene Expression Regulation, Developmental/drug effects , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tamoxifen/pharmacology , White Matter/cytology , White Matter/metabolism
11.
Cancer Discov ; 5(3): 304-15, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25572173

ABSTRACT

UNLABELLED: Gastrointestinal stromal tumor (GIST), originating from the interstitial cells of Cajal (ICC), is characterized by frequent activating mutations of the KIT receptor tyrosine kinase. Despite the clinical success of imatinib, which targets KIT, most patients with advanced GIST develop resistance and eventually die of the disease. The ETS family transcription factor ETV1 is a master regulator of the ICC lineage. Using mouse models of Kit activation and Etv1 ablation, we demonstrate that ETV1 is required for GIST initiation and proliferation in vivo, validating it as a therapeutic target. We further uncover a positive feedback circuit where MAP kinase activation downstream of KIT stabilizes the ETV1 protein, and ETV1 positively regulates KIT expression. Combined targeting of ETV1 stability by imatinib and MEK162 resulted in increased growth suppression in vitro and complete tumor regression in vivo. The combination strategy to target ETV1 may provide an effective therapeutic strategy in GIST clinical management. SIGNIFICANCE: ETV1 is a lineage-specific oncogenic transcription factor required for the growth and survival of GIST. We describe a novel strategy of targeting ETV1 protein stability by the combination of MEK and KIT inhibitors that synergistically suppress tumor growth. This strategy has the potential to change first-line therapy in GIST clinical management.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins/metabolism , Gastrointestinal Stromal Tumors/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Drug Synergism , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
12.
Addict Biol ; 20(5): 941-55, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25619460

ABSTRACT

Despite the fact that several data have supported the involvement of the cerebellum in the functional alterations observed after prolonged cocaine use, this brain structure has been traditionally ignored and excluded from the circuitry affected by addictive drugs. In the present study, we investigated the effects of a chronic cocaine treatment on molecular and structural plasticity in the cerebellum, including BDNF, D3 dopamine receptors, ΔFosB, the Glu2 AMPA receptor subunit, structural modifications in Purkinje neurons and, finally, the evaluation of perineuronal nets (PNNs) in the projection neurons of the medial nucleus, the output of the cerebellar vermis. In the current experimental conditions in which repeated cocaine treatment was followed by a 1-week withdrawal period and a new cocaine challenge, our results showed that cocaine induced a large increase in cerebellar proBDNF levels and its expression in Purkinje neurons, with the mature BDNF expression remaining unchanged. Together with this, cocaine-treated mice exhibited a substantial enhancement of D3 receptor levels. Both ΔFosB and AMPA receptor Glu2 subunit expressions were enhanced in cocaine-treated animals. Significant pruning in Purkinje dendrite arborization and reduction in the size and density of Purkinje boutons contacting deep cerebellar projection neurons accompanied cocaine-dependent increase in proBDNF. Cocaine-associated effects point to the inhibitory Purkinje function impairment, as was evidenced by lower activity in these cells. Moreover, the probability of any remodelling in Purkinje synapses appears to be decreased due to an upregulation of extracellular matrix components in the PNNs surrounding the medial nuclear neurons.


Subject(s)
Cerebellum/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Neuronal Plasticity/drug effects , Animals , Male , Mice , Mice, Inbred BALB C , Models, Animal , Neurons/drug effects
13.
Eur J Neurosci ; 39(11): 1729-41, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24689961

ABSTRACT

Following injury to the adult mammalian cochlea, hair cells cannot be spontaneously replaced. Nonetheless, the postnatal cochlea contains progenitor cells, distinguished by the expression of nestin, which are able to proliferate and form neurospheres in vitro. Such resident progenitors might be endowed with reparative potential. However, to date little is known about their behaviour in situ following hair cell injury. Using adult mice and ex vivo cochlear cultures, we sought to determine whether: (i) resident cochlear progenitors respond to kanamycin ototoxicity and compensate for it; and (ii) the reparative potential of cochlear progenitors can be stimulated by the addition of growth factors. Morphological changes of cochlear tissue, expression of nestin mRNA and protein and cell proliferation were investigated in these models. Our observations show that ototoxic injury has modest effects on nestin expression and cell proliferation. On the other hand, the addition of growth factors to the injured cochlear explants induced the appearance of nestin-positive cells in the supporting cell area of the organ of Corti. The vast majority of nestin-expressing cells, however, were not proliferating. Growth factors also had a robust stimulatory effect on axonal sprouting and the proliferative response, which was more pronounced in injured cochleae. On the whole, our findings indicate that nestin expression after kanamycin ototoxicity is related to tissue reactivity rather than activation of resident progenitors attempting to replace the lost receptors. In addition, administration of growth factors significantly enhances tissue remodelling, suggesting that cochlear repair may be promoted by the exogenous application of regeneration-promoting substances.


Subject(s)
Hair Cells, Auditory/metabolism , Hearing Loss, Sensorineural/metabolism , Nestin/metabolism , Animals , Cell Proliferation , Cells, Cultured , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/physiology , Hearing Loss, Sensorineural/chemically induced , Intercellular Signaling Peptides and Proteins/pharmacology , Kanamycin/toxicity , Mice , Mice, Inbred C57BL , Nestin/genetics , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Neurogenesis , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Mol Cell Neurosci ; 57: 10-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23999154

ABSTRACT

In the adult central nervous system (CNS) subsets of neurons are enwrapped by densely organized extracellular matrix structures, called perineuronal nets (PNNs). PNNs are formed at the end of critical periods and contribute to synapse stabilization. Enzymatic degradation of PNNs or genetic deletion of specific PNN components leads to the prolongation of the plasticity period. PNNs consist of extracellular matrix molecules, including chondroitin sulfate proteoglycans, hyaluronan, tenascins and link proteins. It has been recently shown that the chemorepulsive axon guidance protein semaphorin3A (Sema3A) is also a constituent of PNNs, binding with high affinity to the sugar chains of chondroitin sulfate proteoglycans. To elucidate whether the expression of Sema3A is modified in parallel with structural plasticity in the adult CNS, we examined Sema3A expression in the deep cerebellar nuclei of the adult mouse in a number of conditions associated with structural reorganization of the local connectivity. We found that Sema3A in PNNs is reduced during enhanced neuritic remodeling, in both physiological and injury-induced conditions. Moreover, we provide evidence that Sema3A is tightly associated with Purkinje axons and their terminals and its amount in the PNNs is related to Purkinje cell innervation of DCN neurons, but not to glutamatergic inputs. On the whole these data suggest that Sema3A may contribute to the growth-inhibitory properties of PNNs and Purkinje neurons may directly control their specific connection pattern through the release and capture of this guidance cue in the specialized ECM that surrounds their terminals.


Subject(s)
Cerebellum/metabolism , Extracellular Matrix/metabolism , Purkinje Cells/cytology , Semaphorin-3A/metabolism , Animals , Cerebellum/cytology , Mice , Purkinje Cells/metabolism , Semaphorin-3A/genetics
15.
Prog Neurobiol ; 109: 42-63, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23981535

ABSTRACT

The glial cells of the cerebellum, and particularly astrocytes and oligodendrocytes, are characterized by a remarkable phenotypic variety, in which highly peculiar morphological features are associated with specific functional features, unique among the glial cells of the entire CNS. Here, we provide a critical report about the present knowledge of the development of cerebellar glia, including lineage relationships between cerebellar neurons, astrocytes and oligodendrocytes, the origins and the genesis of the repertoire of glial types, and the processes underlying their acquisition of mature morphological and functional traits. In parallel, we describe and discuss some fundamental roles played by specific categories of glial cells during cerebellar development. In particular, we propose that Bergmann glia exerts a crucial scaffolding activity that, together with the organizing function of Purkinje cells, is necessary to achieve the normal pattern of foliation and layering of the cerebellar cortex. Moreover, we discuss some of the functional tasks of cerebellar astrocytes and oligodendrocytes that are distinctive of cerebellar glia throughout the CNS. Notably, we report about the regulation of synaptic signalling in the molecular and granular layer mediated by Bergmann glia and parenchymal astrocytes, and the functional interaction between oligodendrocyte precursor cells and neurons. On the whole, this review provides an extensive overview of the available literature and some novel insights about the origin and differentiation of the variety of cerebellar glial cells and their function in the developing and mature cerebellum.


Subject(s)
Cell Lineage/physiology , Cerebellum/cytology , Cerebellum/physiology , Neuroglia/physiology , Animals , Cell Differentiation/physiology , Humans , Purkinje Cells/physiology
16.
J Neurosci ; 33(30): 12407-22, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884946

ABSTRACT

Stem cell lines that faithfully maintain the regional identity and developmental potency of progenitors in the human brain would create new opportunities in developmental neurobiology and provide a resource for generating specialized human neurons. However, to date, neural progenitor cultures derived from the human brain have either been short-lived or exhibit restricted, predominantly glial, differentiation capacity. Pluripotent stem cells are an alternative source, but to ascertain definitively the identity and fidelity of cell types generated solely in vitro is problematic. Here, we show that hindbrain neuroepithelial stem (hbNES) cells can be derived and massively expanded from early human embryos (week 5-7, Carnegie stage 15-17). These cell lines are propagated in adherent culture in the presence of EGF and FGF2 and retain progenitor characteristics, including SOX1 expression, formation of rosette-like structures, and high neurogenic capacity. They generate GABAergic, glutamatergic and, at lower frequency, serotonergic neurons. Importantly, hbNES cells stably maintain hindbrain specification and generate upper rhombic lip derivatives on exposure to bone morphogenetic protein (BMP). When grafted into neonatal rat brain, they show potential for integration into cerebellar development and produce cerebellar granule-like cells, albeit at low frequency. hbNES cells offer a new system to study human cerebellar specification and development and to model diseases of the hindbrain. They also provide a benchmark for the production of similar long-term neuroepithelial-like stem cells (lt-NES) from pluripotent cell lines. To our knowledge, hbNES cells are the first demonstration of highly expandable neuroepithelial stem cells derived from the human embryo without genetic immortalization.


Subject(s)
Cell Proliferation/drug effects , Embryonic Stem Cells/cytology , Fetus/cytology , Neural Stem Cells/cytology , Neuroepithelial Cells/cytology , Rhombencephalon/cytology , Animals , Brain Tissue Transplantation/methods , Cell Culture Techniques/methods , Cell Differentiation , Cell Line , Cell Lineage , Cerebellum/cytology , Coculture Techniques , Epidermal Growth Factor/pharmacology , Feeder Cells , Female , Fibroblast Growth Factor 2/pharmacology , Humans , Male , Mice , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Stem Cell Transplantation/methods
17.
Neural Plast ; 2013: 854597, 2013.
Article in English | MEDLINE | ID: mdl-23864962

ABSTRACT

Stroke is a common and disabling global health-care problem, which is the third most common cause of death and one of the main causes of acquired adult disability in many countries. Rehabilitation interventions are a major component of patient care. In the last few years, brain stimulation, mirror therapy, action observation, or mental practice with motor imagery has emerged as interesting options as add-on interventions to standard physical therapies. The neural bases for poststroke recovery rely on the concept of plasticity, namely, the ability of central nervous system cells to modify their structure and function in response to external stimuli. In this review, we will discuss recent noninvasive strategies employed to enhance functional recovery in stroke patients and we will provide an overview of neural plastic events associated with rehabilitation in preclinical models of stroke.


Subject(s)
Disabled Persons/rehabilitation , Exercise Therapy/methods , Physical Therapy Modalities , Recovery of Function/physiology , Stroke Rehabilitation , Humans , Motor Skills/physiology , Neuronal Plasticity/physiology , Stroke/physiopathology
18.
Proc Natl Acad Sci U S A ; 110(11): 4374-9, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23440189

ABSTRACT

Neurons in mammals do not undergo replicative aging, and, in absence of pathologic conditions, their lifespan is limited only by the maximum lifespan of the organism. Whether neuronal lifespan is determined by the strain-specific lifetime or can be extended beyond this limit is unknown. Here, we transplanted embryonic mouse cerebellar precursors into the developing brain of the longer-living Wistar rats. The donor cells integrated into the rat cerebellum developing into mature neurons while retaining mouse-specific morphometric traits. In their new environment, the grafted mouse neurons did not die at or before the maximum lifespan of their strain of origin but survived as long as 36 mo, doubling the average lifespan of the donor mice. Thus, the lifespan of neurons is not limited by the maximum lifespan of the donor organism, but continues when transplanted in a longer-living host.


Subject(s)
Cellular Senescence/physiology , Cerebellum/metabolism , Embryo, Mammalian/metabolism , Neurons/metabolism , Animals , Cells, Cultured , Cerebellum/cytology , Embryo, Mammalian/cytology , Longevity/physiology , Mice , Mice, Transgenic , Neurons/cytology , Neurons/transplantation , Rats , Rats, Wistar , Species Specificity
19.
Stem Cells Dev ; 22(4): 538-47, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23009360

ABSTRACT

Neural stem cells (NSCs) have become promising tools for basic research and regenerative medicine. Intracerebral transplantation studies have suggested that these cells may be able to adopt neuronal phenotypes typical of their engraftment site and to establish appropriate connections in the recipient circuitries. Here, we examined the in vivo neurogenic competence of well-characterized NSC lines subjected to in vitro priming and subsequent implantation into the adult intact mouse brain. Upon implantation into the hippocampus and, less frequently, in the striatum and in the cerebral cortex, numerous green fluorescent protein (GFP)-tagged cells acquired differentiated features indistinguishable from resident neurons. Upon closer examination, however, we found that this outcome resulted from fusion of donor cells with local neuronal elements generating long-term persistent GFP(+) neuronal hybrids. This fusogenic behavior of NSCs was unexpected and also observed in coculture with E18 hippocampal immature neural cells, but not with microglia or astrocytes. Similar findings were consistently obtained with different NSC lines, mouse recipients, and donor cell-labeling methods. The frequent and cell type-specific fusion of donor NSCs with host neurons highlights a previously underestimated biological property of the nervous tissue that might prove profitable for basic and therapeutically oriented studies.


Subject(s)
Brain , Neural Stem Cells , Neurons , Animals , Brain/cytology , Brain/metabolism , Cell Fusion , Mice , Mice, Nude , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Neurons/cytology , Neurons/metabolism , Transplantation, Homologous
20.
J Neurosci ; 32(49): 17788-99, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23223298

ABSTRACT

In the adult mammalian subventricular zone (SVZ), GFAP-positive neural stem cells (NSCs) generate neuroblasts that migrate tangentially along the rostral migratory stream (RMS) toward the olfactory bulb (OB). In the mouse brain, we found that the plasticity inhibitors Nogo-A and Nogo receptor 1 (NgR1) are differentially expressed in the SVZ-OB system, in which Nogo-A identifies immature neuroblasts and NgR1 germinal astrocytes. We therefore examined the role of Nogo-A and NgR1 in the regulation of neurogenesis. Pharmacological experiments show that Nogo-66/NgR1 interaction reduces the proliferation of NSCs. This is consistent with a negative-feedback loop, in which newly generated neurons modulate cell division of SVZ stem cells. Moreover, the Nogo-A-Δ20 domain promotes neuroblast migration toward the OB through activation of the Rho/ROCK (Rho-associated, coiled-coil containing protein kinase) pathway, without the participation of NgR1. Our findings reveal a new unprecedented function for Nogo-A and NgR1 in the homeostatic regulation of the pace of neurogenesis in the adult mouse SVZ and in the migration of neuroblasts along the RMS.


Subject(s)
Cell Movement/physiology , Homeostasis/physiology , Myelin Proteins/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Receptors, Cell Surface/physiology , Animals , Astrocytes/metabolism , Brain/metabolism , Brain/physiology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/physiology , Mice , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/biosynthesis , Neural Stem Cells/metabolism , Nogo Proteins , Nogo Receptor 1 , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/biosynthesis , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...