Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.069
Filtrar
1.
Transplant Proc ; 56(3): 712-714, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38355371

RESUMO

BACKGROUND: Inappropriate matching of motor and sensory fibers after nerve repair or grafting can lead to nerve recovery failure. Identifying the motor and sensory fascicles enables surgeons to match them accurately and correctly align nerve stumps, which is crucial for neural regeneration. Very few methods have been reported to differentiate between the sensory and motor nerve fascicles, and the replicability of these techniques remains unestablished. In this study, we aimed to assess the accuracy of axonal cholinesterase (CE) histochemical staining in distinguishing motor and sensory nerve fibers. METHODS: The femoral and sciatic nerves were harvested from rats. The specimens were immediately cut, frozen in isopentane, and cooled with liquid nitrogen. Nerve serial cross-sections were processed for hematoxylin and eosin staining, followed by CE histochemistry. The staining protocol solutions included acetylthiocholine iodide, phosphate buffer, cobalt sulfate hydrate, potassium phosphate monobasic, sulfuric acid, sodium bicarbonate, glutaraldehyde, and ammonium sulfide. RESULTS: Cross-sections of nerves containing efferent and afferent nerve fibers in segregated fascicles showed that CE activity was confined to motor neurons. A histochemical study revealed that motor fibers with high cholinesterase activity can be differentiated from sensory fibers. The motor branches of the femoral and sciatic nerves showed specific axonal staining, whereas the sensory branch did not show any specific staining. CONCLUSION: CE histochemical staining is a useful technique for distinguishing between motor and sensory nerve fibers. It can be potentially useful in improving the outcomes of nerve grafts or extremity allotransplantation surgery.


Assuntos
Colinesterases , Neurônios Motores , Nervo Isquiático , Coloração e Rotulagem , Animais , Nervo Isquiático/enzimologia , Ratos , Colinesterases/metabolismo , Colinesterases/análise , Coloração e Rotulagem/métodos , Neurônios Motores/enzimologia , Axônios/enzimologia , Células Receptoras Sensoriais/enzimologia , Masculino , Nervo Femoral , Ratos Sprague-Dawley
2.
J Biol Chem ; 298(3): 101647, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35101451

RESUMO

The dual leucine zipper kinase (DLK) is a key regulator of axon regeneration and degeneration in response to neuronal injury; however, regulatory mechanisms of the DLK function via its interacting proteins are largely unknown. To better understand the molecular mechanism of DLK function, we performed yeast two-hybrid screening analysis and identified FK506-binding protein-like (FKBPL, also known as WAF-1/CIP1 stabilizing protein 39) as a DLK-binding protein. FKBPL binds to the kinase domain of DLK and inhibits its kinase activity. In addition, FKBPL induces DLK protein degradation through ubiquitin-dependent pathways. We further assessed other members in the FKBP protein family and found that FK506-binding protein 8 (FKBP8) also induced DLK degradation. We identified the lysine 271 residue in the kinase domain as a major site of DLK ubiquitination and SUMO3 conjugation and was thus responsible for regulating FKBP8-mediated proteasomal degradation that was inhibited by the substitution of the lysine 271 to arginine. FKBP8-mediated degradation of DLK is mediated by autophagy pathway because knockdown of Atg5 inhibited DLK destabilization. We show that in vivo overexpression of FKBP8 delayed the progression of axon degeneration and suppressed neuronal death after axotomy in sciatic and optic nerves. Taken together, this study identified FKBPL and FKBP8 as novel DLK-interacting proteins that regulate DLK stability via the ubiquitin-proteasome and lysosomal protein degradation pathways.


Assuntos
Axônios , MAP Quinase Quinase Quinases , Degeneração Neural , Proteínas de Ligação a Tacrolimo , Axônios/enzimologia , Axônios/metabolismo , Axônios/patologia , Zíper de Leucina , Lisina/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Degeneração Neural/enzimologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Regeneração Nervosa , Proteínas de Ligação a Tacrolimo/metabolismo , Ubiquitina/metabolismo
3.
Invest Ophthalmol Vis Sci ; 63(1): 4, 2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-34982146

RESUMO

Purpose: Netarsudil, a Rho kinase inhibitor with norepinephrine transport inhibitory effect, lowers intraocular pressure, however, its effect on axon damage remains to be elucidated. The aim of the current study was to investigate the effect of netarsudil on TNF-induced axon loss and to examine whether it affects phosphorylated-AMP-activated kinase (p-AMPK) and autophagy in the optic nerve. Methods: Intravitreal administration of TNF or TNF with netarsudil was carried out on rats and quantification of axon number was determined. Electron microscopy determined autophagosome numbers. Localization of p-AMPK expression was examined by immunohistochemistry. The changes in p62, LC3-II, and p-AMPK levels were estimated in the optic nerve by immunoblot analysis. The effect of an AMPK activator A769662 or an AMPK inhibitor dorsomorphin on axon number was evaluated. Results: Morphometric analysis revealed apparent protection by netarsudil against TNF-induced axon degeneration. Netarsudil increased autophagosome numbers inside axons. Netarsudil treatment significantly upregulated optic nerve LC3-II levels in both the TNF-treated eyes and the control eyes. Increased p62 protein level induced by TNF was significantly ameliorated by netarsudil. The netarsudil administration alone lessened p62 levels. Netarsudil significantly upregulated the optic nerve p-AMPK levels. A769662 exhibited obvious axonal protection against TNF-induced damage. A769662 treatment upregulated LC3-II levels and the increment of p62 level induced by TNF was significantly ameliorated by A769662. Immunohistochemical analysis revealed that p-AMPK is present in axons. Netarsudil-mediated axonal protection was significantly suppressed by dorsomorphin administration. Conclusions: Netarsudil upregulated p-AMPK and autophagy. Netarsudil-mediated axonal protection may be associated with upregulated p-AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/fisiologia , Axônios/efeitos dos fármacos , Benzoatos/farmacologia , Degeneração Neural/prevenção & controle , Nervo Óptico/efeitos dos fármacos , Fator de Necrose Tumoral alfa/toxicidade , beta-Alanina/análogos & derivados , Quinases Associadas a rho/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Animais , Axônios/enzimologia , Axônios/patologia , Compostos de Bifenilo/farmacologia , Inibidores Enzimáticos/farmacologia , Imuno-Histoquímica , Injeções Intravítreas , Masculino , Microscopia Eletrônica , Proteínas Associadas aos Microtúbulos/metabolismo , Degeneração Neural/enzimologia , Nervo Óptico/ultraestrutura , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Pironas/farmacologia , Ratos , Ratos Wistar , Proteína Sequestossoma-1/metabolismo , Tiofenos/farmacologia , beta-Alanina/farmacologia
4.
Cell Rep ; 37(3): 109872, 2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34686345

RESUMO

SARM1 is an inducible TIR-domain NAD+ hydrolase that mediates pathological axon degeneration. SARM1 is activated by an increased ratio of NMN to NAD+, which competes for binding to an allosteric activating site. When NMN binds, the TIR domain is released from autoinhibition, activating its NAD+ hydrolase activity. The discovery of this allosteric activating site led us to hypothesize that other NAD+-related metabolites might activate SARM1. Here, we show the nicotinamide analog 3-acetylpyridine (3-AP), first identified as a neurotoxin in the 1940s, is converted to 3-APMN, which activates SARM1 and induces SARM1-dependent NAD+ depletion, axon degeneration, and neuronal death. In mice, systemic treatment with 3-AP causes rapid SARM1-dependent death, while local application to the peripheral nerve induces SARM1-dependent axon degeneration. We identify 2-aminopyridine as another SARM1-dependent neurotoxin. These findings identify SARM1 as a candidate mediator of environmental neurotoxicity and suggest that SARM1 agonists could be developed into selective agents for neurolytic therapy.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Axônios/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Gânglios Espinais/efeitos dos fármacos , Degeneração Neural , Síndromes Neurotóxicas/etiologia , Neurotoxinas/toxicidade , Piridinas/toxicidade , Nervo Isquiático/efeitos dos fármacos , Ativação Metabólica , Regulação Alostérica , Animais , Proteínas do Domínio Armadillo/genética , Axônios/enzimologia , Axônios/patologia , Domínio Catalítico , Morte Celular , Citocinas/genética , Citocinas/metabolismo , Proteínas do Citoesqueleto/genética , Ativação Enzimática , Feminino , Gânglios Espinais/enzimologia , Gânglios Espinais/patologia , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndromes Neurotóxicas/enzimologia , Síndromes Neurotóxicas/patologia , Neurotoxinas/metabolismo , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo , Nicotinamida-Nucleotídeo Adenililtransferase/genética , Nicotinamida-Nucleotídeo Adenililtransferase/metabolismo , Piridinas/metabolismo , Nervo Isquiático/enzimologia , Nervo Isquiático/patologia , Transdução de Sinais
5.
Elife ; 102021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34543184

RESUMO

Synaptotagmin 7 (SYT7) has emerged as a key regulator of presynaptic function, but its localization and precise role in the synaptic vesicle cycle remain the subject of debate. Here, we used iGluSnFR to optically interrogate glutamate release, at the single-bouton level, in SYT7KO-dissociated mouse hippocampal neurons. We analyzed asynchronous release, paired-pulse facilitation, and synaptic vesicle replenishment and found that SYT7 contributes to each of these processes to different degrees. 'Zap-and-freeze' electron microscopy revealed that a loss of SYT7 diminishes docking of synaptic vesicles after a stimulus and inhibits the recovery of depleted synaptic vesicles after a stimulus train. SYT7 supports these functions from the axonal plasma membrane, where its localization and stability require both γ-secretase-mediated cleavage and palmitoylation. In summary, SYT7 is a peripheral membrane protein that controls multiple modes of synaptic vesicle (SV) exocytosis and plasticity, in part, through enhancing activity-dependent docking of SVs.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Axônios/enzimologia , Membrana Celular/enzimologia , Hipocampo/enzimologia , Vesículas Sinápticas/enzimologia , Sinaptotagminas/metabolismo , Animais , Axônios/ultraestrutura , Membrana Celular/ultraestrutura , Células Cultivadas , Exocitose , Hipocampo/ultraestrutura , Lipoilação , Camundongos Knockout , Simulação de Acoplamento Molecular , Plasticidade Neuronal , Processamento de Proteína Pós-Traducional , Transporte Proteico , Proteólise , Ratos Sprague-Dawley , Transmissão Sináptica , Vesículas Sinápticas/ultraestrutura , Sinaptotagminas/genética , Fatores de Tempo
6.
Dev Cell ; 56(7): 976-984.e3, 2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33823136

RESUMO

Axon remodeling through sprouting and pruning contributes to the refinement of developing neural circuits. A prominent example is the pruning of developing sensory axons deprived of neurotrophic support, which is mediated by a caspase-dependent (apoptotic) degeneration process. Distal sensory axons possess a latent apoptotic pathway, but a cell body-derived signal that travels anterogradely down the axon is required for pathway activation. The signaling mechanisms that underlie this anterograde process are poorly understood. Here, we show that the tumor suppressor P53 is required for anterograde signaling. Interestingly loss of P53 blocks axonal but not somatic (i.e., cell body) caspase activation. Unexpectedly, P53 does not appear to have an acute transcriptional role in this process and instead appears to act in the cytoplasm to directly activate the mitochondrial apoptotic pathway in axons. Our data support the operation of a cytoplasmic role for P53 in the anterograde death of developing sensory axons.


Assuntos
Axônios/fisiologia , Células Receptoras Sensoriais/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Axônios/enzimologia , Axônios/metabolismo , Caspases/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Camundongos , Domínios Proteicos , Células Receptoras Sensoriais/enzimologia , Células Receptoras Sensoriais/metabolismo , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Proteína bcl-X/antagonistas & inibidores
7.
Cell Rep ; 34(8): 108778, 2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33626357

RESUMO

The 3' untranslated regions (3' UTRs) of messenger RNAs (mRNAs) are non-coding sequences involved in many aspects of mRNA metabolism, including intracellular localization and translation. Incorrect processing and delivery of mRNA cause severe developmental defects and have been implicated in many neurological disorders. Here, we use deep sequencing to show that in sympathetic neuron axons, the 3' UTRs of many transcripts undergo cleavage, generating isoforms that express the coding sequence with a short 3' UTR and stable 3' UTR-derived fragments of unknown function. Cleavage of the long 3' UTR of Inositol Monophosphatase 1 (IMPA1) mediated by a protein complex containing the endonuclease argonaute 2 (Ago2) generates a translatable isoform that is necessary for maintaining the integrity of sympathetic neuron axons. Thus, our study provides a mechanism of mRNA metabolism that simultaneously regulates local protein synthesis and generates an additional class of 3' UTR-derived RNAs.


Assuntos
Regiões 3' não Traduzidas , Axônios/enzimologia , Corpo Celular/enzimologia , Monoéster Fosfórico Hidrolases/metabolismo , RNA Mensageiro/metabolismo , Gânglio Cervical Superior/enzimologia , Transcrição Gênica , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Proteína Semelhante a ELAV 4/genética , Proteína Semelhante a ELAV 4/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Masculino , Células PC12 , Monoéster Fosfórico Hidrolases/genética , Proteínas de Ligação a Poli(A)/genética , Proteínas de Ligação a Poli(A)/metabolismo , Poliadenilação , Biossíntese de Proteínas , Isoformas de Proteínas , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Gânglio Cervical Superior/citologia , Transativadores/genética , Transativadores/metabolismo
8.
Mol Brain ; 14(1): 31, 2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33579325

RESUMO

Axon regeneration in the central nervous system is inefficient. However, the neurons in the peripheral nervous system display robust regeneration after injury, indicating that axonal regeneration is differentially controlled under various conditions. To identify those molecules regulating axon regeneration, comparative analysis from dorsal root ganglion neurons at embryonic or adult stages is utilized, which reveals that PDK1 is functions as a negative regulator of axon regeneration. PDK1 is downregulated in embryonic neurons after axotomy. In contrast, sciatic nerve axotomy upregulated PDK1 at protein levels from adult mice. The knockdown of PDK1 or the chemical inhibition of PDK1 promotes axon regeneration in vitro and in vivo. Here we present PDK1 as a new player to negatively regulate axon regeneration and as a potential target in the development of therapeutic applications.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Axônios/enzimologia , Axônios/fisiologia , Regeneração Nervosa/fisiologia , Canais de Cátion TRPP/metabolismo , Animais , Axônios/efeitos dos fármacos , Axotomia , Regulação para Baixo/efeitos dos fármacos , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Indazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Regeneração Nervosa/efeitos dos fármacos , Pirimidinas/farmacologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Regulação para Cima/efeitos dos fármacos
9.
Mol Neurobiol ; 58(1): 391-407, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32959171

RESUMO

In peripheral neuropathies, axonal degeneration (AxD) impairs the prognosis for recovery. Here, we describe a role for dual specificity phosphatases (DUSPs; MAP kinase phosphatases, MKPs), in supporting autonomous axon plasticity and viability. Both DUSPs 1 and 4 were identified within intact or axotomized sensory neurons. Knockdown of DUSP 1 or 4 independently or combined impaired neurite outgrowth in adult dissociated sensory neurons. Furthermore, adult sensory neurons with DUSP knockdown were rendered sensitive to axonopathy in vitro following exposure to low, subtoxic TrpV1 (transient receptor potential cation channel subfamily V member 1) activation by capsaicin, an intervention normally supportive of growth. This was not prevented by concurrent DLK (dual leucine zipper kinase) knockdown. Ex vivo neurofilament dissolution was heightened by DUSP inhibition within explanted nerves. In vivo DUSP knockdown or inhibition was associated with more rapid loss of motor axon excitability. The addition of SARM1 (sterile alpha and TIR motif containing 1) siRNA abrogated DUSP1 and 4 mediated loss of excitability. DUSP knockdown accelerated neurofilament breakdown and there was earlier morphological evidence of myelinated axon degeneration distal to axotomy. Taken together, the findings identify a key role for DUSPs in supporting axon plasticity and survival.


Assuntos
Axônios/enzimologia , Axônios/patologia , Fosfatases de Especificidade Dupla/metabolismo , Plasticidade Neuronal , Animais , Axotomia , Sobrevivência Celular , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Gânglios Espinais/metabolismo , Filamentos Intermediários/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Masculino , Degeneração Neural/patologia , Crescimento Neuronal , Neuroproteção , Ratos Sprague-Dawley , Células Receptoras Sensoriais/metabolismo
10.
Proc Natl Acad Sci U S A ; 117(52): 33597-33607, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33318207

RESUMO

Axon injury is a hallmark of many neurodegenerative diseases, often resulting in neuronal cell death and functional impairment. Dual leucine zipper kinase (DLK) has emerged as a key mediator of this process. However, while DLK inhibition is robustly protective in a wide range of neurodegenerative disease models, it also inhibits axonal regeneration. Indeed, there are no genetic perturbations that are known to both improve long-term survival and promote regeneration. To identify such a neuroprotective target, we conducted a set of complementary high-throughput screens using a protein kinase inhibitor library in human stem cell-derived retinal ganglion cells (hRGCs). Overlapping compounds that promoted both neuroprotection and neurite outgrowth were bioinformatically deconvoluted to identify specific kinases that regulated neuronal death and axon regeneration. This work identified the role of germinal cell kinase four (GCK-IV) kinases in cell death and additionally revealed their unexpected activity in suppressing axon regeneration. Using an adeno-associated virus (AAV) approach, coupled with genome editing, we validated that GCK-IV kinase knockout improves neuronal survival, comparable to that of DLK knockout, while simultaneously promoting axon regeneration. Finally, we also found that GCK-IV kinase inhibition also prevented the attrition of RGCs in developing retinal organoid cultures without compromising axon outgrowth, addressing a major issue in the field of stem cell-derived retinas. Together, these results demonstrate a role for the GCK-IV kinases in dissociating the cell death and axonal outgrowth in neurons and their druggability provides for therapeutic options for neurodegenerative diseases.


Assuntos
Axônios/enzimologia , Axônios/patologia , Sistema Nervoso Central/patologia , Quinases do Centro Germinativo/metabolismo , Regeneração Nervosa , Animais , Sequência de Bases , Sistemas CRISPR-Cas/genética , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dependovirus/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL , Regeneração Nervosa/efeitos dos fármacos , Crescimento Neuronal/efeitos dos fármacos , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Organoides/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
Elife ; 92020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33185190

RESUMO

The palmitoyl acyltransferase (PAT) ZDHHC14 is highly expressed in the hippocampus and is the only PAT predicted to bind Type-I PDZ domain-containing proteins. However, ZDHHC14's neuronal roles are unknown. Here, we identify the PDZ domain-containing Membrane-associated Guanylate Kinase (MaGUK) PSD93 as a direct ZDHHC14 interactor and substrate. PSD93, but not other MaGUKs, localizes to the axon initial segment (AIS). Using lentiviral-mediated shRNA knockdown in rat hippocampal neurons, we find that ZDHHC14 controls palmitoylation and AIS clustering of PSD93 and also of Kv1 potassium channels, which directly bind PSD93. Neurodevelopmental expression of ZDHHC14 mirrors that of PSD93 and Kv1 channels and, consistent with ZDHHC14's importance for Kv1 channel clustering, loss of ZDHHC14 decreases outward currents and increases action potential firing in hippocampal neurons. To our knowledge, these findings identify the first neuronal roles and substrates for ZDHHC14 and reveal a previously unappreciated role for palmitoylation in control of neuronal excitability.


Assuntos
Aciltransferases/metabolismo , Axônios/enzimologia , Superfamília Shaker de Canais de Potássio/metabolismo , Aciltransferases/genética , Animais , Fenômenos Eletrofisiológicos , Regulação Enzimológica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Hipocampo/citologia , Humanos , Camundongos , Ligação Proteica , Superfamília Shaker de Canais de Potássio/genética , Técnicas do Sistema de Duplo-Híbrido
12.
Viruses ; 12(9)2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32911874

RESUMO

Since the global outbreak of SARS-CoV-2 (COVID-19), infections of diverse human organs along with multiple symptoms continue to be reported. However, the susceptibility of the brain to SARS-CoV-2, and the mechanisms underlying neurological infection are still elusive. Here, we utilized human embryonic stem cell-derived brain organoids and monolayer cortical neurons to investigate infection of brain with pseudotyped SARS-CoV-2 viral particles. Spike-containing SARS-CoV-2 pseudovirus infected neural layers within brain organoids. The expression of ACE2, a host cell receptor for SARS-CoV-2, was sustained during the development of brain organoids, especially in the somas of mature neurons, while remaining rare in neural stem cells. However, pseudotyped SARS-CoV-2 was observed in the axon of neurons, which lack ACE2. Neural infectivity of SARS-CoV-2 pseudovirus did not increase in proportion to viral load, but only 10% of neurons were infected. Our findings demonstrate that brain organoids provide a useful model for investigating SARS-CoV-2 entry into the human brain and elucidating the susceptibility of the brain to SARS-CoV-2.


Assuntos
Betacoronavirus/fisiologia , Neurônios/virologia , Organoides/virologia , Prosencéfalo/virologia , Glicoproteína da Espícula de Coronavírus/fisiologia , Enzima de Conversão de Angiotensina 2 , Axônios/enzimologia , Diferenciação Celular , Células Cultivadas , Córtex Cerebral/citologia , Células-Tronco Embrionárias/virologia , Células HEK293 , Humanos , Proteínas do Tecido Nervoso/fisiologia , Células-Tronco Neurais/enzimologia , Células-Tronco Neurais/virologia , Neurônios/enzimologia , Peptidil Dipeptidase A/fisiologia , Prosencéfalo/citologia , Receptores Virais/fisiologia , SARS-CoV-2 , Carga Viral , Tropismo Viral , Internalização do Vírus
13.
J Neurosci ; 40(42): 8103-8118, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32917789

RESUMO

Interstitial axon branching is an essential step during the establishment of neuronal connectivity. However, the exact mechanisms on how the number and position of branches are determined are still not fully understood. Here, we investigated the role of Arl8B, an adaptor molecule between lysosomes and kinesins. In chick retinal ganglion cells (RGCs), downregulation of Arl8B reduces axon branch density and shifts their location more proximally, while Arl8B overexpression leads to increased density and more distal positions of branches. These alterations correlate with changes in the location and density of lysosomes and autophagosomes along the axon shaft. Diminishing autophagy directly by knock-down of atg7, a key autophagy gene, reduces branch density, while induction of autophagy by rapamycin increases axon branching, indicating that autophagy plays a prominent role in axon branch formation. In vivo, local inactivation of autophagy in the retina using a mouse conditional knock-out approach disturbs retino-collicular map formation which is dependent on the formation of interstitial axon branches. These data suggest that Arl8B plays a principal role in the positioning of axon branches by spatially controlling autophagy, thus directly controlling formation of neural connectivity in the brain.SIGNIFICANCE STATEMENT The formation of interstitial axonal branches plays a prominent role in numerous places of the developing brain during neural circuit establishment. We show here that the GTPase Arl8B controls density and location of interstitial axon branches, and at the same time controls also density and location of the autophagy machinery. Upregulation or downregulation of autophagy in vitro promotes or inhibits axon branching. Local disruption of autophagy in vivo disturbs retino-collicular mapping. Our data suggest that Arl8B controls axon branching by controlling locally autophagy. This work is one of the first reports showing a role of autophagy during early neural circuit development and suggests that autophagy in general plays a much more prominent role during brain development than previously anticipated.


Assuntos
Fatores de Ribosilação do ADP/fisiologia , Autofagossomos/fisiologia , Axônios/fisiologia , Lisossomos/fisiologia , Fatores de Ribosilação do ADP/metabolismo , Animais , Autofagossomos/enzimologia , Autofagossomos/ultraestrutura , Autofagia/genética , Axônios/enzimologia , Axônios/ultraestrutura , Embrião de Galinha , Regulação para Baixo , Técnicas de Silenciamento de Genes , Lisossomos/enzimologia , Lisossomos/ultraestrutura , Camundongos Knockout , Cultura Primária de Células , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/ultraestrutura
14.
Cell Rep ; 31(6): 107639, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32402271

RESUMO

The generation of axonal and dendritic domains is critical for brain circuitry assembly and physiology. Negative players, such as the RhoA-Rho coiled-coil-associated protein kinase (ROCK) signaling pathway, restrain axon development and polarization. Surprisingly, the genetic control of neuronal polarity has remained largely unexplored. Here, we report that, in primary cultured neurons, expression of the histone methyltransferase G9a and nuclear translocation of its major splicing isoform (G9a/E10+) peak at the time of axon formation. RNAi suppression of G9a/E10+ or pharmacological blockade of G9a constrains neuronal migration, axon initiation, and the establishment of neuronal polarity in situ and in vitro. Inhibition of G9a function upregulates RhoA-ROCK activity by increasing the expression of Lfc, a guanine nucleotide exchange factor (GEF) for RhoA. Together, these results identify G9a as a player in neuronal polarization.


Assuntos
Axônios/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Neurônios/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Axônios/enzimologia , Movimento Celular/fisiologia , Células Cultivadas , Epigênese Genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Gravidez , Ratos , Ratos Wistar , Transdução de Sinais , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Quinases Associadas a rho , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores
15.
Biomolecules ; 10(3)2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32150898

RESUMO

Components of the extracellular matrix (ECM) are key players in regulating cellular functions throughout the whole organism. In fact, ECM components not only participate in tissue organization but also contribute to processes such as cellular maintenance, proliferation, and migration, as well as to support for various signaling pathways. In the central nervous system (CNS), proteoglycans of the lectican family, such as versican, aggrecan, brevican, and neurocan, are important constituents of the ECM. In recent years, members of this family have been found to be involved in the maintenance of CNS homeostasis and to participate directly in processes such as the organization of perineural nets, the regulation of brain plasticity, CNS development, brain injury repair, axonal guidance, and even the altering of synaptic responses. ADAMTSs are a family of "A disintegrin and metalloproteinase with thrombospondin motifs" proteins that have been found to be involved in a multitude of processes through the degradation of lecticans and other proteoglycans. Recently, alterations in ADAMTS expression and activity have been found to be involved in neuronal disorders such as stroke, neurodegeneration, schizophrenia, and even Alzheimer's disease, which in turn may suggest their potential use as therapeutic targets. Herein, we summarize the different roles of ADAMTSs in regulating CNS events through interactions and the degradation of ECM components (more specifically, the lectican family of proteoglycans).


Assuntos
Proteínas ADAMTS/metabolismo , Axônios/enzimologia , Encefalopatias/enzimologia , Encéfalo/enzimologia , Matriz Extracelular/enzimologia , Proteoglicanas/metabolismo , Animais , Axônios/patologia , Encéfalo/patologia , Encefalopatias/patologia , Humanos
16.
Nucleic Acids Res ; 48(8): 3999-4012, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32201888

RESUMO

In eukaryotic cells, with the exception of the specialized genomes of mitochondria and plastids, all genetic information is sequestered within the nucleus. This arrangement imposes constraints on how the information can be tailored for different cellular regions, particularly in cells with complex morphologies like neurons. Although messenger RNAs (mRNAs), and the proteins that they encode, can be differentially sorted between cellular regions, the information itself does not change. RNA editing by adenosine deamination can alter the genome's blueprint by recoding mRNAs; however, this process too is thought to be restricted to the nucleus. In this work, we show that ADAR2 (adenosine deaminase that acts on RNA), an RNA editing enzyme, is expressed outside of the nucleus in squid neurons. Furthermore, purified axoplasm exhibits adenosine-to-inosine activity and can specifically edit adenosines in a known substrate. Finally, a transcriptome-wide analysis of RNA editing reveals that tens of thousands of editing sites (>70% of all sites) are edited more extensively in the squid giant axon than in its cell bodies. These results indicate that within a neuron RNA editing can recode genetic information in a region-specific manner.


Assuntos
Adenosina Desaminase/metabolismo , Neurônios/enzimologia , Edição de RNA , Adenosina/metabolismo , Animais , Axônios/enzimologia , Citoplasma/enzimologia , Decapodiformes/enzimologia , Células HEK293 , Humanos , Inosina/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Sinapses/enzimologia
17.
Neuron ; 106(4): 589-606.e6, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32169171

RESUMO

ACOX1 (acyl-CoA oxidase 1) encodes the first and rate-limiting enzyme of the very-long-chain fatty acid (VLCFA) ß-oxidation pathway in peroxisomes and leads to H2O2 production. Unexpectedly, Drosophila (d) ACOX1 is mostly expressed and required in glia, and loss of ACOX1 leads to developmental delay, pupal death, reduced lifespan, impaired synaptic transmission, and glial and axonal loss. Patients who carry a previously unidentified, de novo, dominant variant in ACOX1 (p.N237S) also exhibit glial loss. However, this mutation causes increased levels of ACOX1 protein and function resulting in elevated levels of reactive oxygen species in glia in flies and murine Schwann cells. ACOX1 (p.N237S) patients exhibit a severe loss of Schwann cells and neurons. However, treatment of flies and primary Schwann cells with an antioxidant suppressed the p.N237S-induced neurodegeneration. In summary, both loss and gain of ACOX1 lead to glial and neuronal loss, but different mechanisms are at play and require different treatments.


Assuntos
Acil-CoA Oxidase/genética , Axônios/enzimologia , Degeneração Neural/genética , Neuroglia/enzimologia , Animais , Axônios/patologia , Drosophila , Humanos , Camundongos , Mutação , Degeneração Neural/enzimologia , Neuroglia/patologia , Ratos
18.
Trends Pharmacol Sci ; 41(4): 281-293, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32107050

RESUMO

Attempts to develop neuroprotective treatments for neurodegenerative disorders have not yet been clinically successful. Axonal degeneration has been recognized as a predominant driver of disability and disease progression in central nervous system (CNS) diseases such as amyotrophic lateral sclerosis (ALS), multiple sclerosis, and Parkinson's disease, peripheral nervous system (PNS) disorders such as chemotherapy-induced, diabetic, and inherited neuropathies, and ocular disorders, such as glaucoma. In recent years, sterile alpha and TIR motif containing 1 (SARM1) has emerged as the first compelling axonal-specific target for therapeutic intervention. In this review, we discuss the role of axonal degeneration in neurodegenerative disorders, with a focus on SARM1 and the discovery of its intrinsic enzymatic function. Establishment of neurofilament light chain (NfL) as a reliable biomarker of axonal damage, and the availability of an ultrasensitive method for measuring NfL in plasma or serum, provide translational tools to make development of axonal protective, SARM1 inhibitors a viable approach to treat multiple neurodegenerative disorders.


Assuntos
Proteínas do Domínio Armadillo/antagonistas & inibidores , Axônios/patologia , Proteínas do Citoesqueleto/antagonistas & inibidores , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/patologia , Animais , Proteínas do Domínio Armadillo/metabolismo , Axônios/efeitos dos fármacos , Axônios/enzimologia , Proteínas do Citoesqueleto/metabolismo , Humanos , Terapia de Alvo Molecular , Doenças Neurodegenerativas/enzimologia
19.
Sci Rep ; 10(1): 1280, 2020 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-31992746

RESUMO

Increases in axonal sodium currents in peripheral nerves are some of the earliest excitability changes observed in Amyotrophic Lateral Sclerosis (ALS) patients. Nothing is known, however, about axonal sodium channels more proximally, particularly at the action potential initiating region - the axon initial segment (AIS). Immunohistochemistry for Nav1.6 sodium channels was used to investigate parameters of AISs of spinal motoneurones in the G127X SOD1 mouse model of ALS in adult mice at presymptomatic time points (~190 days old). In vivo intracellular recordings from lumbar spinal motoneurones were used to determine the consequences of any AIS changes. AISs of both alpha and gamma motoneurones were found to be significantly shorter (by 6.6% and 11.8% respectively) in G127X mice as well as being wider by 9.8% (alpha motoneurones). Measurements from 20-23 day old mice confirmed that this represented a change during adulthood. Intracellular recordings from motoneurones in presymptomatic adult mice, however, revealed no differences in individual action potentials or the cells ability to initiate repetitive action potentials. To conclude, despite changes in AIS geometry, no evidence was found for reduced excitability within the functional working range of firing frequencies of motoneurones in this model of ALS.


Assuntos
Esclerose Lateral Amiotrófica , Axônios/enzimologia , Neurônios Motores , Mutação de Sentido Incorreto , Superóxido Dismutase-1 , Transmissão Sináptica , Substituição de Aminoácidos , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Axônios/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Neurônios Motores/enzimologia , Neurônios Motores/patologia , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo
20.
Science ; 365(6455): 793-799, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31439792

RESUMO

SARM1 (sterile alpha and TIR motif containing 1) is responsible for depletion of nicotinamide adenine dinucleotide in its oxidized form (NAD+) during Wallerian degeneration associated with neuropathies. Plant nucleotide-binding leucine-rich repeat (NLR) immune receptors recognize pathogen effector proteins and trigger localized cell death to restrict pathogen infection. Both processes depend on closely related Toll/interleukin-1 receptor (TIR) domains in these proteins, which, as we show, feature self-association-dependent NAD+ cleavage activity associated with cell death signaling. We further show that SARM1 SAM (sterile alpha motif) domains form an octamer essential for axon degeneration that contributes to TIR domain enzymatic activity. The crystal structures of ribose and NADP+ (the oxidized form of nicotinamide adenine dinucleotide phosphate) complexes of SARM1 and plant NLR RUN1 TIR domains, respectively, reveal a conserved substrate binding site. NAD+ cleavage by TIR domains is therefore a conserved feature of animal and plant cell death signaling pathways.


Assuntos
Proteínas do Domínio Armadillo/química , Proteínas do Citoesqueleto/química , NAD+ Nucleosidase/química , NAD/metabolismo , Proteínas de Plantas/química , Domínios Proteicos , Receptores Imunológicos/química , Animais , Proteínas do Domínio Armadillo/metabolismo , Axônios/enzimologia , Axônios/patologia , Sítios de Ligação , Morte Celular , Sequência Conservada , Cristalografia por Raios X , Proteínas do Citoesqueleto/metabolismo , Células HEK293 , Humanos , Camundongos , NAD+ Nucleosidase/metabolismo , NADP/metabolismo , Neurônios/enzimologia , Proteínas de Plantas/metabolismo , Multimerização Proteica , Receptores Imunológicos/metabolismo , Degeneração Walleriana/enzimologia , Degeneração Walleriana/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...