Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Neurochem ; 161(6): 463-477, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35536742

RESUMO

In the central nervous system, most neurons co-express TrkB and TrkC, the tyrosine kinase receptors for brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT3). As NT3 can also activate TrkB, it has been difficult to understand how NT3 and TrkC can exert unique roles in the assembly of neuronal circuits. Using neurons differentiated from human embryonic stem cells expressing both TrkB and TrkC, we compared Trk activation by BDNF and NT3. To avoid the complications resulting from TrkB activation by NT3, we also generated neurons from stem cells engineered to lack TrkB. We found that NT3 activates TrkC at concentrations lower than those of BDNF needed to activate TrkB. Downstream of Trk activation, the changes in gene expression caused by TrkC activation were found to be similar to those resulting from TrkB activation by BDNF, including a number of genes involved in synaptic plasticity. At high NT3 concentrations, receptor selectivity was lost as a result of TrkB activation. In addition, TrkC was down-regulated, as was also the case with TrkB at high BDNF concentrations. By contrast, receptor selectivity as well as reactivation were preserved when neurons were exposed to low neurotrophin concentrations. These results indicate that the selectivity of NT3/TrkC signalling can be explained by the ability of NT3 to activate TrkC at concentrations lower than those needed to activate TrkB. They also suggest that in a therapeutic perspective, the dosage of Trk receptor agonists will need to be taken into account if prolonged receptor activation is to be achieved.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Glicoproteínas de Membrana/metabolismo , Receptor trkB/metabolismo , Receptor trkC/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Regulação para Baixo , Humanos , Neurônios/metabolismo , Neurotrofina 3/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor trkB/genética , Receptor trkC/genética , Receptores de Fator de Crescimento Neural/genética , Receptores de Fator de Crescimento Neural/metabolismo
2.
Development ; 146(18)2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31488565

RESUMO

Multiple members of the tumour necrosis factor superfamily (TNFSF) regulate the growth and branching of neural processes late in development, when neurons are establishing and refining connections. Here, we present the first evidence that a TNFSF member acts much earlier in development, when axons are growing to their targets. CD40L transiently enhanced axon growth from embryonic mouse DRG neurons cultured at this early stage. Early spinal nerves of embryos lacking the CD40L receptor (Cd40-/- mice) were significantly shorter in vivo than those of Cd40+/+ littermates. CD40L was synthesized in early DRG targets and was co-expressed with CD40 in early DRG neurons. Whereas CD40L enhanced early axon growth independently of neurotrophins, disruption of a CD40L/CD40 autocrine loop impaired early neurotrophin-promoted axon growth. In marked contrast to the widespread regulation of axon and dendrite growth by CD40L reverse signalling later in development, CD40-Fc, which activates reverse signalling, had no effect on early sensory axon growth. These results suggest that CD40 forward signalling is a novel physiological regulator of early axon growth that acts by target-derived and autocrine mechanisms.


Assuntos
Axônios/metabolismo , Antígenos CD40/metabolismo , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais , Animais , Comunicação Autócrina , Ligante de CD40/genética , Ligante de CD40/metabolismo , Sobrevivência Celular , Embrião de Mamíferos/metabolismo , Gânglios Espinais/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Receptoras Sensoriais/citologia , Nervos Espinhais/metabolismo
3.
Mol Cell Neurosci ; 115: 103642, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34119632

RESUMO

Epigenetic modifications in neurodegenerative disease are under investigation for their roles in disease progression. Alterations in acetylation rates of certain Parkinson's disease (PD)-linked genes have been associated with the pathological progression of this disorder. In light of this, and given the lack of disease-modifying therapies for PD, HDAC inhibitors (HDIs) are under consideration as potential pharmacological agents. The neuroprotective effects of pan-HDACs and some class-specific inhibitors have been tested in in vivo and in vitro models of PD, with varying outcomes. Here we used gene co-expression analysis to identify HDACs that are associated with human dopaminergic (DA) neuron development. We identified HDAC3, HDAC5, HDAC6 and HDAC9 as being highly correlated with the DA markers, SLC6A3 and NR4A2. RT-qPCR revealed that mRNA expression of these HDACs exhibited similar temporal profiles during embryonic mouse midbrain DA (mDA) neuron development. We tested the neuroprotective potential of a number of class-specific small molecule HDIs on human SH-SY5Y cells, using neurite growth as a phenotypic readout of neurotrophic action. Neither the class I-specific HDIs, RGFP109 and RGFP966, nor the HDAC6 inhibitor ACY1215, had significant effects on neurite outgrowth. However, the class IIa HDI, LMK235 (a HDAC4/5 inhibitor), significantly increased histone acetylation and neurite outgrowth. We found that LMK235 increased BMP-Smad-dependent transcription in SH-SY5Y cells and that this was required for its neurite growth-promoting effects on SH-SY5Y cells and on DA neurons in primary cultures of embryonic day (E) 14 rat ventral mesencephalon (VM). These effects were also seen in SH-SY5Y cells transfected with HDAC5 siRNA. Furthermore, LMK235 treatment exerted neuroprotective effects against degeneration induced by the DA neurotoxin 1-methyl-4-phenylpyridinium (MPP+), in both SH-SY5Y cells and cultured DA neurons. Treatment with LMK235 was also neuroprotective against axonal degeneration induced by overexpression of wild-type (WT) or A53T mutant α-synuclein in both SH-SY5Y cells and primary cultures of DA neurons. In summary, these data show the neuroprotective potential of the class IIa HDI, LMK235, in cell models of relevance to PD.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Animais , Neurônios Dopaminérgicos , Histona Desacetilases , Camundongos , Neurotoxinas/farmacologia , Doença de Parkinson/tratamento farmacológico , Ratos , alfa-Sinucleína/genética
4.
Development ; 145(22)2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30337376

RESUMO

TWE-PRIL is a naturally occurring fusion protein of components of two TNF superfamily members: the extracellular domain of APRIL; and the intracellular and transmembrane domains of TWEAK with no known function. Here, we show that April-/- mice (which lack APRIL and TWE-PRIL) exhibited overgrowth of sympathetic fibres in vivo, and sympathetic neurons cultured from these mice had significantly longer axons than neurons cultured from wild-type littermates. Enhanced axon growth from sympathetic neurons cultured from April-/- mice was prevented by expressing full-length TWE-PRIL in these neurons but not by treating them with soluble APRIL. Soluble APRIL, however, enhanced axon growth from the sympathetic neurons of wild-type mice. siRNA knockdown of TWE-PRIL but not siRNA knockdown of APRIL alone also enhanced axon growth from wild-type sympathetic neurons. Our work reveals the first and physiologically relevant role for TWE-PRIL and suggests that it mediates reverse signalling.


Assuntos
Axônios/metabolismo , Transdução de Sinais , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Antígeno de Maturação de Linfócitos B/genética , Antígeno de Maturação de Linfócitos B/metabolismo , Células Cultivadas , Citocina TWEAK/genética , Citocina TWEAK/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Modelos Biológicos , Fator de Crescimento Neural/farmacologia , Fenótipo , RNA Interferente Pequeno/metabolismo , Solubilidade , Gânglio Cervical Superior/metabolismo , Sistema Nervoso Simpático/crescimento & desenvolvimento , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
6.
Development ; 140(10): 2108-17, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23633509

RESUMO

The somatosensory and sympathetic innervation of the vertebrate head is derived principally from the neurons of trigeminal and superior cervical ganglia (SCG), respectively. During development, the survival of both populations of neurons and the terminal growth and branching of their axons in the tissues they innervate is regulated by the supply of nerve growth factor (NGF) produced by these tissues. NGF is derived by proteolytic cleavage of a large precursor protein, proNGF, which is recognised to possess distinctive biological functions. Here, we show that proNGF promotes profuse neurite growth and branching from cultured postnatal mouse SCG neurons. In marked contrast, proNGF does not promote the growth of trigeminal neurites. Studies using compartment cultures demonstrated that proNGF acts locally on SCG neurites to promote growth. The neurite growth-promoting effect of proNGF is not observed in SCG neurons cultured from p75(NTR)-deficient mice, and proNGF does not phosphorylate the NGF receptor tyrosine kinase TrkA. These findings suggest that proNGF selectively promotes the growth of neurites from a subset of NGF-responsive neurons by a p75(NTR)-dependent mechanism during postnatal development when the axons of these neurons are ramifying within their targets in vivo.


Assuntos
Fator de Crescimento Neural/metabolismo , Neuritos/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Fatores de Crescimento Neural/metabolismo , Neurônios/citologia , Neurônios/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor trkA/metabolismo , Transdução de Sinais , Fatores de Tempo , Nervo Trigêmeo/metabolismo
7.
Development ; 140(23): 4751-62, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24173804

RESUMO

Dendrite size and morphology are key determinants of the functional properties of neurons. Here, we show that growth differentiation factor 5 (GDF5), a member of the bone morphogenetic protein (BMP) subclass of the transforming growth factor ß superfamily with a well-characterised role in limb morphogenesis, is a key regulator of the growth and elaboration of pyramidal cell dendrites in the developing hippocampus. Pyramidal cells co-express GDF5 and its preferred receptors, BMP receptor 1B and BMP receptor 2, during development. In culture, GDF5 substantially increased dendrite, but not axon, elongation from these neurons by a mechanism that depends on activation of SMADs 1/5/8 and upregulation of the transcription factor HES5. In vivo, the apical and basal dendritic arbours of pyramidal cells throughout the hippocampus were markedly stunted in both homozygous and heterozygous Gdf5 null mutants, indicating that dendrite size and complexity are exquisitely sensitive to the level of endogenous GDF5 synthesis.


Assuntos
Dendritos/metabolismo , Fator 5 de Diferenciação de Crescimento/metabolismo , Hipocampo/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Células Cultivadas , Ativação Enzimática , Regulação da Expressão Gênica no Desenvolvimento , Fator 5 de Diferenciação de Crescimento/biossíntese , Fator 5 de Diferenciação de Crescimento/genética , Hipocampo/embriologia , Hipocampo/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Piramidais/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Regulação para Cima
8.
Mol Cell Neurosci ; 59: 24-36, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24444792

RESUMO

APRIL (A Proliferation-Inducing Ligand, TNFSF13) is a member of the tumor necrosis factor superfamily that regulates lymphocyte survival and activation and has been implicated in tumorigenesis and autoimmune diseases. Here we report the expression and first known activity of APRIL in the nervous system. APRIL and one of its receptors, BCMA (B-Cell Maturation Antigen, TNFRSF17), are expressed by hippocampal pyramidal cells of fetal and postnatal mice. In culture, these neurons secreted APRIL, and function-blocking antibodies to either APRIL or BCMA reduced axonal elongation. Recombinant APRIL enhanced axonal elongation, but did not influence dendrite elongation. The effect of APRIL on axon elongation was inhibited by anti-BCMA and the expression of a signaling-defective BCMA mutant in these neurons, suggesting that the axon growth-promoting effect of APRIL is mediated by BCMA. APRIL promoted phosphorylation and activation of ERK1, ERK2 and Akt and serine phosphorylation and inactivation of GSK-3ß in cultured hippocampal pyramidal cells. Inhibition of MEK1/MEK2 (activators of ERK1/ERK2), PI3-kinase (activator of Akt) or Akt inhibited the axon growth-promoting action of APRIL, as did pharmacological activation of GSK-3ß and the expression of a constitutively active form of GSK-3ß. These findings suggest that APRIL promotes axon elongation by a mechanism that depends both on ERK signaling and PI3-kinase/Akt/GSK-3ß signaling.


Assuntos
Axônios/metabolismo , Hipocampo/metabolismo , Neurogênese , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Antígeno de Maturação de Linfócitos B/metabolismo , Células Cultivadas , Dendritos/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Transdução de Sinais , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
9.
Differentiation ; 88(4-5): 124-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25582843

RESUMO

During development, the growth of neural processes is regulated by an array of cellular and molecular mechanisms which influence growth rate, direction and branching. Recently, many members of the TNF superfamily have been shown to be key regulators of neurite growth during development. The founder member of this family, TNFα can both promote and inhibit neurite growth depending on the cellular context. Specifically, transmembrane TNFα promotes neurite growth, while soluble TNFα inhibits it. While the growth promoting effects of TNFα are restricted to a defined developmental window of early postnatal development, whether the growth inhibitory effects of soluble TNFα occur throughout development is unknown. In this study we used the extensively studied, well characterised neurons of the superior cervical ganglion to show that the growth inhibitory effects of soluble TNFα are restricted to a specific period of late embryonic and early postnatal development. Furthermore, we show that this growth inhibitory effect of soluble TNFα requires NF-κB signalling at all developmental stages at which soluble TNFα inhibits neurite growth. These findings raise the possibility that increases in the amount of soluble TNFα in vivo, for example as a result of maternal inflammation, could negatively affect neurite growth in developing neurons at specific stages of development.


Assuntos
Neuritos/efeitos dos fármacos , Neurogênese , Sistema Nervoso Simpático/citologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Neuritos/metabolismo , Neuritos/fisiologia , Ratos , Ratos Sprague-Dawley , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/crescimento & desenvolvimento
10.
Int J Neurosci ; 125(1): 70-7, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24628580

RESUMO

It is well established that neuroinflammation is associated with the progression of many neurodegenerative diseases, including Parkinson's disease (PD). Activated microglia and elevated levels of pro-inflammatory cytokines such as interleukin-1ß (IL-1ß) have been found in the brain and cerebrospinal fluid of PD patients, suggesting that IL-1ß may be involved in the pathogenesis of this disease. This study aimed to knock down the expression of the interleukin-1 type 1 receptor (IL-1R1) to evaluate any potential therapeutic effect of limiting the action of IL-1ß in the substantia nigra following a unilateral intrastriatal 6-hydroxydopamine (6-OHDA) lesion in rats. Adult Sprague-Dawley rats received intranigral injections of shRNA specific for IL-1R1, followed 2 weeks later by intrastriatal 6-OHDA. Injection of IL-1R1 shRNA did not prevent 6-OHDA-induced loss of motor function or loss of nigral dopamine neurons. IL-1R1 expression was increased in the midbrain following 6-OHDA injection; this effect was attenuated in 6-OHDA-treated animals that had received IL-1R1 shRNA. These data suggest that while IL-1R1 was increased in 6-OHDA-treated animals and reduced following shRNA injection, the neurodegeneration induced by 6-OHDA was not mediated through IL-1R1.


Assuntos
Corpo Estriado/fisiologia , Doença de Parkinson/patologia , Doença de Parkinson/prevenção & controle , Receptores Tipo I de Interleucina-1/metabolismo , Adrenérgicos/toxicidade , Anfetaminas , Análise de Variância , Animais , Corpo Estriado/efeitos dos fármacos , Modelos Animais de Doenças , Masculino , Oxidopamina/toxicidade , Doença de Parkinson/etiologia , RNA Interferente Pequeno/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores Tipo I de Interleucina-1/genética , Comportamento Estereotipado/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/metabolismo
11.
Anat Histol Embryol ; 51(3): 347-354, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35170797

RESUMO

Anomalies in the subclavian and common carotid arteries can be of interest in cases of cranial mediastinal surgeries, as well as to diagnose the cause of oesophageal constrictions leading to clinical signs of dysphagia (dysphagia lusoria). The development and regression of the aortic arches are of key importance in understanding the origin of these type of vascular anomalies. This report describes the congenital anomalous aortic origin of the common carotid and the subclavian arteries in a 14-year-old dog and the plausible developmental pattern failure. Academic dissection revealed a common bicarotid trunk and bisubclavian trunk arising from the most cranial aspect of the aortic arch. Despite the abnormal origin, these vessels displayed a predominantly standard anatomical course. All the anticipated branches were identified and described. Cardiac abnormalities were also noted including right atrial dilation, coronary sinus enlargement, right and left valvular endocardiosis, a patent foramen ovale and marked concentric left ventricular hypertrophy with compensatory left atrial dilation. Additionally, the right recurrent laryngeal nerve demonstrated an aberrant course consistent with a 'non-recurrent laryngeal nerve' (non-RLN). Awareness of the anatomical variations of the aortic arch is important for surgical interventions of the cranial mediastinum as well as radiological interpretation. Although infrequent, the variants similar to the one described here have been reported in different species.


Assuntos
Transtornos de Deglutição , Doenças do Cão , Cardiopatias Congênitas , Animais , Aorta Torácica/anormalidades , Aorta Torácica/diagnóstico por imagem , Artéria Carótida Primitiva , Transtornos de Deglutição/veterinária , Cães , Cardiopatias Congênitas/veterinária , Artéria Subclávia/anormalidades
12.
Dev Neurobiol ; 81(2): 139-148, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33369884

RESUMO

Recent work has shown that neuregulin-4 (NRG4) is a physiological regulator of the growth of sympathetic axons and CNS dendrites in the developing nervous system. Here, we have investigated whether NRG4 plays a role in sensory axon growth and the establishment of cutaneous sensory innervation. Imaging early nerve fibers in the well-characterized cutaneous trigeminal territory, the brachial plexus, and thorax revealed very marked and highly significant decreases in nerve fiber length and branching density in Nrg4-/- embryos compared with Nrg4+/+ littermates. NRG4 promoted neurotrophin-independent sensory axon growth from correspondingly early trigeminal ganglion and DRG neurons in culture but not from enteroceptive nodose ganglion neurons. High levels of Nrg4 mRNA were detected in cutaneous tissues but not in sensory ganglia. Our findings suggest that NRG4 is an important target-derived factor that participates in the establishment of early cutaneous sensory innervation.


Assuntos
Fatores de Crescimento Neural , Neurregulinas/fisiologia , Axônios/fisiologia , Neurregulinas/química , Neurregulinas/metabolismo , Neurônios/fisiologia , Neurônios Aferentes/fisiologia
13.
Mol Cell Neurosci ; 41(2): 175-85, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19285136

RESUMO

Macrophage stimulating protein (MSP) is a pleiotropic growth factor that signals via the RON receptor tyrosine kinase. Here we demonstrate that MSP increases the proportion of cultured adult mouse DRG neurons displaying discernable neuritic processes and promotes the elongation and branching of these processes in a dose dependent manner. RON expression in adult DRG is largely restricted to nerve growth factor (NGF)-responsive nociceptive neurons, and MSP mimics the effects of NGF by increasing the expression of several mRNAs that encode functionally important proteins that are characteristically expressed by this neuronal sub-population. MSP mRNA is expressed at high levels in the peripheral target fields of DRG somatic afferents, but is undetectable in DRG, spinal cord or freshly dissected sciatic nerve. These results suggest that MSP is a peripheral target-derived neurotrophic factor for NGF-responsive adult DRG neurons.


Assuntos
Gânglios Espinais/citologia , Fator de Crescimento de Hepatócito/farmacologia , Fatores de Crescimento Neural/metabolismo , Nociceptores/efeitos dos fármacos , Nociceptores/fisiologia , Proteínas Proto-Oncogênicas/farmacologia , Animais , Camundongos , Neuritos/metabolismo , Neuritos/ultraestrutura , Nociceptores/citologia , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor trkA/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Canal de Cátion TRPA1 , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo
14.
Front Cell Dev Biol ; 8: 254, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32411702

RESUMO

CD40-activated CD40L reverse signaling is a major physiological regulator of neural process growth from many kinds of developing neurons. Here we have investigated whether CD40L-reverse signaling also influences dendrite spine number and morphology in striatal medium spiny neurons (MSNs). Golgi preparations revealed no differences in the spine density, but because the dendrite arbors of MSNs were larger and branched in Cd40 -/- mice, the total number of spines was greater in Cd40 -/- mice. We also detected more mature spines compared with wild-type littermates. Western blot revealed that MSN cultures from Cd40 -/- mice had significantly less PSD-95 and there were changes in RhoA/B/C and Cdc42. Immunocytochemistry revealed that PSD-95 was clustered in spines in Cd40 -/- neurons compared with more diffuse labeling in Cd40 +/+ neurons. Activation of CD40L-reverse signaling with CD40-Fc prevented the changes observed in Cd40 -/- cultures. Our findings suggest that CD40L-reverse signaling influences dendrite spine morphology and related protein expression and distribution.

15.
iScience ; 23(9): 101457, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32853992

RESUMO

Loss of midbrain dopaminergic (mDA) neurons and their axons is central to Parkinson's disease (PD). Growth differentiation factor (GDF)5 is a potential neurotrophic factor for PD therapy. However, the molecular mediators of its neurotrophic action are unknown. Our proteomics analysis shows that GDF5 increases the expression of serine threonine receptor-associated protein kinase (STRAP) and nucleoside diphosphate kinase (NME)1 in the SH-SY5Y neuronal cell line. GDF5 overexpression increased NME1 expression in adult rat brain in vivo. NME and STRAP mRNAs are expressed in developing and adult rodent midbrain. Expression of both STRAP and NME1 is necessary and sufficient for the promotion of neurite growth in SH-SY5Y cells by GDF5. NME1 treatment increased neurite growth in both SH-SY5Y cells and cultured mDA neurons. Expression patterns of NME and STRAP are altered in PD midbrain. NME1 and STRAP are thus key mediators of GDF5's neurotrophic effects, rationalizing their future study as therapeutic targets for PD.

16.
Neuronal Signal ; 4(1): NS20200006, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32714600

RESUMO

Neuroblastoma (NB) is a paediatric cancer that arises in the sympathetic nervous system. Patients with stage 4 tumours have poor outcomes and 20% of high-risk cases have MYCN amplification. The bone morphogenetic proteins (BMPs) play roles in sympathetic neuritogenesis, by signalling through bone morphogenetic protein receptor (BMPR)2 and either BMPR1A or BMPR1B. Alterations in BMPR2 expression have been reported in NB; it is unknown if the expression of BMPR1A or BMPR1B is altered. We report lower BMPR2 and BMPR1B, and higher BMPR1A, expression in stage 4 and in MYCN-amplified NB. Kaplan-Meier plots showed that high BMPR2 or BMPR1B expression was linked to better survival, while high BMPR1A was linked to worse survival. Gene ontology enrichment and pathway analyses revealed that BMPR2 and BMPR1B co-expressed genes were enriched in those associated with NB differentiation. BMPR1A co-expressed genes were enriched in those associated with cell proliferation. Moreover, the correlation between BMPR2 and BMPR1A was strengthened, while the correlation between BMPR2 and BMPR1B was lost, in MYCN-amplified NB. This suggested that differentiation should decrease BMPR1A and increase BMPR1B expression. In agreement, nerve growth factor treatment of cultured sympathetic neurons decreased Bmpr1a expression and increased Bmpr1b expression. Overexpression of dominant negative BMPR1B, treatment with a BMPR1B inhibitor and treatment with GDF5, which signals via BMPR1B, showed that BMPR1B signalling is required for optimal neuritogenesis in NB cells, suggesting that loss of BMPR1B may alter neuritogenesis. The present study shows that expression of distinct BMPRs is associated with different survival outcomes in NB.

17.
J Neuropathol Exp Neurol ; 78(8): 725-734, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31225596

RESUMO

Medium spiny neurons (MSNs) comprise the vast majority of neurons in the striatum. Changes in the exuberant dendrites of these widely connected neurons are associated with a multitude of neurological conditions and are caused by a variety of recreational and medicinal drugs. However, we have a poor understanding of the physiological regulators of dendrite growth and elaboration of this clinically important population of neurons. Here, we show that MSN dendrites are markedly smaller and less branched in neonatal mice that possess a homozygous null mutation in the neuregulin-4 gene (Nrg4-/-) compared with wild type (Nrg4+/+) littermates. Nrg4-/- mice also had a highly significant reduction in MSN dendrite spine number in neonates and adults. The striking stunted dendrite arbor phenotype of MSNs observed in Nrg4-/- neonates was replicated in MSNs cultured from Nrg4-/- embryos and was completely rescued by soluble recombinant neuregulin-4. MSNs cultured from wild type mice coexpressed NRG4 and its receptor ErbB4. Our findings show that NRG4 is a major novel regulator of dendritic growth and arborization and spine formation in the striatum and suggest that it exerts its effects by an autocrine/paracrine mechanism.

18.
Dev Neurobiol ; 79(11-12): 949-962, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-32077240

RESUMO

CD40-activated CD40L reverse signaling is a major physiological regulator of the growth of neural processes in the developing nervous system. Previous work on superior cervical ganglion (SCG) neurons of the paravertebral sympathetic chain has shown that CD40L reverse signaling enhances NGF-promoted axon growth and tissue innervation. Here we show that CD40L reverse signaling has the opposite function in prevertebral ganglion (PVG) sympathetic neurons. During a circumscribed perinatal window of development, PVG neurons cultured from Cd40-/- mice had substantially larger, more exuberant axon arbors in the presence of NGF than PVG neurons cultured from wild-type mice. Tissues that receive their sympathetic innervation from PVG neurons were markedly hyperinnervated in Cd40-/- mice compared with wild-type mice. The exuberant axonal growth phenotype of cultured CD40-deficient perinatal PVG neurons was pared back to wild-type levels by activating CD40L reverse signaling with a CD40-Fc chimeric protein, but not by activating CD40 forward signaling with CD40L. The co-expression of CD40 and CD40L in PVG neurons suggests that these proteins engage in an autocrine signaling loop in these neurons. Our work shows that CD40L reverse signaling is a physiological regulator of NGF-promoted sympathetic axon growth and tissue innervation with opposite effects in paravertebral and prevertebral neurons.


Assuntos
Axônios/metabolismo , Ligante de CD40/metabolismo , Neurônios/metabolismo , Gânglio Cervical Superior/metabolismo , Animais , Gânglios Simpáticos/metabolismo , Camundongos Transgênicos , Fator de Crescimento Neural/metabolismo
19.
Dev Neurobiol ; 79(4): 317-334, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31004466

RESUMO

Members of the TNF and TNF receptor superfamilies acting by both forward and reverse signaling are increasingly recognized as major physiological regulators of axon growth and tissue innervation in development. Studies of the experimentally tractable superior cervical ganglion (SCG) neurons and their targets have shown that only TNF reverse signaling, not forward signaling, is a physiological regulator of sympathetic innervation. Here, we compared SCG neurons and their targets with prevertebral ganglion (PVG) neurons and their targets. Whereas all SCG targets were markedly hypoinnervated in both TNF-deficient and TNFR1-deficient mice, PVG targets were not hypoinnervated in these mice and one PVG target, the spleen, was significantly hyperinnervated. These in vivo regional differences in innervation density were related to in vitro differences in the responses of SCG and PVG neurons to TNF reverse and forward signaling. Though TNF reverse signaling enhanced SCG axon growth, it did not affect PVG axon growth. Whereas activation of TNF forward signaling in PVG axons inhibited growth, TNF forward signaling could not be activated in SCG axons. These latter differences in the response of SCG and PVG axons to TNF forward signaling were related to TNFR1 expression, whereas PVG axons expressed TNFR1, SCG axons did not. These results show that both TNF reverse and forward signaling are physiological regulators of sympathetic innervation in different tissues.


Assuntos
Axônios/metabolismo , Gânglios Simpáticos/crescimento & desenvolvimento , Gânglios Simpáticos/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células Cultivadas , Camundongos Knockout , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética
20.
Front Cell Dev Biol ; 7: 191, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31572723

RESUMO

Parkinson's disease is characterized by the intracellular accumulation of α-synuclein which has been linked to early dopaminergic axonal degeneration. Identifying druggable targets that can promote axonal growth in cells overexpressing α-synuclein is important in order to develop strategies for early intervention. Class-IIa histone deacetylases (HDACs) have previously emerged as druggable targets, however, it is not known which specific class-IIa HDACs should be targeted to promote neurite growth in dopaminergic neurons. To provide insight into this, we used gene co-expression analysis to identify which, if any, of the class-IIa HDACs had a positive correlation with markers of dopaminergic neurons in the human substantia nigra. This revealed that two histone deacetylases, HDAC5 and HDAC9, are co-expressed with TH, GIRK2 and ALDH1A1 in the human SN. We further found that HDAC5 and HDAC9 are expressed in dopaminergic neurons in the adult mouse substantia nigra. We show that siRNAs targeting HDAC5 or HDAC9 can promote neurite growth in SH-SY5Y cells, and that their pharmacological inhibition, using the drug MC1568, promoted neurite growth in cultured rat dopaminergic neurons. Moreover, MC1568 treatment upregulated the expression of the neurotrophic factor, BMP2, and its downstream transcription factor, SMAD1. In addition, MC1568 or siRNAs targeting HDAC5 or HDAC9 led to an increase in Smad-dependent GFP expression in a reporter assay. Furthermore, MC1568 treatment of cultured rat dopaminergic neurons increased cellular levels of phosphorylated Smad1, which was prevented by the BMP receptor inhibitor, dorsomorphin. Dorsomorphin treatment prevented the neurite growth-promoting effects of siRNAs targeting HDAC5, as did overexpression of dominant-negative Smad4 or of the inhibitory Smad7, demonstrating a functional link to BMP signaling. Supplementation with BMP2 prevented the neurite growth-inhibitory effects of nuclear-restricted HDAC5. Finally, we report that siRNAs targeting HDAC5 or HDAC9 promoted neurite growth in cells overexpressing wild-type or A53T-α-synuclein and that MC1568 protected cultured rat dopaminergic neurons against the neurotoxin, MPP+. These findings establish HDAC5 and HDAC9 as novel regulators of BMP-Smad signaling, that additionally may be therapeutic targets worthy of further exploration in iPSC-derived human DA neurons and in vivo models of Parkinson's disease.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA