Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39201743

ABSTRACT

Neurodegenerative disorders, including traumatic injuries to the central nervous system (CNS) and neurodegenerative diseases, are characterized by early axonal damage, which does not regenerate in the adult mammalian CNS, leading to permanent neurological deficits. One of the primary causes of the loss of regenerative ability is thought to be a developmental decline in neurons' intrinsic capability for axon growth. Different molecules are involved in the developmental loss of the ability for axon regeneration, including many transcription factors. However, the function of microRNAs (miRNAs), which are also modulators of gene expression, in axon re-growth is still unclear. Among the various miRNAs recently identified with roles in the CNS, miR-17, which is highly expressed during early development, emerges as a promising target to promote axon regeneration. Here, we used adeno-associated viral (AAV) vectors to overexpress miR-17 (AAV.miR-17) in primary cortical neurons and evaluate its effects on neurite and axon regeneration in vitro. Although AAV.miR-17 had no significant effect on neurite outgrowth and arborization, it significantly enhances neurite regeneration after scratch lesion and axon regeneration after axotomy of neurons cultured in microfluidic chambers. Target prediction and functional annotation analyses suggest that miR-17 regulates gene expression associated with autophagy and cell metabolism. Our findings suggest that miR-17 promotes regenerative response and thus could mitigate neurodegenerative effects.


Subject(s)
Axons , Dependovirus , MicroRNAs , Nerve Regeneration , Neurites , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Axons/metabolism , Axons/physiology , Nerve Regeneration/genetics , Neurites/metabolism , Dependovirus/genetics , Cells, Cultured , Genetic Vectors/genetics , Mice , Neurons/metabolism
2.
Cells ; 12(17)2023 08 28.
Article in English | MEDLINE | ID: mdl-37681887

ABSTRACT

Advances in RNA-sequencing technologies have led to the identification of molecular biomarkers for several diseases, including neurodegenerative diseases, such as Alzheimer's, Parkinson's, Huntington's diseases and Amyotrophic Lateral Sclerosis. Despite the nature of glaucoma as a neurodegenerative disorder with several similarities with the other above-mentioned diseases, transcriptional data about this disease are still scarce. microRNAs are small molecules (~17-25 nucleotides) that have been found to be specifically expressed in the CNS as major components of the system regulating the development signatures of neurodegenerative diseases and the homeostasis of the brain. In this review, we sought to identify similarities between the functional mechanisms and the activated pathways of the most common neurodegenerative diseases, as well as to discuss how those mechanisms are regulated by miRNAs, using RNA-Seq as an approach to compare them. We also discuss therapeutically suitable applications for these disease hallmarks in clinical future studies.


Subject(s)
Glaucoma , MicroRNAs , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/therapy , RNA-Seq , Homeostasis , Glaucoma/genetics , Glaucoma/therapy , MicroRNAs/genetics
4.
Int J Mol Sci ; 23(7)2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35409291

ABSTRACT

Glaucoma is a neurodegenerative disease that affects the retinal ganglion cells (RGCs) and leads to progressive vision loss. The first pathological signs can be seen at the optic nerve head (ONH), the structure where RGC axons leave the retina to compose the optic nerve. Besides damage of the axonal cytoskeleton, axonal transport deficits at the ONH have been described as an important feature of glaucoma. Axonal transport is essential for proper neuronal function, including transport of organelles, synaptic components, vesicles, and neurotrophic factors. Impairment of axonal transport has been related to several neurodegenerative conditions. Studies on axonal transport in glaucoma include analysis in different animal models and in humans, and indicate that its failure happens mainly in the ONH and early in disease progression, preceding axonal and somal degeneration. Thus, a better understanding of the role of axonal transport in glaucoma is not only pivotal to decipher disease mechanisms but could also enable early therapies that might prevent irreversible neuronal damage at an early time point. In this review we present the current evidence of axonal transport impairment in glaucomatous neurodegeneration and summarize the methods employed to evaluate transport in this disease.


Subject(s)
Glaucoma , Neurodegenerative Diseases , Animals , Axonal Transport , Axons/metabolism , Disease Models, Animal , Glaucoma/metabolism , Neurodegenerative Diseases/metabolism , Retinal Ganglion Cells/pathology
5.
Invest Ophthalmol Vis Sci ; 63(2): 5, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35103748

ABSTRACT

Purpose: Based on our preview evidence that reduced nuclear content of the transcription factor Myc-associated protein X (MAX) is an early event associated with degeneration of retinal ganglion cells (RGCs), in the present study, our purpose was to test whether the overexpression of human MAX had a neuroprotective effect against RGC injury. Methods: Overexpression of either MAX or green fluorescent protein (GFP) in the retina was achieved by intravitreal injections of recombinant adenovirus-associated viruses (rAAVs). Lister Hooded rats were used in three models of RGC degeneration: (1) cultures of retinal explants for 30 hours ex vivo from the eyes of 14-day-old rats that had received intravitreal injections of rAAV2-MAX or the control vector rAAV2-GFP at birth; (2) an optic nerve crush model, in which 1-month-old rats received intravitreal injection of either rAAV2-MAX or rAAV2-GFP and, 4 weeks later, were operated on; and (3) an ocular hypertension (OHT) glaucoma model, in which 1-month-old rats received intravitreal injection of either rAAV2-MAX or rAAV2-GFP and, 4 weeks later, were subject to cauterization of the limbal plexus. Cell death was estimated by detection of pyknotic nuclei and TUNEL technique and correlated with MAX immunocontent in an ex vivo model of retinal explants. MAX expression was detected by quantitative RT-PCR. In the OHT model, survival of RGCs was quantified by retrograde labeling with DiI or immunostaining for BRN3a at 14 days after in vivo injury. Functional integrity of RGCs was analyzed through pattern electroretinography, and damage to the optic nerve was examined in semithin sections. Results: In all three models of RGC insult, gene therapy by overexpression of MAX prevented RGC death. Also, ON degeneration and electrophysiologic deficits were prevented in the OHT model. Conclusions: Our experiments offer proof of concept for a novel neuroprotective gene therapy for glaucomatous neurodegeneration based on overexpression of MAX.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Gene Expression Regulation , Genetic Therapy/methods , Glaucoma/complications , Nerve Regeneration/genetics , Neurodegenerative Diseases/therapy , Neuroprotection/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Cell Death , Disease Models, Animal , Female , Glaucoma/genetics , Glaucoma/pathology , Male , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Rats , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
6.
Cell Death Dis ; 12(2): 213, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637688

ABSTRACT

Axonal damage is an early step in traumatic and neurodegenerative disorders of the central nervous system (CNS). Damaged axons are not able to regenerate sufficiently in the adult mammalian CNS, leading to permanent neurological deficits. Recently, we showed that inhibition of the autophagic protein ULK1 promotes neuroprotection in different models of neurodegeneration. Moreover, we demonstrated previously that axonal protection improves regeneration of lesioned axons. However, whether axonal protection mediated by ULK1 inhibition could also improve axonal regeneration is unknown. Here, we used an adeno-associated viral (AAV) vector to express a dominant-negative form of ULK1 (AAV.ULK1.DN) and investigated its effects on axonal regeneration in the CNS. We show that AAV.ULK1.DN fosters axonal regeneration and enhances neurite outgrowth in vitro. In addition, AAV.ULK1.DN increases neuronal survival and enhances axonal regeneration after optic nerve lesion, and promotes long-term axonal protection after spinal cord injury (SCI) in vivo. Interestingly, AAV.ULK1.DN also increases serotonergic and dopaminergic axon sprouting after SCI. Mechanistically, AAV.ULK1.DN leads to increased ERK1 activation and reduced expression of RhoA and ROCK2. Our findings outline ULK1 as a key regulator of axonal degeneration and regeneration, and define ULK1 as a promising target to promote neuroprotection and regeneration in the CNS.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Axons/metabolism , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Nerve Regeneration , Optic Nerve Injuries/therapy , Optic Nerve/metabolism , Spinal Cord Injuries/therapy , Spinal Cord/metabolism , Animals , Autophagy-Related Protein-1 Homolog/genetics , Axons/pathology , Cells, Cultured , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Down-Regulation , Female , Mitogen-Activated Protein Kinase 3/metabolism , Neuronal Outgrowth , Optic Nerve/pathology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Rats, Wistar , Serotonergic Neurons/metabolism , Serotonergic Neurons/pathology , Spinal Cord/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Time Factors , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
7.
Curr Gene Ther ; 21(5): 362-381, 2021.
Article in English | MEDLINE | ID: mdl-33573569

ABSTRACT

Glaucoma leads to irreversible vision loss and current therapeutic strategies are often insufficient to prevent the progression of the disease and consequent blindness. Elevated intraocular pressure is an important risk factor, but not required for the progression of glaucomatous neurodegeneration. The demise of retinal ganglion cells represents the final common pathway of glaucomatous vision loss. Still, lifelong control of intraocular pressure is the only current treatment to prevent severe vision loss, although it frequently fails despite best practices. This scenario calls for the development of neuroprotective and pro-regenerative therapies targeting the retinal ganglion cells as well as the optic nerve. Several experimental studies have shown the potential of gene modulation as a tool for neuroprotection and regeneration. In this context, gene therapy represents an attractive approach as a persistent treatment for glaucoma. Viral vectors engineered to promote overexpression of a broad range of cellular factors have been shown to protect retinal ganglion cells and/or promote axonal regeneration in experimental models. Here, we review the mechanisms involved in glaucomatous neurodegeneration and regeneration in the central nervous system. Then, we point out the current limitations of gene therapy platforms and review a myriad of studies that use viral vectors to manipulate genes in retinal ganglion cells, as a strategy to promote neuroprotection and regeneration. Finally, we address the potential of combining neuroprotective and regenerative gene therapies as an approach to glaucomatous neurodegeneration.


Subject(s)
Glaucoma , Genetic Therapy , Glaucoma/genetics , Glaucoma/therapy , Humans , Intraocular Pressure , Neuroprotection , Retinal Ganglion Cells
8.
Cell Death Differ ; 27(10): 2810-2827, 2020 10.
Article in English | MEDLINE | ID: mdl-32341448

ABSTRACT

Axonal degeneration is a key and early pathological feature in traumatic and neurodegenerative disorders of the CNS. Following a focal lesion to axons, extended axonal disintegration by acute axonal degeneration (AAD) occurs within several hours. During AAD, the accumulation of autophagic proteins including Unc-51 like autophagy activating kinase 1 (ULK1) has been demonstrated, but its role is incompletely understood. Here, we study the effect of ULK1 inhibition in different models of lesion-induced axonal degeneration in vitro and in vivo. Overexpression of a dominant negative of ULK1 (ULK1.DN) in primary rat cortical neurons attenuates axotomy-induced AAD in vitro. Both ULK1.DN and the ULK1 inhibitor SBI-0206965 protect against AAD after rat optic nerve crush in vivo. ULK1.DN additionally attenuates long-term axonal degeneration after rat spinal cord injury in vivo. Mechanistically, ULK1.DN decreases autophagy and leads to an mTOR-mediated increase in translational proteins. Consistently, treatment with SBI-0206965 results in enhanced mTOR activation. ULK1.DN additionally modulates the differential splicing of the degeneration-associated genes Kif1b and Ddit3. These findings uncover ULK1 as an important mediator of axonal degeneration in vitro and in vivo, and elucidate its function in splicing, defining it as a putative therapeutic target.


Subject(s)
Autophagy-Related Protein-1 Homolog , Axons , Central Nervous System , Nerve Degeneration , Neurodegenerative Diseases , Animals , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Autophagy-Related Protein-1 Homolog/physiology , Axons/metabolism , Axons/pathology , Cells, Cultured , Central Nervous System/injuries , Central Nervous System/metabolism , Female , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Primary Cell Culture , Rats
9.
Mol Neurobiol ; 57(2): 685-697, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31446549

ABSTRACT

Loss of nigrostriatal projections by axonal degeneration is a key early event in Parkinson's disease (PD) pathophysiology, being accountable for the lack of dopamine in the nigrostriatal system and resulting in motor symptoms such as bradykinesia, rigidity, and tremor. Since autophagy is an important mechanism contributing to axonal degeneration, we aimed to evaluate the effects of competitive autophagy inhibition in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD in vivo. Adeno-associated viral vector (AAV)-mediated overexpression of a dominant-negative form of the unc-51 like autophagy-initiating kinase (ULK1.DN) in the substantia nigra was induced 3 weeks before MPTP treatment. Analysis of motor behavior demonstrated a significant improvement of ULK1.DN expressing mice after MPTP treatment. Immunohistochemical analyses of dopaminergic nigral neurons and nigrostriatal projections revealed a significant protection from MPTP-induced neurotoxicity after ULK1.DN expression. Western blot analysis linked these findings to an activation of mTOR signaling. Taken together, our results indicate that expression of ULK1.DN can attenuate MPTP-induced axonal neurodegeneration, suggesting that ULK1 could be a promising novel target in the treatment of PD.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Dependovirus/metabolism , Genes, Dominant , Motor Activity , Neurons/enzymology , Neurons/pathology , Parkinson Disease/enzymology , Parkinson Disease/physiopathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Behavior, Animal , Cell Survival , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , HEK293 Cells , Humans , Male , Metabolome , Mice, Inbred C57BL , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
10.
J Med Chem ; 63(2): 490-511, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31518122

ABSTRACT

Injury to the adult central nervous system (CNS) usually leads to permanent deficits of cognitive, sensory, and/or motor functions. The failure of axonal regeneration in the damaged CNS limits functional recovery. The lack of information concerning the biological mechanism of axonal regeneration and its complexity has delayed the process of drug discovery for many years compared to other drug classes. Starting in the early 2000s, the ability of many molecules to stimulate axonal regrowth was evaluated through automated screening techniques; many hits and some new mechanisms involved in axonal regeneration were identified. In this Perspective, we discuss the rise of the CNS regenerative drugs, the main biological techniques used to test these drug candidates, some of the most important screens performed so far, and the main challenges following the identification of a drug that is able to induce axonal regeneration in vivo.


Subject(s)
Central Nervous System Agents/pharmacology , Nerve Regeneration/drug effects , Animals , Axons/drug effects , Central Nervous System/drug effects , Humans , Neurites/drug effects
11.
Front Cell Neurosci ; 11: 231, 2017.
Article in English | MEDLINE | ID: mdl-28824380

ABSTRACT

Limited axon regeneration in the injured adult mammalian central nervous system (CNS) usually results in irreversible functional deficits. Both the presence of extrinsic inhibitory molecules at the injury site and the intrinsically low capacity of adult neurons to grow axons are responsible for the diminished capacity of regeneration in the adult CNS. Conversely, in the embryonic CNS, neurons show a high regenerative capacity, mostly due to the expression of genes that positively control axon growth and downregulation of genes that inhibit axon growth. A better understanding of the role of these key genes controlling pro-regenerative mechanisms is pivotal to develop strategies to promote robust axon regeneration following adult CNS injury. Genetic manipulation techniques have been widely used to investigate the role of specific genes or a combination of different genes in axon regrowth. This review summarizes a myriad of studies that used genetic manipulations to promote axon growth in the injured CNS. We also review the roles of some of these genes during CNS development and suggest possible approaches to identify new candidate genes. Finally, we critically address the main advantages and pitfalls of gene-manipulation techniques, and discuss new strategies to promote robust axon regeneration in the mature CNS.

12.
Mol Neurobiol ; 54(1): 72-86, 2017 01.
Article in English | MEDLINE | ID: mdl-26732591

ABSTRACT

Axonal degeneration is one of the initial steps in many traumatic and neurodegenerative central nervous system (CNS) disorders and thus a promising therapeutic target. A focal axonal lesion is followed by acute axonal degeneration (AAD) of both adjacent axon parts, before proximal and distal parts follow different degenerative fates at later time points. Blocking calcium influx by calcium channel inhibitors was previously shown to attenuate AAD after optic nerve crush (ONC). However, it remains unclear whether the attenuation of AAD also promotes consecutive axonal regeneration. Here, we used a rat ONC model to study the effects of calcium channel inhibitors on axonal degeneration, retinal ganglion cell (RGC) survival, and axonal regeneration, as well as the molecular mechanisms involved. Application of calcium channel inhibitors attenuated AAD after ONC and preserved axonal integrity as visualized by live imaging of optic nerve axons. Consecutively, this resulted in improved survival of RGCs and improved axonal regeneration at 28 days after ONC. We show further that calcium channel inhibition attenuated lesion-induced calpain activation in the proximity of the crush and inhibited the activation of the c-Jun N-terminal kinase pathway. Pro-survival signaling via Akt in the retina was also increased. Our data thus show that attenuation of AAD improves consecutive neuronal survival and axonal regeneration and that calcium channel inhibitors could be valuable tools for therapeutic interventions in traumatic and degenerative CNS disorders.


Subject(s)
Axons/physiology , Calcium Channel Blockers/therapeutic use , Nerve Regeneration/physiology , Optic Nerve Injuries/prevention & control , Retinal Ganglion Cells/physiology , Animals , Axons/drug effects , Axons/pathology , Calcium Channel Blockers/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Female , Nerve Crush , Nerve Regeneration/drug effects , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve/physiology , Optic Nerve Injuries/pathology , Rats , Rats, Wistar , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
15.
J Neurochem ; 134(2): 261-75, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25807858

ABSTRACT

A lesion to the rat rubrospinal tract is a model for traumatic spinal cord lesions and results in atrophy of the red nucleus neurons, axonal dieback, and locomotor deficits. In this study, we used adeno-associated virus (AAV)-mediated over-expression of BAG1 and ROCK2-shRNA in the red nucleus to trace [by co-expression of enhanced green fluorescent protein (EGFP)] and treat the rubrospinal tract after unilateral dorsal hemisection. We investigated the effects of targeted gene therapy on neuronal survival, axonal sprouting of the rubrospinal tract, and motor recovery 12 weeks after unilateral dorsal hemisection at Th8 in rats. In addition to the evaluation of BAG1 and ROCK2 as therapeutic targets in spinal cord injury, we aimed to demonstrate the feasibility and the limits of an AAV-mediated protein over-expression versus AAV.shRNA-mediated down-regulation in this traumatic CNS lesion model. Our results demonstrate that BAG1 and ROCK2-shRNA both promote neuronal survival of red nucleus neurons and enhance axonal sprouting proximal to the lesion.


Subject(s)
DNA-Binding Proteins/biosynthesis , Nerve Regeneration/physiology , Neurons/pathology , Spinal Cord Injuries/pathology , Transcription Factors/biosynthesis , rho-Associated Kinases/biosynthesis , Animals , Axons , Base Sequence , Blotting, Western , Cell Survival , DNA-Binding Proteins/genetics , Dependovirus , Disease Models, Animal , Female , Genetic Therapy/methods , Genetic Vectors , Immunohistochemistry , Molecular Sequence Data , RNA, Small Interfering , Rats , Rats, Wistar , Recovery of Function , Red Nucleus/pathology , Transcription Factors/genetics , rho-Associated Kinases/genetics
16.
Neurobiol Dis ; 73: 150-62, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25283984

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder with prominent neuronal cell death in the substantia nigra (SN) and other parts of the brain. Previous studies in models of traumatic and neurodegenerative CNS disease showed that pharmacological inhibition of Rho-associated kinase (ROCK), a molecule involved in inhibitory signaling in the CNS, by small-molecule inhibitors improves neuronal survival and increases regeneration. Most small-molecule inhibitors, however, offer only limited target specificity and also inhibit other kinases, including both ROCK isoforms. To establish the role of the predominantly brain-expressed ROCK2 isoform in models of regeneration and PD, we used adeno-associated viral vectors (AAV) to specifically knockdown ROCK2 in neurons. Rat primary midbrain neurons (PMN) were transduced with AAV expressing short-hairpin-RNA (shRNA) against ROCK2 and LIM-domain kinase 1 (LIMK1), one of the downstream targets of ROCK2. While knock-down of ROCK2 and LIMK1 both enhanced neurite regeneration in a traumatic scratch lesion model, only ROCK2-shRNA protected PMN against 1-methyl-4-phenylpyridinium (MPP+) toxicity. Moreover, AAV.ROCK2-shRNA increased levels of the pro-survival markers Bcl-2 and phospho-Erk1. In vivo, AAV.ROCK2-shRNA vectors were injected into the ipsilateral SN and a unilateral 6-OHDA striatal lesion was performed. After four weeks, behavioral, immunohistochemical and biochemical alterations were investigated. Downregulation of ROCK2 protected dopaminergic neurons in the SN from 6-OHDA-induced degeneration and resulted in significantly increased TH-positive neuron numbers. This effect, however, was confined to nigral neuronal somata as striatal terminal density, dopamine and metabolite levels were not significantly preserved. Interestingly, motor behavior was improved in the ROCK2-shRNA treated animals compared to control after four weeks. Our studies thus confirm ROCK2 as a promising therapeutic target in models of PD and demonstrate that neuron-specific inhibition of ROCK2 promotes survival of lesioned dopaminergic neurons.


Subject(s)
Dopaminergic Neurons/metabolism , Down-Regulation/physiology , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Parkinson Disease/complications , rho-Associated Kinases/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adrenergic Agents/toxicity , Animals , Dependovirus/genetics , Disease Models, Animal , Down-Regulation/genetics , Genetic Vectors/physiology , Homovanillic Acid , Lim Kinases/genetics , Lim Kinases/metabolism , Oxidopamine/toxicity , Parkinson Disease/etiology , Psychomotor Performance , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase , rho-Associated Kinases/genetics
17.
Brain Pathol ; 25(2): 157-70, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25040536

ABSTRACT

Axonal degeneration is one of the initial steps in many neurological disorders and has been associated with increased autophagic activity. Although there are increasing data on the regulation of autophagy proteins in the neuronal soma after spinal cord injury (SCI), their characterization in the axon is scarce. Here, we examined the regulation of autophagy during axonal degeneration in a rat model of SCI following a lesion at Th 8. We analyzed the morphological and ultrastructural changes in injured axons by immunohistochemical evaluation of autophagy-related proteins and electron microscopy at different time points following SCI. The expression of ULK1, Atg7 and Atg5 in damaged axons was rapidly upregulated within hours after SCI. The number of axonal LC3-positive autophagosomes was also rapidly increased after SCI and remained at an increased level for up to 6 weeks. Ultrastructural analysis showed early signs of axonal degeneration and increased autophagy. In conclusion, we show that autophagy is increased early and for a sustained period in degenerating axons after SCI and that it might be an important executive step involved in axonal degeneration. Therefore, autophagy may represent a promising target for future therapeutic interventions in the treatment of axonal degeneration in traumatic central nervous system disorders.


Subject(s)
Autophagy , Axons/metabolism , Nerve Degeneration/metabolism , Spinal Cord Injuries/metabolism , Animals , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Autophagy-Related Protein-1 Homolog , Axons/ultrastructure , Female , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Proteins/metabolism , Rats , Rats, Wistar , Spinal Cord Injuries/pathology , Ubiquitin-Activating Enzymes/metabolism , Up-Regulation
18.
PLoS One ; 7(4): e34483, 2012.
Article in English | MEDLINE | ID: mdl-22496813

ABSTRACT

Most studies of c-Jun N-terminal Kinase (JNK) activation in retinal tissue were done in the context of neurodegeneration. In this study, we investigated the behavior of JNK during mitosis of progenitor cells in the retina of newborn rats. Retinal explants from newborn rats were kept in vitro for 3 hours and under distinct treatments. Sections of retinal explants or freshly fixed retinal tissue were used to detect JNK phosphorylation by immunohistochemistry, and were examined through both fluorescence and confocal microscopy. Mitotic cells were identified by chromatin morphology, histone-H3 phosphorylation, and location in the retinal tissue. The subcellular localization of proteins was analyzed by double staining with both a DNA marker and an antibody to each protein. Phosphorylation of JNK was also examined by western blot. The results showed that in the retina of newborn rats (P1), JNK is phosphorylated during mitosis of progenitor cells, mainly during the early stages of mitosis. JNK1 and/or JNK2 were preferentially phosphorylated in mitotic cells. Inhibition of JNK induced cell cycle arrest, specifically in mitosis. Treatment with the JNK inhibitor decreased the number of cells in anaphase, but did not alter the number of cells in either prophase/prometaphase or metaphase. Moreover, cells with aberrant chromatin morphology were found after treatment with the JNK inhibitor. The data show, for the first time, that JNK is activated in mitotic progenitor cells of developing retinal tissue, suggesting a new role of JNK in the control of progenitor cell proliferation in the retina.


Subject(s)
Gene Expression Regulation, Developmental , JNK Mitogen-Activated Protein Kinases/metabolism , Mitosis/physiology , Retina/cytology , Retina/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Animals, Newborn , Blotting, Western , Cell Cycle Checkpoints , Cell Proliferation , Immunoenzyme Techniques , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Rats , Rats, Inbred Strains
19.
Arq. bras. neurocir ; 26(3): 88-92, set. 2007. ilus
Article in Portuguese | LILACS | ID: lil-586458

ABSTRACT

Introdução: Estudos in vitro mostram que radioterapia e/ou quimioterapia podem ativar as vias de sinalizaçãodo receptor do fator de crescimento epidérmico (EGFR) e Ras, aumentando a resistência cruzada dascélulas de glioblastomas multiformes (GBM) ao tratamento. A inibição das atividades de EGFR e Rasatravés de inibidores tirosinas cinases elimina o antagonismo observado à administração seqüencialdestas modalidades terapêuticas, induzindo apoptose nestas células. Em estudo prévio demonstramosque o tratamento com o álcool perílico (AP), inibidor da farnesilação da Ras, induz apoptose em linhagenscelulares e células de explante de GBM. Objetivo: No presente estudo investigamos se a regressãoparcial observada em GBM recorrente de paciente tratado com administração intranasal de AP é mediadapor apoptose. Resultado: Ensaios com TUNEL (deoxynucleotidyl-mediated deoxyuridine triphosphate) ecaspase-3 ativada evidenciaram presença de células apoptóticas nas lâminas de GBM tratado. Conclusão:Esses achados sugerem que estratégias adjuvantes visando à inativação das vias de sinalização do EGFRe Ras podem melhorar tanto a eficácia de terapia isolada como de terapia multimodal em gliomas.


Background: In vitro studies demonstrated that both radiation and chemotherapy can activate EGFRand Ras signaling pathways, leading to increased cross-resistance to treatment of GBM cell. Inhibition of either EGFR or Ras activity with tytosine kinase inhibitor appears to abrogate the observed antagonism between sequentially administration of these therapeutic modalities inducing apoptosis in these cells. In a previous study, we demonstrated that in vitro treatment with perillyl alcohol (POH), an inhibitor of Ras farnezilation, induced apoptosis in human GBM cell lines and explants. Objective: In the presentstudy, we investigated if the partial regression observed in a patient with a recurrent GBM after treatmentby intranasal delivery of POH, is mediated by apoptosis. Result: Data from classical histology, terminaldeoxynucleotidyl-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) assay, as well asactivation of caspase 3, showed increased apoptosis in the treated tumor. Conclusion: These findings suggest that strategies to inactivate EGFR and RAS signaling may be critical to improving not only theefficacy of single-agent therapy but also of multimodal therapy in gliomas.


Subject(s)
Humans , Male , Middle Aged , Apoptosis , Glioma/surgery , Glioma/drug therapy , Glioma/radiotherapy , Immunohistochemistry , Monoterpenes/therapeutic use , Administration, Inhalation
SELECTION OF CITATIONS
SEARCH DETAIL