Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Fa Yi Xue Za Zhi ; 34(6): 635-639, 2018 Jun.
Article in English, Chinese | MEDLINE | ID: mdl-30896102

ABSTRACT

OBJECTIVES: To investigate the expression changes of the hydrogen sulfide synthases cystathionine γ-lyase (CSE), cystathionine ß-synthase (CBS), and 3-mercaptopyruvate sulfurtransferase (3-MST), after optic nerve crush (ONC) in rat the retina. METHODS: The rat model of ONC was established. Rats were divided into normal control, ONC, and sham control groups. Histopathologic changes in retina, the number of retinal ganglion cells (RGC) and retinal thickness of inner part (RTIP) were measured. The changes of CSE, CBS and 3-MST mRNA expression were detected with quantitative real-time PCR. RESULTS: The retinal histostructure was normal in normal controls and with minor changes in sham controls, respectively. Compared with sham group, significant retina damages were found in the ONC group: a time-dependent reduction of RGC number and RTIP. Expressions of CSE, CBS and 3-MST mRNA in rat retina were detected in normal control. Compared with normal controls, the expressions of CSE, CBS and 3-MST mRNA did not show any significant changes in the sham controls. Compared with sham controls, CBS mRNA expressions showed a time-dependent increase at 3 d, 7 d and 14 d after crush in the ONC group; CSE mRNA expressions increased to the peak at 3 d and then slightly reduced at 14 d after crush; 3-MST mRNA expressions showed the trend of increase at 3 d and 7 d and then enhanced remarkably at 14 d after crush. CONCLUSIONS: Hydrogen sulfide synthases CSE, CBS and 3-MST expressions were up-regulated in rat retina following ONC, which may cause an increase in local endogenous hydrogen sulfide production in the retina and a compensatory protective effect.


Subject(s)
Hydrogen Sulfide , Optic Nerve Injuries , Retina , Animals , Cystathionine beta-Synthase , Cystathionine gamma-Lyase , Hydrogen Sulfide/metabolism , Optic Nerve , Optic Nerve Injuries/enzymology , Rats , Retina/enzymology
2.
Cell Death Dis ; 8(6): e2847, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28569783

ABSTRACT

Damage-induced neuronal endopeptidase (DINE)/endothelin-converting enzyme-like 1 (ECEL1) is a membrane-bound metalloprotease that we identified as a nerve regeneration-associated molecule. The expression of DINE is upregulated in response to nerve injury in both the peripheral and central nervous systems, while its transcription is regulated by the activating transcription factor 3 (ATF3), a potent hub-transcription factor for nerve regeneration. Despite its unique hallmark of injury-induced upregulation, the physiological relevance of DINE in injured neurons has been unclear. In this study, we have demonstrated that the expression of DINE is upregulated in injured retinal ganglion cells (RGCs) in a coordinated manner with that of ATF3 after optic nerve injury, whereas DINE and ATF3 are not observed in any normal retinal cells. Recently, we have generated a mature DINE-deficient (KOTg) mouse, in which exogenous DINE is overexpressed specifically in embryonic motor neurons to avoid aberrant arborization of motor nerves and lethality after birth that occurs in the conventional DINE KO mouse. The DINE KOTg mice did not show any difference in retinal structure and the projection to brain from that of wild-type (wild type) mice under normal conditions. However, injured RGCs of DINE KOTg mice failed to regenerate even after the zymosan treatment, which is a well-known regeneration-promoting reagent. Furthermore, a DINE KOTg mouse crossed with a Atf3:BAC Tg mouse, in which green fluorescent protein (GFP) is visualized specifically in injured RGCs and optic nerves, has verified that DINE deficiency leads to regeneration failure. These findings suggest that injury-induced DINE is a crucial endopeptidase for injured RGCs to promote axonal regeneration after optic nerve injury. Thus, a DINE-mediated proteolytic mechanism would provide us with a new therapeutic strategy for nerve regeneration.


Subject(s)
Activating Transcription Factor 3/genetics , Metalloendopeptidases/genetics , Nerve Regeneration/genetics , Optic Nerve Injuries/genetics , Retinal Ganglion Cells/enzymology , Activating Transcription Factor 3/metabolism , Animals , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Metalloendopeptidases/deficiency , Mice , Mice, Knockout , Neuroprotective Agents/pharmacology , Optic Nerve/drug effects , Optic Nerve/enzymology , Optic Nerve/pathology , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Signal Transduction , Zymosan/pharmacology
3.
eNeuro ; 3(6)2016.
Article in English | MEDLINE | ID: mdl-28032120

ABSTRACT

After traumatic damage of the brain or spinal cord, many surviving neurons are disconnected, and recovery of function is limited by poor axon regeneration. Recent data have suggested that poly ADP-ribosylation plays a role in limiting axonal regrowth such that inhibition of poly (ADP-ribose) polymerase (PARP) may have therapeutic efficacy for neurological recovery after trauma. Here, we tested systemic administration of the PARP inhibitor, veliparib, and showed effective suppression of PARylation in the mouse CNS. After optic nerve crush injury or dorsal hemisection of the thoracic spinal cord in mice, treatment with veliparib at doses with pharmacodynamic action had no benefit for axonal regeneration or functional recovery. We considered whether PARP gene family specificity might play a role. In vitro mouse cerebral cortex axon regeneration experiments revealed that short hairpin RNA (shRNA)-mediated suppression of PARP1 promoted axonal regeneration, whereas suppression of other PARP isoforms either had no effect or decreased regeneration. Therefore, we examined recovery from neurological trauma in mice lacking PARP1. No increase of axonal regeneration was observed in Parp1-/- mice after optic nerve crush injury or dorsal hemisection of the thoracic spinal cord, and there was no improvement in motor function recovery. Thus, comprehensive in vivo analysis reveals no indication that clinical PARP inhibitors will on their own provide benefit for recovery from CNS trauma.


Subject(s)
Axons/drug effects , Benzimidazoles/pharmacology , Nerve Regeneration/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Recovery of Function/drug effects , Animals , Axons/enzymology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Disease Models, Animal , Female , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Nerve Regeneration/physiology , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/pathology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Recovery of Function/physiology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/pathology , Thoracic Vertebrae
4.
Mol Neurodegener ; 10: 40, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26306916

ABSTRACT

BACKGROUND: Axonal injury of the optic nerve (ON) is involved in various ocular diseases, such as glaucoma and traumatic optic neuropathy, which leads to apoptotic death of retinal ganglion cells (RGCs) and loss of vision. Caspases have been implicated in RGC pathogenesis. However, the role of caspase-7, a functionally unique caspase, in ON injury and RGC apoptosis has not been reported previously. The purpose of this study is to evaluate the role of caspase-7 in ON injury-induced RGC apoptosis. RESULTS: C57BL/6 (wildtype, WT) and caspase-7 knockout (Casp7(-/-)) mice were used. We show that ON crush activated caspase-7 and calpain-1, an upstream activator of caspase-7, in mouse RGCs, as well as hydrolysis of kinectin and co-chaperone P23, specific substrates of caspase-7. ON crush caused a progressive loss of RGCs to 28 days after injury. Knockout of caspase-7 partially and significantly protected against the ON injury-induced RGC loss; RGC density at 28 days post ON crush in Casp7(-/-) mice was approximately twice of that in WT ON injured retinas. Consistent with changes in RGC counts, spectral-domain optical coherence tomography analysis revealed that ON crush significantly reduced the in vivo thickness of the ganglion cell complex layer (including ganglion cell layer, nerve fiber layer, and inner plexiform layer) in the retina. The ON crush-induced thinning of retinal layer was significantly ameliorated in Casp7(-/-) mice when compared to WT mice. Moreover, electroretinography analysis demonstrated a decline in the positive component of scotopic threshold response amplitude in ON crushed eyes of the WT mice, whereas this RGC functional response was significantly higher in Casp7(-/-) mice at 28 days post injury. CONCLUSION: Altogether, our findings indicate that caspase-7 plays a critical role in ON injury-induced RGC death, and inhibition of caspase-7 activity may be a novel therapeutic strategy for glaucoma and other neurodegenerative diseases of the retina.


Subject(s)
Caspase 7/physiology , Eye Proteins/physiology , Optic Nerve Injuries/enzymology , Retinal Ganglion Cells/pathology , Animals , Apoptosis , Calpain/metabolism , Caspase 7/deficiency , Caspase 7/genetics , Cell Count , Cytoplasm/enzymology , Electroretinography , Enzyme Activation , Enzyme Induction , Eye Proteins/genetics , Female , Intramolecular Oxidoreductases/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Crush , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , Prostaglandin-E Synthases , Retinal Ganglion Cells/enzymology , Retinal Ganglion Cells/physiology , Tomography, Optical Coherence
5.
ASN Neuro ; 7(3)2015.
Article in English | MEDLINE | ID: mdl-26129908

ABSTRACT

Histones deacetylases (HDACs), besides their function as epigenetic regulators, deacetylate and critically regulate the activity of nonhistone targets. In particular, HDACs control partially the proapoptotic activity of p53 by balancing its acetylation state. HDAC inhibitors have revealed neuroprotective properties in different models, but the exact mechanisms of action remain poorly understood. We have generated a conditional knockout mouse model targeting retinal ganglion cells (RGCs) to investigate specifically the functional role of HDAC1 and HDAC2 in an acute model of optic nerve injury. Our results demonstrate that combined HDAC1 and HDAC2 ablation promotes survival of axotomized RGCs. Based on global gene expression analyses, we identified the p53-PUMA apoptosis-inducing axis to be strongly activated in axotomized mouse RGCs. Specific HDAC1/2 ablation inhibited this apoptotic pathway by impairing the crucial acetylation status of p53 and reducing PUMA expression, thereby contributing to the ensuing enhanced neuroprotection due to HDAC1/2 depletion. HDAC1/2 inhibition and the affected downstream signaling components emerge as specific targets for developing therapeutic strategies in neuroprotection.


Subject(s)
Cell Survival/physiology , Genes, p53 , Histone Deacetylase 1/deficiency , Histone Deacetylase 2/deficiency , Neuroprotection , Optic Nerve Injuries/enzymology , Retinal Ganglion Cells/enzymology , Retinal Ganglion Cells/physiology , Acetylation , Acute Disease , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Axotomy , Disease Models, Animal , Gene Expression Regulation , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , MAP Kinase Signaling System , Mice, Knockout , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Int J Clin Exp Pathol ; 8(11): 14717-24, 2015.
Article in English | MEDLINE | ID: mdl-26823796

ABSTRACT

OBJECTIVE: The aim of this study was to study the impacts of Rho kinase inhibitor Fasudil on expressions of Rho/ROCK signaling pathway associated genes in rabbits with optic nerve injury (ONI), and to explore the therapeutic mechanisms towards ONI. METHODS: The rabbit ONI model was established, then the rabbits were divided into model group (treated with saline), control group (treated with dexamethasone, Dex), and intervention group (treated with Fasudil, Fas). The eyeball and optic nerve were sampled at 3, 7, 14 and 21 days after injury. The morphological changes of retina and optic nerve were observed. The expressions of RhoA, Caspase-3, Rock 2 and Nogo-A gene were determined by immunohistochemistry and real-time polymerase chain reaction (RT-PCR) methods. RESULTS: At different time after injury, there were significant differences of RhoA, Caspase-3, Rock 2 and Nogo-A gene expression among three groups (P < 0.05). CONCLUSIONS: After ONI, Fas can decrease the expression of Caspase-3 gene, and down-regulate the expressions of Nogo-A and Rock 2 gene. Therefore, it can treat ONI through affecting the Rho/ROCK signaling pathway.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/pathology , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Disease Models, Animal , Female , Gene Expression/drug effects , Immunohistochemistry , Male , Rabbits , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Mol Neurodegener ; 9: 39, 2014 Sep 28.
Article in English | MEDLINE | ID: mdl-25261965

ABSTRACT

BACKGROUND: Optic nerve damage initiates a series of early atrophic events in retinal ganglion cells (RGCs) that precede the BAX-dependent committed step of the intrinsic apoptotic program. Nuclear atrophy, including global histone deacetylation, heterochromatin formation, shrinkage and collapse of nuclear structure, and the silencing of normal gene expression, comprise an important obstacle to overcome in therapeutic approaches to preserve neuronal function. Several studies have implicated histone deacetylases (HDACs) in the early stages of neuronal cell death, including RGCs. Importantly, these neurons exhibit nuclear translocation of HDAC3 shortly after optic nerve damage. Additionally, HDAC3 activity has been reported to be selectively toxic to neurons. RESULTS: RGC-specific conditional knockout of Hdac3 was achieved by transducing the RGCs of Hdac3fl/fl mice with an adeno-associated virus serotype 2 carrying CRE recombinase and GFP (AAV2-Cre/GFP). Controls included similar viral transduction of Rosa26fl/fl reporter mice. Optic nerve crush (ONC) was then performed on eyes. The ablation of Hdac3 in RGCs resulted in significant amelioration of characteristics of ONC-induced nuclear atrophy such as H4 deacetylation, heterochromatin formation, and the loss of nuclear structure. RGC death was also significantly reduced. Interestingly, loss of Hdac3 expression did not lead to protection against RGC-specific gene silencing after ONC, although this effect was achieved using the broad spectrum inhibitor, Trichostatin A. CONCLUSION: Although other HDACs may be responsible for gene expression changes in RGCs, our results indicate a critical role for HDAC3 in nuclear atrophy in RGC apoptosis following axonal injury. This study provides a framework for studying the roles of other prevalent retinal HDACs in neuronal death as a result of axonal injury.


Subject(s)
Apoptosis/physiology , Histone Deacetylases/metabolism , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/enzymology , Animals , Blotting, Western , Female , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Real-Time Polymerase Chain Reaction , Retinal Ganglion Cells/pathology
8.
Neurosci Lett ; 581: 89-93, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25172145

ABSTRACT

The dedicator of cytokinesis 3 (Dock3) is an atypical guanine nucleotide exchange factor that is predominantly expressed in the CNS. Dock3 exerts neuroprotective effects and stimulates optic nerve regeneration. The p38 mitogen-activated protein kinase acts downstream of apoptosis signal-regulating kinase 1 (ASK1) signaling and plays an important role in neural cell death. We assessed a therapeutic efficacy of Dock3 stimulation and p38 inhibition in retinal degeneration induced by optic nerve injury (ONI). In vivo retinal imaging using optical coherence tomography revealed that ONI-induced retinal degeneration was ameliorated in SB203580 (a p38 inhibitor)-treated WT mice and PBS-treated Dock3 overexpressing (Dock3 Tg) mice, and SB203580 further stimulated retinal protection in Dock3 Tg mice. In addition, SB203580 increased the number of regenerating axons after ONI in both WT and Dock3 Tg mice. ONI-induced phosphorylation of ASK1, p38 and the N-methyl-d-aspartate receptor 2B subunit were suppressed in the retina of Dock3 Tg mice. Inhibition of the ASK1 pathway in Dock3 Tg mice suggests that Dock3 may have an antioxidant-like property. These results indicate that overexpression of Dock3 and pharmacological interruption of p38 have synergistic effects for both neuroprotection and axon regeneration, thus combined application may be beneficial for the treatment of ONI.


Subject(s)
Axons/physiology , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Optic Nerve Injuries/metabolism , Regeneration/physiology , Retinal Ganglion Cells/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Carrier Proteins/therapeutic use , Cell Count , Enzyme Inhibitors/pharmacology , Guanine Nucleotide Exchange Factors , Imidazoles/pharmacology , Mice , Mice, Transgenic , Nerve Tissue Proteins/therapeutic use , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/therapy , Phosphorylation , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/therapeutic use
9.
Neurobiol Dis ; 69: 108-16, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24878510

ABSTRACT

Injury to retinal ganglion cell (RGC) axons triggers rapid activation of Jun N-terminal kinase (JNK) signaling, a major prodeath pathway in injured RGCs. Of the multiple kinases that can activate JNK, dual leucine kinase (Dlk) is known to regulate both apoptosis and Wallerian degeneration triggered by axonal insult. Here we tested the importance of Dlk in regulating somal and axonal degeneration of RGCs following axonal injury. Removal of DLK from the developing optic cup did not grossly affect developmental RGC death or inner plexiform layer organization. In the adult, Dlk deficiency significantly delayed axonal-injury induced RGC death. The activation of JUN was also attenuated in Dlk deficient retinas. Dlk deficiency attenuated the activation of the somal pool of JNK but did not prevent activation of the axonal pool of JNK after axonal injury, indicating that JNK activation in different cellular compartments of an RGC following axonal injury is regulated by distinct upstream kinases. In contrast to its robust influence on somal degeneration, Dlk deficiency did not alter RGC axonal degeneration after axonal injury as assessed using physiological readouts of optic nerve function.


Subject(s)
Axons/enzymology , MAP Kinase Kinase Kinases/deficiency , Optic Nerve Injuries/enzymology , Retinal Ganglion Cells/enzymology , Wallerian Degeneration/enzymology , Animals , Axons/pathology , Cell Death/physiology , Cell Survival/physiology , Disease Models, Animal , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Mice, Transgenic , Optic Nerve Injuries/pathology , Phosphorylation/physiology , Retina/enzymology , Retina/growth & development , Retina/pathology , Retinal Ganglion Cells/pathology , Signal Transduction , Tissue Culture Techniques , Wallerian Degeneration/pathology
10.
Cell Death Dis ; 5: e1225, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24832597

ABSTRACT

The Rho/ROCK/LIMK pathway is central for the mediation of repulsive environmental signals in the central nervous system. Several studies using pharmacological Rho-associated protein kinase (ROCK) inhibitors have shown positive effects on neurite regeneration and suggest additional pro-survival effects in neurons. However, as none of these drugs is completely target specific, it remains unclear how these effects are mediated and whether ROCK is really the most relevant target of the pathway. To answer these questions, we generated adeno-associated viral vectors to specifically downregulate ROCK2 and LIM domain kinase (LIMK)-1 in rat retinal ganglion cells (RGCs) in vitro and in vivo. We show here that specific knockdown of ROCK2 and LIMK1 equally enhanced neurite outgrowth of RGCs on inhibitory substrates and both induced substantial neuronal regeneration over distances of more than 5 mm after rat optic nerve crush (ONC) in vivo. However, only knockdown of ROCK2 but not LIMK1 increased survival of RGCs after optic nerve axotomy. Moreover, knockdown of ROCK2 attenuated axonal degeneration of the proximal axon after ONC assessed by in vivo live imaging. Mechanistically, we demonstrate here that knockdown of ROCK2 resulted in decreased intraneuronal activity of calpain and caspase 3, whereas levels of pAkt and collapsin response mediator protein 2 and autophagic flux were increased. Taken together, our data characterize ROCK2 as a specific therapeutic target in neurodegenerative diseases and demonstrate new downstream effects of ROCK2 including axonal degeneration, apoptosis and autophagy.


Subject(s)
Nerve Degeneration , Nerve Regeneration , Optic Nerve Injuries/enzymology , Optic Nerve/enzymology , Retinal Ganglion Cells/enzymology , rho-Associated Kinases/metabolism , Animals , Apoptosis , Autophagy , Axons/enzymology , Axons/pathology , Calpain/metabolism , Caspase 3/metabolism , Cell Death , Cells, Cultured , Dependovirus/genetics , Disease Models, Animal , Female , Gene Knockdown Techniques , Gene Transfer Techniques , Genetic Vectors , Intercellular Signaling Peptides and Proteins/metabolism , Lim Kinases/genetics , Lim Kinases/metabolism , Nerve Crush , Nerve Tissue Proteins/metabolism , Neurites/enzymology , Neurites/pathology , Optic Nerve/pathology , Optic Nerve/physiopathology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rats , Rats, Wistar , Retinal Ganglion Cells/pathology , Signal Transduction , Time Factors , Transfection , rho-Associated Kinases/genetics
11.
Exp Cell Res ; 319(15): 2384-94, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23830879

ABSTRACT

Rho GTPases are molecular switches that modulate multiple intracellular signaling processes by means of various effector proteins. As a result, Rho GTPase activities are tightly spatiotemporally regulated in order to ensure homeostasis within the cell. Though the roles of Rho GTPases during neural development have been well documented, their participation during neurodegeneration has been far less characterized. Herein we discuss our current knowledge of the role and function of Rho GTPases and regulators during neurodegeneration, and highlight their potential as targets for therapeutic intervention in common neurodegenerative disorders.


Subject(s)
Gene Expression Regulation , Neurites/metabolism , Neurodegenerative Diseases/genetics , Signal Transduction , rho GTP-Binding Proteins/metabolism , Animals , Humans , Isoenzymes/classification , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Nerve Regeneration/physiology , Neurites/pathology , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Neurogenesis/genetics , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/genetics , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/genetics , rho GTP-Binding Proteins/classification , rho GTP-Binding Proteins/genetics
12.
Proc Natl Acad Sci U S A ; 110(10): 4045-50, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23431148

ABSTRACT

Glaucoma, a major cause of blindness worldwide, is a neurodegenerative optic neuropathy in which vision loss is caused by loss of retinal ganglion cells (RGCs). To better define the pathways mediating RGC death and identify targets for the development of neuroprotective drugs, we developed a high-throughput RNA interference screen with primary RGCs and used it to screen the full mouse kinome. The screen identified dual leucine zipper kinase (DLK) as a key neuroprotective target in RGCs. In cultured RGCs, DLK signaling is both necessary and sufficient for cell death. DLK undergoes robust posttranscriptional up-regulation in response to axonal injury in vitro and in vivo. Using a conditional knockout approach, we confirmed that DLK is required for RGC JNK activation and cell death in a rodent model of optic neuropathy. In addition, tozasertib, a small molecule protein kinase inhibitor with activity against DLK, protects RGCs from cell death in rodent glaucoma and traumatic optic neuropathy models. Together, our results establish a previously undescribed drug/drug target combination in glaucoma, identify an early marker of RGC injury, and provide a starting point for the development of more specific neuroprotective DLK inhibitors for the treatment of glaucoma, nonglaucomatous forms of optic neuropathy, and perhaps other CNS neurodegenerations.


Subject(s)
MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/physiology , Retinal Ganglion Cells/enzymology , Retinal Ganglion Cells/pathology , Animals , Cell Death/genetics , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Down-Regulation , Glaucoma/drug therapy , Glaucoma/etiology , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Mice , Optic Nerve Diseases/etiology , Optic Nerve Diseases/pathology , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/pathology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA Interference , Rats , Rats, Wistar , Retinal Ganglion Cells/drug effects , Signal Transduction , Up-Regulation
13.
Neurobiol Dis ; 46(2): 393-401, 2012 May.
Article in English | MEDLINE | ID: mdl-22353563

ABSTRACT

Glaucoma is a neurodegenerative disease characterized by the apoptotic death of retinal ganglion cells (RGCs). The primary insult to RGCs in glaucoma is thought to occur to their axons as they exit the eye in the optic nerve head. However, pathological signaling pathways that exert central roles in triggering RGC death following axonal injury remain unidentified. It is likely that the first changes to occur following axonal injury are signal relay events that transduce the injury signal from the axon to the cell body. Here we focus on the c-Jun N-terminal kinase (JNK1-3) family, a signaling pathway implicated in axonal injury signaling and neurodegenerative apoptosis, and likely to function as a central node in axonal injury-induced RGC death. We show that JNK signaling is activated immediately after axonal injury in RGC axons at the site of injury. Following its early activation, sustained JNK signaling is observed in axonally-injured RGCs in the form of JUN phosphorylation and upregulation. Using mice lacking specific Jnk isoforms, we show that Jnk2 and Jnk3 are the isoforms activated in injured axons. Combined deficiency of Jnk2 and Jnk3 provides robust long-term protection against axonal injury-induced RGC death and prevents downregulation of the RGC marker, BRN3B, and phosphorylation of JUN. Finally, using Jun deficient mice, we show that JUN-dependent pathways are important for axonal injury-induced RGC death. Together these data demonstrate that JNK signaling is the major early pathway triggering RGC death after axonal injury and may directly link axon injury to transcriptional activity that controls RGC death.


Subject(s)
Axons/enzymology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 10/physiology , Mitogen-Activated Protein Kinase 9/physiology , Retinal Ganglion Cells/enzymology , Animals , Axons/pathology , Cell Death , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/pathology , Transcriptional Activation/physiology
14.
J Neurosci ; 29(30): 9545-52, 2009 Jul 29.
Article in English | MEDLINE | ID: mdl-19641117

ABSTRACT

Adult spinal axons do not spontaneously regenerate after injury. However, if the peripheral branch of dorsal root ganglion neurons is lesioned before lesioning the central branch of the same neurons in the dorsal column, these central axons will regenerate and, if cultured, are not inhibited from extending neurites by myelin-associated inhibitors of regeneration such as myelin-associated glycoprotein (MAG). This effect can be mimicked by elevating cAMP and is transcription dependent. The ability of cAMP to overcome inhibition by MAG in culture involves the upregulation of the enzyme arginase I (Arg I) and subsequent increase in synthesis of polyamines such as putrescine. Now we show that a peripheral lesion also induces an increase in Arg I expression and synthesis of polyamines. We also show that the conditioning lesion effect in overcoming inhibition by MAG is initially dependent on ongoing polyamine synthesis but, with time after lesion, becomes independent of ongoing synthesis. However, if synthesis of polyamines is blocked in vivo the early phase of good growth after a conditioning lesion is completely blocked and the later phase of growth, when ongoing polyamine synthesis is not required during culture, is attenuated. We also show that putrescine must be converted to spermidine both in culture and in vivo to overcome inhibition by MAG and that spermidine can promote optic nerve regeneration in vivo. These results suggest that spermidine could be a useful tool in promoting CNS axon regeneration after injury.


Subject(s)
Arginase/metabolism , Axons/physiology , Nerve Regeneration/physiology , Spermidine/metabolism , Animals , Axons/enzymology , Cells, Cultured , Ganglia, Spinal/enzymology , Ganglia, Spinal/physiology , Male , Myelin Sheath/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Crush , Neurons/enzymology , Neurons/physiology , Optic Nerve/enzymology , Optic Nerve/physiology , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/physiopathology , Polyamines/metabolism , Putrescine/metabolism , Rats , Rats, Inbred F344 , Sciatic Nerve/enzymology , Sciatic Nerve/injuries , Sciatic Nerve/physiology , Signal Transduction/drug effects , Up-Regulation
15.
Exp Eye Res ; 89(5): 665-77, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19560459

ABSTRACT

Granulocyte-macrophage-colony-stimulating-factor (GM-CSF) is a potent hematopoietic cytokine. In the present study, we examined whether GM-CSF is neuroprotective in retinal ganglion cells (RGCs). First, we studied the expression of GM-CSF and the GM-CSF-alpha-receptor in rat and human retina and in RGC-5 cells. Then, RGC-5 cells were incubated with apoptosis-inducing agents (e.g., staurosporine, glutamate and NOR3). The cell death was assessed by Live-Death-Assays and apoptosis-related-proteins were examined by immunoblotting. In addition, the expression of phosphorylated ERK1/2-pathway-proteins after incubation with GM-CSF and after inhibiting MEK1/2 with U0126 was analyzed. To assess the in vivo-effect, first staurosporine or GM-CSF plus staurosporine was injected into the vitreous body of Sprague-Dawley rats. In a second axotomy model the optic nerve was cut and GM-CSF was injected into the vitreous body. In both models, the RGCs were labeled retrogradely with either Fluoro-Gold or 4-Di-10-Asp and counted. As a first result, we identified GM-CSF and the GM-CSF-alpha-receptor in rat and human retina as well as in RGC-5 cells. Then, in the RGC-5 cells GM-CSF counteracts induced cell death in a dose-and time-dependent manner. With respect to apoptosis, Western blot analysis revealed a decreased Bad-expression and an increased Bcl-2-expression after co-incubation with GM-CSF. Concerning signaling pathways, incubation with GM-CSF activates the ERK1/2 pathway, whereas inhibition of MEK1/2 with U0126 strongly decreased the phosphorylation downstream in the ERK1/2 pathway, and the antiapoptotic activity of GM-CSF in vitro. Like in vitro, GM-CSF counteracts the staurosporine-induced cell death in vivo and protects RGCs from axotomy-induced degeneration. Our data suggest that GM-CSF might be a novel therapeutic agent in neuropathic disease of the eye.


Subject(s)
Apoptosis , Glaucoma/enzymology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Optic Nerve Injuries/enzymology , Retinal Ganglion Cells/enzymology , Adult , Aged , Animals , Apoptosis/drug effects , Blotting, Western , Butadienes/pharmacology , Cells, Cultured , Disease Models, Animal , Glaucoma/pathology , Glutamic Acid/toxicity , Humans , Hydroxylamines/toxicity , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Neuroanatomical Tract-Tracing Techniques , Nitriles/pharmacology , Nitro Compounds , Optic Nerve Injuries/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Staurosporine/toxicity , bcl-Associated Death Protein/metabolism
16.
Brain Res ; 1201: 23-33, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18313036

ABSTRACT

A ROCK inhibitor Fasudil is widely administered to relieve vasospasm in patients after subarachnoid hemorrhage in Japan. We investigated the difference of Fasudil and Y-27632, a common ROCK inhibitor, on neurite regeneration in culture and axonal regeneration after injuring the optic nerve (OpN) in cats. The optimal dose of Y-27632, determined by counting the number and length of neurites in retinal explants, was found to be 100 microM: the only effect of Fasudil was to promote extension of glial processes. We next examined the effects of Fasudil (10 microM-100 microM) and Y-27632 (10 microM-300 microM) on axonal regeneration in the crushed OpN model in vivo. Immediately after crushing the left OpN, Fasudil or Y-27632 was injected into the vitreous and the crushed site. Injection of 10 microM and 100 microM Y-27632 induced extension of the optic axons beyond the crush site, with the latter dosage giving stronger regeneration. Very few axons passed beyond the crush site in the optic nerve with phosphate-buffered saline injection, and no axons elongated in the OpN with Fasudil injection.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Amides/pharmacology , Nerve Regeneration/drug effects , Optic Nerve Injuries/drug therapy , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Amides/therapeutic use , Animals , Axons/drug effects , Axons/enzymology , Axons/pathology , Cats , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Male , Nerve Crush , Nerve Regeneration/physiology , Optic Nerve/drug effects , Optic Nerve/enzymology , Optic Nerve/physiopathology , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/physiopathology , Organ Culture Techniques , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Retina/cytology , Retina/drug effects , Treatment Outcome , Wallerian Degeneration/drug therapy , Wallerian Degeneration/enzymology , Wallerian Degeneration/physiopathology , rho-Associated Kinases/metabolism
17.
J Neurochem ; 103(1): 181-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17608642

ABSTRACT

Inhibitory molecules derived from CNS myelin and glial scar tissue are major causes for insufficient functional regeneration in the mammalian CNS. A multitude of these molecules signal through the Rho/Rho kinase (ROCK) pathway. We evaluated three inhibitors of ROCK, Y- 27632, Fasudil (HA-1077), and Dimethylfasudil (H-1152), in models of neurite outgrowth in vitro. We show, that all three ROCK inhibitors partially restore neurite outgrowth of Ntera-2 neurons on the inhibitory chondroitin sulphate proteoglycan substrate. In the rat optic nerve crush model Y-27632 dose-dependently increased regeneration of retinal ganglion cell axons in vivo. Application of Dimethylfasudil showed a trend towards increased axonal regeneration in an intermediate concentration. We demonstrate that inhibition of ROCK can be an effective therapeutic approach to increase regeneration of CNS neurons. The selection of a suitable inhibitor with a broad therapeutic window, however, is crucial in order to minimize unwanted side effects and to avoid deleterious effects on nerve fiber growth.


Subject(s)
Chondroitin Sulfate Proteoglycans/pharmacology , Enzyme Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Nerve Regeneration/drug effects , Neurites/drug effects , Optic Nerve Injuries/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Amides/pharmacology , Animals , Axons/drug effects , Axons/pathology , Cell Line , Dose-Response Relationship, Drug , Female , Humans , Nerve Crush , Neurites/physiology , Neurons/drug effects , Neurons/pathology , Optic Nerve/drug effects , Optic Nerve/enzymology , Optic Nerve/pathology , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/pathology , Pyridines/pharmacology , Rats , Rats, Wistar , rho-Associated Kinases
18.
Brain Res ; 1139: 68-84, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17280650

ABSTRACT

We investigated the effects of neonatal optic nerve transection on cortical acetylcholinesterase (AChE) activity in hooded rats during postnatal development and following behavioral manipulation after weaning. AChE reaction product was quantified on digitized images of histochemically stained sections in layer IV of primary somatic sensory, primary visual and visual association cortex. Rats with optic nerve transection were compared to sham-operated littermates. In all cortical regions of both types of animal, AChE reaction product was increased to peak 2 weeks after birth and decreased thereafter, reaching adult levels at the end of the third postnatal week. During postnatal development, reaction product in primary visual cortex was lower in rats deprived of retinal input than in sham-operated littermates and the area delineated by reaction product was smaller. However, optic nerve transection did not modify the time course of postnatal development or statistically significantly diminish adult levels of AChE activity. Behavioral manipulations after weaning statistically significantly increased enzyme activity in sham-operated rats in all cortical areas examined. Compared with cage rearing, training in a discrimination task with food reward had a greater impact than environmental enrichment. By contrast, in the rats with optic nerve transection enrichment and training resulted in statistically significantly increased AChE activity only in lateral visual association cortex. Our findings provide evidence for intra- and supramodal influences of the neonatal removal of retinal input on neural activity- and use-dependent modifications of cortical AChE activity. The laminar distribution of the AChE reaction product suggests that the observed changes in AChE activity were mainly related to cholinergic basal forebrain afferents. These afferents may facilitate the stabilization of transient connections between the somatic sensory and the visual pathway.


Subject(s)
Acetylcholinesterase/metabolism , Neocortex/enzymology , Neuronal Plasticity/physiology , Optic Nerve Injuries/enzymology , Visual Pathways/enzymology , Animals , Arousal/physiology , Cholinergic Fibers/enzymology , Discrimination Learning/physiology , Environment , Female , Male , Neocortex/cytology , Neocortex/growth & development , Neurons/enzymology , Optic Nerve Injuries/physiopathology , Rats , Rats, Long-Evans , Visual Pathways/cytology , Visual Pathways/growth & development
19.
Neuroscience ; 142(4): 1081-92, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16997488

ABSTRACT

Fish CNS neurons can repair their axons following nerve injury, whereas mammalian CNS neurons cannot regenerate, and become apoptotic within 1-2 weeks after the nerve lesion. One explanation for these differences is that one, or several molecules are upregulated in fish CNS neurons during nerve regeneration, and this same molecule is downregulated in mammalian CNS neurons before the development of apoptosis caused by nerve injury. A molecule satisfying these criteria might successfully rescue and repair the mammalian CNS neurons. In this study, we looked for such a candidate molecule from goldfish retinas. Transglutaminase derived from goldfish retina (TG(R)) was characterized as a regenerating molecule after optic nerve injury. A full-length cDNA for TG(R) was isolated from the goldfish retinal cDNA library prepared from axotomized retinas. Levels of TG(R) mRNA and protein increased only in the retinal ganglion cells (RGCs) between 10 and 40 days after optic nerve transection. Recombinant TG(R) protein enhanced neurite outgrowth from adult fish RGCs in culture. Specific interference RNA and antibodies for TG(R) inhibited neurite outgrowth both in vitro and in vivo. In contrast, the level of TG(R) protein decreased in rat RGCs within 1-3 days after nerve injury. Furthermore, the addition of recombinant TG(R) to retinal cultures induced striking neurite outgrowth from adult rat RGCs. These molecular and cellular data strongly suggest that TG(R) promotes axonal elongation at the surface of injured RGCs after optic nerve injury.


Subject(s)
Growth Cones/enzymology , Nerve Regeneration/physiology , Optic Nerve Injuries/enzymology , Optic Nerve/enzymology , Retinal Ganglion Cells/enzymology , Transglutaminases/metabolism , Animals , Cells, Cultured , DNA, Complementary/analysis , DNA, Complementary/genetics , Disease Models, Animal , Gene Library , Goldfish , Growth Cones/drug effects , Growth Cones/ultrastructure , Nerve Regeneration/drug effects , Neurites/drug effects , Neurites/enzymology , Optic Nerve/drug effects , Optic Nerve/physiopathology , Optic Nerve Injuries/physiopathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Retinal Ganglion Cells/drug effects , Transglutaminases/genetics , Transglutaminases/pharmacology , Up-Regulation/physiology
20.
J Neurochem ; 93(6): 1422-33, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15935058

ABSTRACT

We have shown that application of basic fibroblast growth factor (FGF-2) to axotomized optic nerve promotes the survival of frog retinal ganglion cells (RGCs). In the present study we used western blotting and immunocytochemistry to investigate the effects of this FGF-2 treatment upon the activation of the extracellular signal-regulated kinase (ERK) pathway, the amounts and distribution of Bcl-2 family proteins, and the activation of caspase-3. Axotomy alone temporarily increased ERK activation; FGF-2 treatment to the nerve prolonged this activation. This effect was blocked by U0126, a selective ERK kinase (MEK) inhibitor. Axotomy caused a decrease in Bcl-2 and a small increase in Bcl-x(L). FGF-2 treatment caused an ERK-dependent increase in Bcl-2 and an ERK-independent increase in Bcl-x(L). The pro-apoptotic Bax was increased by axotomy; FGF-2 treatment greatly decreased Bax levels, an effect that was inhibited by U0126. Axotomy induced the cleavage of caspase-3; FGF-2 treatment blocked this effect in an ERK-dependent manner. Finally, intraocular application of the MEK inhibitor caused a large reduction in the survival-promoting effect that FGF-2 application to the nerve stump had on RGCs. Our results suggest that FGF-2 acts, at least in part, via the ERK pathway to prevent apoptosis of axotomized RGCs not only by increasing amounts of anti-apoptotic proteins, but also by a striking reduction in the levels of apoptotic effectors themselves.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Degeneration/metabolism , Nerve Regeneration/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinal Ganglion Cells/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Axotomy , Caspase 3 , Caspases/drug effects , Caspases/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Down-Regulation/physiology , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nerve Degeneration/drug therapy , Nerve Degeneration/prevention & control , Nerve Regeneration/drug effects , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/enzymology , Proto-Oncogene Proteins c-bcl-2/drug effects , Rana pipiens , Retinal Ganglion Cells/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology , bcl-2-Associated X Protein , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...