Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
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
2.
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
3.
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
4.
Development ; 139(13): 2308-20, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22669821

ABSTRACT

By serving as the sole output of the cerebellar cortex, integrating a myriad of afferent stimuli, Purkinje cells (PCs) constitute the principal neuron in cerebellar circuits. Several neurodegenerative cerebellar ataxias feature a selective cell-autonomous loss of PCs, warranting the development of regenerative strategies. To date, very little is known as to the regulatory cascades controlling PC development. During central nervous system development, the proneural gene neurogenin 2 (Neurog2) contributes to many distinct neuronal types by specifying their fate and/or dictating development of their morphological features. By analyzing a mouse knock-in line expressing Cre recombinase under the control of Neurog2 cis-acting sequences we show that, in the cerebellar primordium, Neurog2 is expressed by cycling progenitors cell-autonomously fated to become PCs, even when transplanted heterochronically. During cerebellar development, Neurog2 is expressed in G1 phase by progenitors poised to exit the cell cycle. We demonstrate that, in the absence of Neurog2, both cell-cycle progression and neuronal output are significantly affected, leading to an overall reduction of the mature cerebellar volume. Although PC fate identity is correctly specified, the maturation of their dendritic arbor is severely affected in the absence of Neurog2, as null PCs develop stunted and poorly branched dendrites, a defect evident from the early stages of dendritogenesis. Thus, Neurog2 represents a key regulator of PC development and maturation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Cycle , Cerebellum/growth & development , Dendrites/physiology , Nerve Tissue Proteins/physiology , Purkinje Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage , Cerebellum/physiology , Female , Gene Knock-In Techniques , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neurogenesis/physiology , Pregnancy , Stem Cell Transplantation , Stem Cells/physiology
5.
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
6.
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
7.
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
8.
Development ; 138(16): 3463-72, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21771816

ABSTRACT

The progenitors of cerebellar GABAergic interneurons proliferate up to postnatal development in the prospective white matter, where they give rise to different neuronal subtypes, in defined quantities and according to precise spatiotemporal sequences. To investigate the mechanisms that regulate the specification of distinct interneuron phenotypes, we examined mice lacking the G1 phase-active cyclin D2. It has been reported that these mice show severe reduction of stellate cells, the last generated interneuron subtype. We found that loss of cyclin D2 actually impairs the whole process of interneuron genesis. In the mutant cerebella, progenitors of the prospective white matter show reduced proliferation rates and enhanced tendency to leave the cycle, whereas young postmitotic interneurons undergo severe delay of their maturation and migration. As a consequence, the progenitor pool is precociously exhausted and the number of interneurons is significantly reduced, although molecular layer interneurons are more affected than those of granular layer or deep nuclei. The characteristic inside-out sequence of interneuron placement in the cortical layers is also reversed, so that later born cells occupy deeper positions than earlier generated ones. Transplantation experiments show that the abnormalities of cyclin D2(-/-) interneurons are largely caused by cell-autonomous mechanisms. Therefore, cyclin D2 is not required for the specification of particular interneuron subtypes. Loss of this protein, however, disrupts regulatory mechanisms of cell cycle dynamics that are required to determine the numbers of interneurons of different types and impairs their rhythm of maturation and integration in the cerebellar circuitry.


Subject(s)
Brain/metabolism , Cell Cycle , Cyclin D2/metabolism , Interneurons/cytology , Interneurons/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Brain/embryology , Brain/growth & development , Cell Movement , Cyclin D2/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Mol Pain ; 8: 39, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22616849

ABSTRACT

BACKGROUND: Current tools for analgesia are often only partially successful, thus investigations of new targets for pain therapy stimulate great interest. Consequent to peripheral nerve injury, c-Jun N-terminal kinase (JNK) activity in cells of the dorsal root ganglia (DRGs) and spinal cord is involved in triggering neuropathic pain. However, the relative contribution of distinct JNK isoforms is unclear. Using knockout mice for single isoforms, and blockade of JNK activity by a peptide inhibitor, we have used behavioral tests to analyze the contribution of JNK in the development of neuropathic pain after unilateral sciatic nerve transection. In addition, immunohistochemical labelling for the growth associated protein (GAP)-43 and Calcitonin Gene Related Peptide (CGRP) in DRGs was used to relate injury related compensatory growth to altered sensory function. RESULTS: Peripheral nerve injury produced pain-related behavior on the ipsilateral hindpaw, accompanied by an increase in the percentage of GAP43-immunoreactive (IR) neurons and a decrease in the percentage of CGRP-IR neurons in the lumbar DRGs. The JNK inhibitor, D-JNKI-1, successfully modulated the effects of the sciatic nerve transection. The onset of neuropathic pain was not prevented by the deletion of a single JNK isoform, leading us to conclude that all JNK isoforms collectively contribute to maintain neuropathy. Autotomy behavior, typically induced by sciatic nerve axotomy, was absent in both the JNK1 and JNK3 knockout mice. CONCLUSIONS: JNK signaling plays an important role in regulating pain threshold: the inhibition of all of the JNK isoforms prevents the onset of neuropathic pain, while the deletion of a single splice JNK isoform mitigates established sensory abnormalities. JNK inactivation also has an effect on axonal sprouting following peripheral nerve injury.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Neuralgia/metabolism , Sciatic Nerve/injuries , Animals , GAP-43 Protein/metabolism , Ganglia, Spinal/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Mice , Mice, Knockout , Neurons/metabolism , Peptides/pharmacology , Sciatic Nerve/metabolism
15.
Cell Tissue Res ; 349(1): 161-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22143260

ABSTRACT

During developmental critical periods, external stimuli are crucial for information processing, acquisition of new functions or functional recovery after CNS damage. These phenomena depend on the capability of neurons to modify their functional properties and/or their connections, generally defined as "plasticity". Although plasticity decreases after the closure of critical periods, the adult CNS retains significant capabilities for structural remodelling and functional adaptation. At the molecular level, structural modifications of neural circuits depend on the balance between intrinsic growth properties of the involved neurons and growth-regulatory cues of the extracellular milieu. Interestingly, experience acts on this balance, so as to create permissive conditions for neuritic remodelling. Here, we present an overview of recent findings concerning the effects of experience on cellular and molecular processes responsible for producing structural plasticity of neural networks or functional recovery after an insult to the adult CNS (e.g. traumatic injury, ischemia or neurodegenerative disease). Understanding experience-dependent mechanisms is crucial for the development of tailored rehabilitative strategies, which can be exploited alone or in combination with specific therapeutic interventions to improve neural repair after damage.


Subject(s)
Aging/pathology , Central Nervous System/physiopathology , Environment , Neuronal Plasticity/physiology , Wound Healing , Central Nervous System/surgery , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/rehabilitation , Central Nervous System Diseases/surgery , Humans
16.
Cerebellum ; 11(2): 434-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22090364

ABSTRACT

Cerebellar GABAergic projection neurons and interneurons originate from the ventricular neuroepithelium of the cerebellar primordium. However, while projection neurons are born within this germinal layer, interneurons derive from progenitors that delaminate into the prospective white matter. In spite of this common origin, the two main classes of GABAergic neurons are generated according to distinct strategies. Projection neurons are committed to their fate at early ontogenetic stages and acquire their mature phenotypes through cell-autonomous mechanisms. On the contrary, the different categories of cerebellar interneurons derive from a single pool of multipotent progenitors, whose fate choices, production rates and differentiation schedules are strongly influenced by environmental cues.


Subject(s)
Cell Differentiation/physiology , Cerebellum/cytology , Neurons/physiology , gamma-Aminobutyric Acid/physiology , Animals , Cell Count , Cell Size , Cerebellum/growth & development , Cerebral Ventricles/cytology , Cerebral Ventricles/growth & development , Cerebral Ventricles/physiology , Humans , Interneurons/physiology , Neural Stem Cells/physiology , PAX2 Transcription Factor/genetics
17.
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
18.
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
19.
Neurobiol Dis ; 41(3): 640-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21111821

ABSTRACT

Recent evidence suggests that adult bone marrow stem cells reduce tissue damage and promote repair following CNS ischemic injury. Since granulocyte-colony stimulating factor (G-CSF) mobilizes hematopoietic stem cells to the circulating compartment, here we tested whether administration of this drug modifies the outcome of a permanent occlusion of the middle cerebral artery in adult mice. To elucidate the behavior and fate of blood-borne cells in the ischemic brain, we produced chimeric animals, in which hematopoietic derivatives are genetically tagged. G-CSF administration enhances the proliferation of microglia in the uninjured CNS but has no effect on the amount of hematopoietic cells that infiltrate the ischemic tissue and on the size of the lesion. The blood-borne elements acquire different mesodermal identities but fail to adopt neural phenotypes, even though they occasionally fuse with Purkinje neurons. These results indicate that G-CSF treatment does not exert a significant beneficial effect on the ischemic injury.


Subject(s)
Brain Ischemia/therapy , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/physiology , Infarction, Middle Cerebral Artery/pathology , Microglia/pathology , Microglia/physiology , Age Factors , Animals , Brain Ischemia/pathology , Cell Proliferation , Humans , Infarction, Middle Cerebral Artery/therapy , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/cytology , Radiation Chimera , Treatment Outcome
20.
J Neurosci ; 29(21): 7079-91, 2009 May 27.
Article in English | MEDLINE | ID: mdl-19474334

ABSTRACT

In most CNS regions, the variety of inhibitory interneurons originates from separate pools of progenitors residing in discrete germinal domains, where they become committed to specific phenotypes and positions during their last mitosis. We show here that GABAergic interneurons of the rodent cerebellum are generated through a different mechanism. Progenitors for these interneurons delaminate from the ventricular neuroepithelium of the embryonic cerebellar primordium and continue to proliferate in the prospective white matter during late embryonic and postnatal development. Young postmitotic interneurons do not migrate immediately to their final destination, but remain in the prospective white matter for several days. The different interneuron categories are produced according to a continuous inside-out positional sequence, and cell identity and laminar placement in the cerebellar cortex are temporally related to birth date. However, terminal commitment does not occur while precursors are still proliferating, and postmitotic cells heterochronically transplanted to developing cerebella consistently adopt host-specific phenotypes and positions. However, solid grafts of prospective white matter implanted into the adult cerebellum, when interneuron genesis has ceased, produce interneuron types characteristic of the donor age. Therefore, specification of cerebellar GABAergic interneurons occurs through a hitherto unknown process, in which postmitotic neurons maintain broad developmental potentialities and their phenotypic choices are dictated by instructive cues provided by the microenvironment of the prospective white matter. Whereas in most CNS regions the repertoire of inhibitory interneurons is produced by recruiting precursors from different origins, in the cerebellum it is achieved by creating phenotypic diversity from a single source.


Subject(s)
Cerebellum/cytology , Interneurons/physiology , Phenotype , gamma-Aminobutyric Acid/metabolism , Actins/genetics , Age Factors , Animals , Animals, Newborn , Bromodeoxyuridine , Cell Count , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement , Cell Proliferation , Cerebellum/embryology , Cerebellum/growth & development , Embryo, Mammalian , Flow Cytometry , Gene Expression Regulation, Developmental/genetics , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Interneurons/classification , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Pathways/metabolism , PAX2 Transcription Factor/genetics , Rats , Rats, Wistar , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL