Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(22): e2400648121, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38781210

RESUMO

After central nervous system injury, a rapid cellular and molecular response is induced. This response can be both beneficial and detrimental to neuronal survival in the first few days and increases the risk for neurodegeneration if persistent. Semaphorin4B (Sema4B), a transmembrane protein primarily expressed by cortical astrocytes, has been shown to play a role in neuronal cell death following injury. Our study shows that after cortical stab wound injury, cytokine expression is attenuated in Sema4B-/- mice, and microglia/macrophage reactivity is altered. In vitro, Sema4B enhances the reactivity of microglia following injury, suggesting astrocytic Sema4B functions as a ligand. Moreover, injury-induced microglia reactivity is attenuated in the presence of Sema4B-/- astrocytes compared to Sema4B+/- astrocytes. In vitro experiments indicate that Plexin-B2 is the Sema4B receptor on microglia. Consistent with this, in microglia/macrophage-specific Plexin-B2-/- mice, similar to Sema4B-/- mice, microglial/macrophage reactivity and neuronal cell death are attenuated after cortical injury. Finally, in Sema4B/Plexin-B2 double heterozygous mice, microglial/macrophage reactivity is also reduced after injury, supporting the idea that both Sema4B and Plexin-B2 are part of the same signaling pathway. Taken together, we propose a model in which following injury, astrocytic Sema4B enhances the response of microglia/macrophages via Plexin-B2, leading to increased reactivity.


Assuntos
Astrócitos , Camundongos Knockout , Microglia , Proteínas do Tecido Nervoso , Semaforinas , Animais , Camundongos , Astrócitos/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/genética , Comunicação Celular , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Semaforinas/metabolismo , Semaforinas/genética
2.
Front Cell Neurosci ; 16: 1076281, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36531136

RESUMO

Injury to the central nervous system induces neuronal cell death and astrogliosis, an astrocyte-mediated response that has both a beneficial and detrimental impact on surrounding neuronal cells. The circumstance however, in which astrogliosis improves neuronal survival after an injury is not fully characterized. We have recently shown that Semaphorin4B (Sema4B) in the cortex is mostly expressed by astrocytes, and in its absence, astrocyte activation after an injury is altered. Here we find that in Sema4B knockout mice, neuronal cell death is reduced; as a result, more neurons survive near the injury site. Sema4B protein applied directly to neurons does not affect neuronal survival. In contrast, survival of wild-type neurons is increased when plated on glial culture isolated from the Sema4B knockout mice, as compared to Sema4B heterozygous cultures. Furthermore, this increased survival is also observed with conditioned medium collected from glial cultures of Sema4B knockout mice compared to heterozygous mice. This indicates that the increased survival is glial cell-dependent and mediated by a secreted factor(s). Together, our results imply that following injury, the lack of Sema4B expression in glial cells improves neuronal survival either as a result of reduced toxic factors, or perhaps increased survival factors under these conditions.

3.
Neurooncol Adv ; 4(1): vdac117, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35990702

RESUMO

Background: High-grade gliomas (HGG) in children have a devastating prognosis and occur in a remarkable spatiotemporal pattern. Diffuse midline gliomas (DMG), including diffuse intrinsic pontine gliomas (DIPG), typically occur in mid-childhood, while cortical HGGs are more frequent in older children and adults. The mechanisms behind this pattern are not clear. Methods: We used mouse organotypic slice cultures and glial cell cultures to test the impact of the microenvironment on human DIPG cells. Comparing the expression between brainstem and cortical microglia identified differentially expressed secreted proteins. The impact of some of these proteins on DIPGs was tested. Results: DIPGs, pediatric HGGs of brainstem origin, survive and divide more in organotypic slice cultures originating in the brainstem as compared to the cortex. Moreover, brainstem microglia are better able to support tumors of brainstem origin. A comparison between the two microglial populations revealed differentially expressed genes. One such gene, interleukin-33 (IL33), is highly expressed in the pons of young mice and its DIPG receptor is upregulated in this context. Consistent with this observation, the expression levels of IL33 and its receptor, IL1RL1, are higher in DIPG biopsies compared to low-grade cortical gliomas. Furthermore, IL33 can enhance proliferation and clonability of HGGs of brainstem origin, while blocking IL33 in brainstem organotypic slice cultures reduced the proliferation of these tumor cells. Conclusions: Crosstalk between DIPGs and the brainstem microenvironment, in particular microglia, through IL33 and other secreted factors, modulates spatiotemporal patterning of this HGG and could prove to be an important future therapeutic target.

4.
Brain ; 144(10): 3061-3077, 2021 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-33914858

RESUMO

WWOX-related epileptic encephalopathy (WOREE) syndrome caused by human germline bi-allelic mutations in WWOX is a neurodevelopmental disorder characterized by intractable epilepsy, severe developmental delay, ataxia and premature death at the age of 2-4 years. The underlying mechanisms of WWOX actions are poorly understood. In the current study, we show that specific neuronal deletion of murine Wwox produces phenotypes typical of the Wwox-null mutation leading to brain hyperexcitability, intractable epilepsy, ataxia and postnatal lethality. A significant decrease in transcript levels of genes involved in myelination was observed in mouse cortex and hippocampus. Wwox-mutant mice exhibited reduced maturation of oligodendrocytes, reduced myelinated axons and impaired axonal conductivity. Brain hyperexcitability and hypomyelination were also revealed in human brain organoids with a WWOX deletion. These findings provide cellular and molecular evidence for myelination defects and hyperexcitability in the WOREE syndrome linked to neuronal function of WWOX.


Assuntos
Epilepsia/genética , Deleção de Genes , Bainha de Mielina/genética , Neurônios/fisiologia , Oxidorredutase com Domínios WW/deficiência , Oxidorredutase com Domínios WW/genética , Animais , Encéfalo/patologia , Técnicas de Cocultura , Epilepsia/patologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Bainha de Mielina/patologia , Neurônios/patologia , Organoides , Oxidorredutase com Domínios WW/antagonistas & inibidores
5.
EBioMedicine ; 50: 274-289, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31787569

RESUMO

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that affects motor neurons (MNs). It was shown that human astrocytes with mutations in genes associated with ALS, like C9orf72 (C9) or SOD1, reduce survival of MNs. Astrocyte toxicity may be related to their dysfunction or the release of neurotoxic factors. METHODS: We used human induced pluripotent stem cell-derived astrocytes from ALS patients carrying C9orf72 mutations and non-affected donors. We utilized these cells to investigate astrocytic induced neuronal toxicity, changes in astrocyte transcription profile as well as changes in secretome profiles. FINDINGS: We report that C9-mutated astrocytes are toxic to MNs via soluble factors. The toxic effects of astrocytes are positively correlated with the length of astrocyte propagation in culture, consistent with the age-related nature of ALS. We show that C9-mutated astrocytes downregulate the secretion of several antioxidant proteins. In line with these findings, we show increased astrocytic oxidative stress and senescence. Importantly, media conditioned by C9-astrocytes increased oxidative stress in wild type MNs. INTERPRETATION: Our results suggest that dysfunction of C9-astrocytes leads to oxidative stress of themselves and MNs, which probably contributes to neurodegeneration. Our findings suggest that therapeutic strategies in familial ALS must not only target MNs but also focus on astrocytes to abrogate nervous system injury.


Assuntos
Esclerose Lateral Amiotrófica/etiologia , Esclerose Lateral Amiotrófica/metabolismo , Astrócitos/citologia , Astrócitos/metabolismo , Proteína C9orf72/genética , Células-Tronco Pluripotentes Induzidas/citologia , Mutação , Estresse Oxidativo , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Biomarcadores , Células Cultivadas , Reprogramação Celular , Senescência Celular/genética , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Ácido Glutâmico/metabolismo , Humanos , Camundongos , Neurônios Motores/metabolismo , Proteômica/métodos , Espécies Reativas de Oxigênio/metabolismo
6.
J Cell Sci ; 132(6)2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30796102

RESUMO

Genomic editing methods, such as the CRISPR/Cas9 system, are routinely used to study gene function in somatic cells. Owing to the heterogeneity of mutations, it is necessary to purify cell clones grown from high dilution to the point of colony formation, which can be a time-consuming process. Here, we tested a modified approach in which we seeded cells at high dilution, together with non-edited carrier cells. As a comparison, cells were also grown at high dilution with conditioned medium from a high-density culture. When using carrier cells or conditioned medium, the formation of cell colonies is accelerated. Additionally, clones grown with carrier cells are more similar to the parental lines in terms of their tumorigenic properties. Surprisingly, key signaling cascades are highly divergent between clones isolated from low-density cultures, even with conditioned medium, in contrast to clones isolated with carrier cells. Thus, our study uncovers a significant limitation using the common approach of isolating cell clones following genetic modifications and suggests an alternative method that mitigates the problem of heterogeneity of gene expression between clones.This article has an associated First Person interview with the first author of the paper.


Assuntos
Sistemas CRISPR-Cas/genética , Células Clonais/fisiologia , Meios de Cultivo Condicionados , Técnicas de Cultura de Células/métodos , Linhagem Celular , Ensaio de Unidades Formadoras de Colônias , Edição de Genes , Humanos , Mutação , Transdução de Sinais
7.
J Neurosci ; 38(24): 5478-5494, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29773756

RESUMO

Axon degeneration and disruption of neuromuscular junctions (NMJs) are key events in amyotrophic lateral sclerosis (ALS) pathology. Although the disease's etiology is not fully understood, it is thought to involve a non-cell-autonomous mechanism and alterations in RNA metabolism. Here, we identified reduced levels of miR126-5p in presymptomatic ALS male mice models, and an increase in its targets: axon destabilizing Type 3 Semaphorins and their coreceptor Neuropilins. Using compartmentalized in vitro cocultures, we demonstrated that myocytes expressing diverse ALS-causing mutations promote axon degeneration and NMJ dysfunction, which were inhibited by applying Neuropilin1 blocking antibody. Finally, overexpressing miR126-5p is sufficient to transiently rescue axon degeneration and NMJ disruption both in vitro and in vivo Thus, we demonstrate a novel mechanism underlying ALS pathology, in which alterations in miR126-5p facilitate a non-cell-autonomous mechanism of motor neuron degeneration in ALS.SIGNIFICANCE STATEMENT Despite some progress, currently no effective treatment is available for amyotrophic lateral sclerosis (ALS). We suggest a novel regulatory role for miR126-5p in ALS and demonstrate, for the first time, a mechanism by which alterations in miR126-5p contribute to axon degeneration and NMJ disruption observed in ALS. We show that miR126-5p is altered in ALS models and that it can modulate Sema3 and NRP protein expression. Furthermore, NRP1 elevations in motor neurons and muscle secretion of Sema3A contribute to axon degeneration and NMJ disruption in ALS. Finally, overexpressing miR126-5p is sufficient to transiently rescue NMJ disruption and axon degeneration both in vitro and in vivo.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , MicroRNAs/metabolismo , Degeneração Neural/metabolismo , Esclerose Lateral Amiotrófica/genética , Animais , Axônios/metabolismo , Axônios/patologia , Regulação para Baixo , Regulação da Expressão Gênica , Humanos , Camundongos , MicroRNAs/genética , Degeneração Neural/genética , Degeneração Neural/patologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Neuropilina-1/biossíntese , Neuropilina-1/genética , Semaforina-3A/biossíntese , Semaforina-3A/genética
8.
Cell Death Dis ; 9(3): 256, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29449528

RESUMO

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by cell death of upper and lower motor neurons (MNs). The cause of MN cell loss is not completely understood but involves both cell autonomous and non-cell autonomous mechanisms. Numerous molecules have been implicated to be involved in the death of MNs. One such candidate is semaphorin 3A (Sema3A). In ALS patients, Sema3A was shown to be significantly upregulated in the motor cortex and downregulated in the spinal cord. In the mouse, Sema3A was shown to be an axon repellent molecule for MNs. Sema3A could also induce death of different neuronal types that are also repelled by it, including sensory, sympathetic, retinal, and cortical neurons. In contrast, astrocyte-specific knockout of Sema3A results in motor neuron cell death, consistent with the idea that Sema3A is a survival factor for mouse motor neurons. Here, we tested the response of human cortical neurons and spinal cord MNs to Sema3A. We found that Sema3A enhances the survival of spinal cord MNs. In contrast, Sema3A reduces the survival of cortical neurons. Thus, both upregulation of Sema3A in the cortex, or downregulation in the spinal cord of ALS patients is likely to directly contribute to MNs cell loss in ALS patients.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Córtex Cerebral/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , Semaforina-3A/farmacologia , Nervos Espinhais/efeitos dos fármacos , Esclerose Lateral Amiotrófica/metabolismo , Morte Celular , Linhagem Celular , Sobrevivência Celular , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Humanos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Neuropilinas/metabolismo , Semaforina-3A/metabolismo , Nervos Espinhais/metabolismo , Nervos Espinhais/patologia
9.
Sci Rep ; 8(1): 93, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311693

RESUMO

Inhibition of genes is a powerful approach to study their function. While RNA interference is a widely used method to achieve this goal, mounting evidence indicates that such an approach is prone to off-target effects. An alternative approach to gene function inhibition is genetic mutation, such as the CRISPR/cas9 method. A recent report, however, demonstrated that genetic mutation and inhibition of gene expression do not always give corresponding results. This can be explained by off-target effects, but it was recently shown, at least in one case, that these differences are the result of a compensatory mechanism induced only by genetic mutation. We present here a combination of RNA inhibition and CRISPR/cas9 methods to identify possible off targets as well as potential compensatory effects. This approach is demonstrated by testing a possible role for Sema4B in glioma biology, in which our results implicate Sema4B as having a critical function. In stark contrast, by using shRNA over CRISPR/cas9 combined methodology, we clearly demonstrate that the Sema4B targeted shRNA effects on cell proliferation is the result of off-target effects. Nevertheless, it also revealed that certain splice variants of Sema4B are important for the ability of glioma cells to grow as individual clones.


Assuntos
Sistemas CRISPR-Cas , RNA Interferente Pequeno/genética , Animais , Morte Celular/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Xenoenxertos , Humanos , Camundongos , Interferência de RNA , Semaforinas/genética , Semaforinas/metabolismo
10.
Sci Rep ; 7: 43421, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28266523

RESUMO

Adenosine to Inosine (A-to-I) RNA editing is a co- or post-transcriptional mechanism that modifies genomically encoded nucleotides at the RNA level. A-to-I RNA editing is abundant in the brain, and altered editing levels have been reported in various neurological pathologies and following spinal cord injury (SCI). The prevailing concept is that the RNA editing process itself is dysregulated by brain pathologies. Here we analyzed recent RNA-seq data, and found that, except for few mammalian conserved editing sites, editing is significantly higher in neurons than in other cell populations of the brain. We studied A-to-I RNA editing in stab wound injury (SWI) and SCI models and showed that the apparent under-editing observed after injury correlates with an approximately 20% reduction in the relative density of neurons, due to cell death and immune cell infiltration that may account for the observed under-editing. Studies of neuronal and astrocyte cultures and a computational analysis of SCI RNA-seq data further supported the possibility that a reduction in neuronal density is responsible for alterations in the tissue-wide editing patterns upon injury. Thus, our data suggest that the case for a mechanistic linkage between A-to-I RNA editing and brain pathologies should be revisited.


Assuntos
Astrócitos/metabolismo , Córtex Cerebral/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Oligodendroglia/metabolismo , RNA/metabolismo , Traumatismos da Medula Espinal/metabolismo , Adenosina/genética , Adenosina/metabolismo , Animais , Astrócitos/patologia , Córtex Cerebral/lesões , Córtex Cerebral/patologia , Feminino , Inosina/genética , Inosina/metabolismo , Camundongos , Microglia/patologia , Neurônios/patologia , Oligodendroglia/patologia , Especificidade de Órgãos , Cultura Primária de Células , RNA/genética , Edição de RNA , Medula Espinal/metabolismo , Medula Espinal/patologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia
11.
J Vis Exp ; (105): e53304, 2015 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-26575352

RESUMO

Brain tumors are a major cause of cancer-related morbidity and mortality. Developing new therapeutics for these cancers is difficult, as many of these tumors are not easily grown in standard culture conditions. Neurosphere cultures under serum-free conditions and orthotopic xenografts have expanded the range of tumors that can be maintained. However, many types of brain tumors remain difficult to propagate or study. This is particularly true for pediatric brain tumors such as pilocytic astrocytomas and medulloblastomas. This protocol describes a system that allows primary human brain tumors to be grown in culture. This quantitative assay can be used to investigate the effect of microenvironment on tumor growth, and to test new drug therapies. This protocol describes a system where fluorescently labeled brain tumor cells are grown on an organotypic brain slice from a juvenile mouse. The response of tumor cells to drug treatments can be studied in this assay, by analyzing changes in the number of cells on the slice over time. In addition, this system can address the nature of the microenvironment that normally fosters growth of brain tumors. This brain tumor organotypic slice co-culture assay provides a propitious system for testing new drugs on human tumor cells within a brain microenvironment.


Assuntos
Neoplasias Encefálicas/patologia , Técnicas de Cocultura/métodos , Técnicas de Cultura de Órgãos/métodos , Animais , Astrocitoma/patologia , Corantes Fluorescentes/química , Camundongos , Microscopia de Fluorescência/métodos , Microesferas , Microambiente Tumoral
12.
eNeuro ; 2(3)2015.
Artigo em Inglês | MEDLINE | ID: mdl-26464987

RESUMO

Injury to the CNS induces astrogliosis, an astrocyte-mediated response that has both beneficial and detrimental impacts on surrounding neural and non-neural cells. The precise signaling events underlying astrogliosis are not fully characterized. Here, we show that astrocyte activation was altered and proliferation was reduced in Semaphorin 4B (Sema4B)-deficient mice following injury. Proliferation of cultured Sema4B(-/-) astrocytes was also significantly reduced. In contrast to its expected role as a ligand, the Sema4B ectodomain was not able to rescue Sema4B(-/-) astrocyte proliferation but instead acted as an antagonist against Sema4B(+/-) astrocytes. Furthermore, the effects of Sema4B on astrocyte proliferation were dependent on phosphorylation of the intracellular domain at Ser825. Our results suggest that Sema4B functions as an astrocyte receptor, defining a novel signaling pathway that regulates astrogliosis after CNS injury.

13.
Eur J Cell Biol ; 94(10): 453-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26111659

RESUMO

The biological activity of a recombinant protein is routinely measured using a bioassay such as an enzyme assay. However, many proteins have no enzymatic activity and in many cases it is difficult to devise a simple and reliable approach to test their activity. Semaphorins, Ephrins, Slits, Netrins or amylin-assisted proteins have numerous activities affecting many systems and cell types in the human body. Most of them are also able to induce rapid cytoskeleton changes at least in some cell types. We assumed therefore, that such proteins might be tested based on their ability to modulate the cytoskeleton. Here we tested a number of semaphorins in an impedance based label-free platform that allows for dynamic monitoring of subtle morphological and adhesive changes. This system has proved to be a very fast, sensitive and effective way to monitor and determine the activity of such proteins. Furthermore we showed that it is possible to customize a cell-protein system by transfecting the cells with specific receptors and test the cell response following the addition of the recombinant ligand protein. Since other protein families such as Ephrins and Netrins can also influence the cytoskeleton of some cells, this approach may be applicable to a large number of proteins.


Assuntos
Bioensaio/métodos , Proteínas Recombinantes/análise , Semaforinas/análise , Animais , Citoesqueleto , Impedância Elétrica , Humanos , Ligantes , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Transfecção
14.
PLoS One ; 8(7): e70085, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23922915

RESUMO

Axon guidance molecules determine the pattern of neuronal circuits. Accuracy of the process is ensured by unknown mechanisms that correct early guidance errors. Since the time frame of error correction in Sema3A null mice partly overlaps with the period of naturally occurring cell death in dorsal root ganglia (DRG) development, we tested the hypothesis that apoptosis of misguided neurons enables error correction. We crossed BAX null mice, in which DRG apoptosis is blocked, with Sema3A null mice to induce errors. Analyses of these double-null mouse embryos showed that the elimination of abnormal projections is not blocked in the absence of BAX. Surprisingly however, there are fewer surviving neurons in Sema3A null or Sema3A/BAX double-null newborn mice than in wild-type mice. These results suggest that guidance errors are corrected by a BAX-independent cell death mechanism. Thus, aberrant axonal guidance may lead to reductions in neuronal numbers to suboptimal levels, perhaps increasing the likelihood of neuropathological consequences later in life.


Assuntos
Apoptose , Gânglios Espinais/citologia , Gânglios Espinais/embriologia , Semaforina-3A/genética , Proteína X Associada a bcl-2/genética , Animais , Sobrevivência Celular , Deleção de Genes , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo
15.
J Neurosci ; 28(47): 12427-32, 2008 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19020035

RESUMO

Extensive neuronal cell death during development is believed to be due to a limiting supply of neurotrophic factors. In vitro studies suggest that axon guidance molecules directly regulate neuronal survival, raising the possibility that they play a direct role in neuronal cell death in vivo. However, guidance errors may also influence survival indirectly due to loss of target-derived neurotrophic support. The role of guidance molecules in neuronal death in vivo has thus been difficult to decipher. Semaphorin3A, a repulsive guidance cue for sensory neurons, can induce sensory neuron death in vitro. Null mice studies of the Semaphorin3A coreceptors showed that guidance activity is mediated by PlexinA4, but PlexinA3 partially compensates in PlexinA4(-/-) mice. Here we demonstrate that both Plexins contribute to Sema3A-induced cell death in vitro, albeit in a different hierarchy. PlexinA3 is absolutely required, while PlexinA4 makes a smaller contribution to cell death. We found that PlexinA3(-/-) mice, which, unlike PlexinA4(-/-) mice, do not exhibit sensory axon patterning defects, show reduced neuronal apoptosis and an increased number of DRG neurons. Semaphorin3A involvement in neuronal death in vivo was demonstrated by a sensitization experiment using the proapoptotic effector Bax. Our results identify Plexins as mediators of Semaphorin-induced cell death in vitro, and provide the first evidence implicating Semaphorin/Plexin signaling in neuronal survival independent of its role in axon guidance. The results also support the idea that naturally occurring neuronal cell death reflects not only competition for target-derived trophic factors, but also the action of proapoptotic signaling via a Semaphorin/Plexin pathway.


Assuntos
Apoptose/fisiologia , Gânglios Espinais/citologia , Gânglios Espinais/embriologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Contagem de Células/métodos , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos , Feminino , Proteínas de Homeodomínio/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas/métodos , Proteínas com Homeodomínio LIM , Camundongos , Camundongos Knockout , Camundongos Mutantes , Mutação , Fator de Crescimento Neural/farmacologia , Gravidez , Semaforina-3A/farmacologia , Semaforinas/genética , Estatísticas não Paramétricas , Fatores de Tempo , Fatores de Transcrição , Transfecção/métodos , Proteína X Associada a bcl-2/deficiência
16.
J Neurosci ; 27(47): 13000-11, 2007 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18032673

RESUMO

The p75 neurotrophin receptor (p75(NTR)) interacts with multiple ligands and coreceptors. It is thought to mediate myelin growth inhibition as part of the Nogo receptor complex, in addition to its other roles. Paradoxically, however, peripheral axons of p75(ExonIII-/-) mutant embryos are severely stunted. This inhibition of axon growth may be a result of neurite elongation defects in p75(NTR) mutant neurons. Here, we show that p75(ExonIII-/-) DRG neurons are hypersensitive to the repellent molecule Semaphorin3A (Sema3A). NGF modulates Sema3A activity equally well in both the p75(NTR) mutant and wild-type neurons, indicating that the hypersensitivity of p75(NTR) mutant neurons is probably not related to their NGF receptor activity. Neuropilin1 and p75(NTR) partially colocalize in DRG growth cones. After Sema3A stimulation, the degree of colocalization is dramatically increased, particularly in clusters associated with Sema3A receptor complex activation. Coimmunoprecipitation studies show that p75(NTR) interacts directly with the Sema3A receptors Neuropilin1 and PlexinA4. When coexpressed with both Neuropilin1 and PlexinA4, p75(NTR) reduces the interaction between these two receptor components. Finally, p75(NTR)/Sema3A double-mutant embryos show growth similar to that observed in Sema3A-null mice. These data indicate that p75(NTR) is an important functional modulator of Sema3A activity and that, in the absence of p75(NTR), oversensitivity to Sema3A leads to severe reduction in sensory innervation. Our results also suggest that while inhibition of p75(NTR) in CNS injury may enhance nerve regeneration resulting from the inhibition of myelin-associated protein, it may also inhibit nerve regeneration through its modulation of Sema3A.


Assuntos
Axônios/fisiologia , Padronização Corporal/fisiologia , Nervos Periféricos/fisiologia , Receptor de Fator de Crescimento Neural/fisiologia , Semaforina-3A/metabolismo , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Padronização Corporal/genética , Células COS , Células Cultivadas , Chlorocebus aethiops , Gânglios Espinais/citologia , Gânglios Espinais/embriologia , Gânglios Espinais/patologia , Gânglios Espinais/fisiologia , Camundongos , Camundongos Knockout , Camundongos Mutantes , Neurônios/citologia , Neurônios/patologia , Neurônios/fisiologia , Nervos Periféricos/citologia , Nervos Periféricos/embriologia , Nervos Periféricos/patologia , Receptor de Fator de Crescimento Neural/deficiência , Receptor de Fator de Crescimento Neural/genética , Semaforina-3A/deficiência , Semaforina-3A/genética , Semaforina-3A/fisiologia
17.
Mol Cell Neurosci ; 36(2): 222-34, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17728139

RESUMO

Axon guidance cues are critical for neuronal circuitry formation. Guidance molecules may repel or attract axons directly by effecting growth cone motility, or by impinging on neuronal polarity. In Semaphorin3A null mice, many axonal errors are detected, most prominently in DRG neurons. It has been generally assumed the repellent properties of Semaphorin3A are the cause of these erroneous axonal projections. Here we show that, in semaphorin3A-null mice, the initial trajectory of neurons in the DRG is abnormal, suggesting that Semaphorin3A may instruct neuronal polarity. In corroboration, in vitro Semaphorin3A dramatically increases neuronal polarization, as indicated by GSK3beta and Rac1 sub-cellular localization in DRG neurons. Polarization effects of Semaphorin3A are regulated by activated MAPK, as indicated by p-MAPK 42/44 polarization and the need for its activity for Rac1 and GSK3beta polarization. Taken together, our findings suggest that Semaphorin3A plays a role in the formation of neuronal polarity, in addition to its classic repellent role.


Assuntos
Polaridade Celular/fisiologia , Gânglios Espinais/citologia , Neurônios/fisiologia , Semaforina-3A/fisiologia , Animais , Axônios/fisiologia , Catequina/análogos & derivados , Catequina/farmacologia , Movimento Celular/genética , Polaridade Celular/efeitos dos fármacos , Células Cultivadas , Distribuição de Qui-Quadrado , Dendritos/fisiologia , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Neurônios/citologia , Neurônios/efeitos dos fármacos , Gravidez , Semaforina-3A/deficiência , Semaforina-3A/farmacologia , Fatores de Tempo , Tubulina (Proteína)/metabolismo
18.
J Neurochem ; 96(2): 585-97, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16336628

RESUMO

Large numbers of neurons are eliminated by apoptosis during nervous system development. For instance, in the mouse dorsal root ganglion (DRG), the highest incidence of cell death occurs between embryonic days 12 and 14 (E12-E14). While the cause of cell death and its biological significance in the nervous system is not entirely understood, it is generally believed that limiting quantities of neurotrophins are responsible for neuronal death. Between E12 and E14, developing DRG neurons pass through tissues expressing high levels of axonal guidance molecules such as Semaphorin 3A (Sema3A) while navigating to their targets. Here, we demonstrate that Sema3A acts as a death-inducing molecule in neurotrophin-3 (NT-3)-, brain-derived neurotrophic factor (BDNF)- and nerve growth factor (NGF)-dependent E12 and E13 cultured DRG neurons. We show that Sema3A most probably induces cell death through activation of the c-Jun N-terminal kinase (JNK)/c-Jun signaling pathway, and that this cell death is blocked by a moderate increase in NGF concentration. Interestingly, increasing concentrations of other neurotrophic factors, such as NT-3 or BDNF, do not elicit similar effects. Our data suggest that the number of DRG neurons is determined by a fine balance between neurotrophins and Semaphorin 3A, and not only by neurotrophin levels.


Assuntos
Apoptose/fisiologia , Gânglios Espinais/embriologia , Fatores de Crescimento Neural/fisiologia , Neurônios/fisiologia , Semaforina-3A/fisiologia , Transdução de Sinais , Animais , Antracenos/farmacologia , Apoptose/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/administração & dosagem , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Éxons , Gânglios Espinais/citologia , Cones de Crescimento/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Fator de Crescimento Neural/administração & dosagem , Fator de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/farmacologia , Neurotrofina 3/administração & dosagem , Neurotrofina 3/farmacologia , Receptor de Fator de Crescimento Neural/deficiência , Receptor de Fator de Crescimento Neural/genética , Semaforina-3A/administração & dosagem , Semaforina-3A/farmacologia
19.
Development ; 132(2): 323-34, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15604101

RESUMO

During development, trigeminal nerve fibers navigate and establish their axonal projections to the developing tooth in a highly spatiotemporally controlled manner. By analyzing Sema3a and its receptor Npn1 knockout mouse embryos, we found that Sema3a regulates dental trigeminal axon navigation and patterning, as well as the timing of the first mandibular molar innervation, and that the effects of Sema3a appear to be mediated by Npn1 present in the axons. By performing tissue recombinant experiments and analyzing the effects of signaling molecules, we found that early oral and dental epithelia, which instruct tooth formation, and epithelial Wnt4 induce Sema3a expression in the presumptive dental mesenchyme before the arrival of the first dental nerve fibers. Later, at the bud stage, epithelial Wnt4 and Tgfbeta1 regulate Sema3a expression in the dental mesenchyme. In addition, Wnt4 stimulates mesenchymal expression of Msx1 transcription factor, which is essential for tooth formation, and Tgfbeta1 proliferation of the dental mesenchymal cells. Thus, epithelial-mesenchymal interactions control Sema3a expression and may coordinate axon navigation and patterning with tooth formation. Moreover, our results suggest that the odontogenic epithelium possesses the instructive information to control the formation of tooth nerve supply.


Assuntos
Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Semaforina-3A/biossíntese , Dente/embriologia , Dente/inervação , Nervo Trigêmeo/fisiologia , Animais , Axônios/metabolismo , Padronização Corporal , Proliferação de Células , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Imageamento Tridimensional , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Fator de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/biossíntese , Ratos , Fatores de Tempo , Distribuição Tecidual , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1 , Proteínas Wnt , Proteína Wnt4
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...