Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Gene Med ; 19(3)2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28087981

RESUMO

BACKGROUND: Malignant gliomas (glioblastomas; GBMs) are extremely aggressive and have a median survival of approximately 15 months. Current treatment modalities, which include surgical resection, radiation and chemotherapy, have done little to prolong the lives of GBM patients. Chondroitin sulfate proteoglycans (CSPG) are critical for cell-cell and cell-extracellular matrix (ECM) interactions and are implicated in glioma growth and invasion. Chondroitinase (Chase) ABC is a bacterial enzyme that cleaves chondroitin sulfate disaccharide chains from CSPGs in the tumor ECM. Wild-type Chase ABC has limited stability and/or activity in mammalian cells; therefore, we created a mutant humanized version (Chase M) with enhanced function in mammalian cells. METHODS: We hypothesized that disruption of cell-cell and cell-ECM interactions by ChaseM and temozolomide (TMZ) will enhance the chemotherapeutic availability and sensitivity of glioma cells. RESULTS: Utilizing primary patient-derived neurospheres, we found that ChaseM decreases glioma neurosphere aggregation in vitro. Furthermore, an oncolytic HSV-1 virus expressing secreted ChaseM (OV-ChaseM) enhanced viral spread and glioma cell killing compared to OV-Control, in vitro. OV-ChaseM plus TMZ combinatorial treatment resulted in a significant synergistic enhancement of glioma cell killing accompanied by an increase in apoptotic cell death. Intracellular flow cytometric analysis revealed a significant reduction in the phosphorylation of the pro-survival AKT protein following OV-ChaseM plus TMZ treatment. Lastly, in nude mice bearing intracranial GBM30 glioma xenografts, intratumoral OV-ChaseM plus TMZ (10 mg/kg by oral gavage) combination therapy resulted in a significant (p < 0.02) enhancement of survival compared to each individual treatment alone. CONCLUSIONS: These data reveal that OV-ChaseM enhances glioma cell viral susceptibility and sensitivity to TMZ.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Condroitina ABC Liase/genética , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/genética , Alelos , Substituição de Aminoácidos , Animais , Apoptose/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Chlorocebus aethiops , Condroitina ABC Liase/metabolismo , Dacarbazina/farmacologia , Modelos Animais de Doenças , Ativação Enzimática , Expressão Gênica , Vetores Genéticos/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Herpesvirus Humano 1/genética , Humanos , Camundongos , Mutação , Terapia Viral Oncolítica , Temozolomida , Transdução Genética , Resultado do Tratamento , Carga Tumoral , Células Tumorais Cultivadas , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Mol Ther ; 18(2): 285-94, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19844198

RESUMO

Oncolytic viral (OV) therapy is a promising therapeutic modality for brain tumors. Vasculostatin (Vstat120) is the cleaved and secreted extracellular fragment of brain-specific angiogenesis inhibitor 1 (BAI1), a brain-specific receptor. To date, the therapeutic efficacy of Vstat120 delivery into established tumors has not been investigated. Here we tested the therapeutic efficacy of combining Vstat120 gene delivery in conjunction with OV therapy. We constructed RAMBO (Rapid Antiangiogenesis Mediated By Oncolytic virus), which expresses Vstat120 under the control of the herpes simplex virus (HSV) IE4/5 promoter. Secreted Vstat120 was detected as soon as 4 hours postinfection in vitro and was retained for up to 13 days after OV therapy in subcutaneous tumors. RAMBO-produced Vstat120 efficiently inhibited endothelial cell migration and tube formation in vitro (P = 0.0005 and P = 0.0184, respectively) and inhibited angiogenesis (P = 0.007) in vivo. There was a significant suppression of intracranial and subcutaneous glioma growth in mice treated with RAMBO compared to the control virus, HSVQ (P = 0.0021 and P < 0.05, respectively). Statistically significant reduction in tumor vascular volume fraction (VVF) and microvessel density (MVD) was observed in tumors treated with RAMBO. This is the first study to report the antitumor effects of Vstat120 delivery into established tumors and supports the further development of RAMBO as a possible cancer therapy.


Assuntos
Proteínas Angiogênicas/fisiologia , Glioma/terapia , Vírus Oncolíticos/fisiologia , Proteínas Angiogênicas/genética , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos , Camundongos Nus , Vírus Oncolíticos/genética , Simplexvirus/genética , Células Vero
3.
Mol Ther ; 17(9): 1517-26, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19352323

RESUMO

The microtubule-associated protein tau (MAPT) and alpha-synuclein (SNCA) genes play central roles in neurodegenerative disorders. Mutations in each gene cause familial disease, whereas common genetic variation at both loci contributes to susceptibility to sporadic neurodegenerative disease. Here, we demonstrate exquisite gene regulation of the human MAPT and SNCA transgene loci and functional complementation in neuronal cell cultures and organotypic brain slices using the herpes simplex virus type 1 (HSV-1) amplicon-based infectious bacterial artificial chromosome (iBAC) vector to express complete loci >100 kb. Cell cultures transduced by iBAC vectors carrying a 143 kb MAPT or 135 kb SNCA locus expressed the human loci similar to the endogenous gene. We focused on analysis of the iBAC-MAPT vector carrying the complete MAPT locus. On transduction into neuronal cultures, multiple MAPT transcripts were expressed from iBAC-MAPT under strict developmental and cell type-specific control. In primary neurons from Mapt(-/-) mice, the iBAC-MAPT vector expressed the human tau protein, as detected by enzyme-linked immunosorbent assay and immunocytochemistry, and restored sensitivity of Mapt(-/-) neurons to Abeta peptide treatment in dissociated neuronal cultures and in organotypic slice cultures. The faithful retention of gene expression and phenotype complementation by the system provides a novel method to analyze neurological disease genes.


Assuntos
Doenças do Sistema Nervoso/terapia , Neurônios/patologia , Transgenes/genética , Transgenes/fisiologia , Animais , Células Cultivadas , Cromossomos Artificiais Bacterianos , Terapia Genética , Vetores Genéticos/genética , Humanos , Imuno-Histoquímica , Camundongos , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Neurônios/metabolismo , Ratos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , alfa-Sinucleína/fisiologia , Proteínas tau/genética , Proteínas tau/metabolismo , Proteínas tau/fisiologia
4.
Cancer Res ; 67(19): 9398-406, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17909049

RESUMO

Clinical trials have proven oncolytic virotherapy to be safe but not effective. We have shown that oncolytic viruses (OV) injected into intracranial gliomas established in rodents are rapidly cleared, and this is associated with up-regulation of markers (CD68 and CD163) of cells of monocytic lineage (monocytes/microglia/macrophages). However, it is unclear whether these cells directly impede intratumoral persistence of OV through phagocytosis and whether they infiltrate the tumor from the blood or the brain parenchyma. To investigate this, we depleted phagocytes with clodronate liposomes (CL) in vivo through systemic delivery and ex vivo in brain slice models with gliomas. Interestingly, systemic CL depleted over 80% of peripheral CD163+ macrophages in animal spleen and peripheral blood, thereby decreasing intratumoral infiltration of these cells, but CD68+ cells were unchanged. Intratumoral viral titers increased 5-fold. In contrast, ex vivo CL depleted only CD68+ cells from brain slices, and intratumoral viral titers increased 10-fold. These data indicate that phagocytosis by both peripheral CD163+ and brain-resident CD68+ cells infiltrating tumor directly affects viral clearance from tumor. Thus, improved therapeutic efficacy may require modulation of these innate immune cells. In support of this new therapeutic paradigm, we observed intratumoral up-regulation of CD68+ and CD163+ cells following treatment with OV in a patient with glioblastoma.


Assuntos
Neoplasias Encefálicas/virologia , Glioma/virologia , Macrófagos/virologia , Terapia Viral Oncolítica/métodos , Animais , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Glioma/sangue , Glioma/patologia , Glioma/terapia , Masculino , Camundongos , Camundongos Nus , Microglia/patologia , Fagocitose , Ratos , Ratos Endogâmicos F344
5.
Neuro Oncol ; 10(5): 690-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18715951

RESUMO

Therapies targeting glioma cells that diffusely infiltrate normal brain are highly sought after. Our aim was to identify novel approaches to this problem using glioma spheroid migration assays. Lithium, a currently approved drug for the treatment of bipolar illnesses, has not been previously examined in the context of glioma migration. We found that lithium treatment potently blocked glioma cell migration in spheroid, wound-healing, and brain slice assays. The effects observed were dose dependent and reversible, and worked using every glioma cell line tested. In addition, there was little effect on cell viability at lithium concentrations that inhibit migration, showing that this is a specific effect. Lithium treatment was associated with a marked change in cell morphology, with cells retracting the long extensions at their leading edge. Examination of known targets of lithium showed that inositol monophosphatase inhibition had no effect on glioma migration, whereas inhibition of glycogen synthase kinase-3 (GSK-3) did. This suggested that the effects of lithium on glioma cell migration could possibly be mediated through GSK-3. Specific pharmacologic GSK-3 inhibitors and siRNA knockdown of GSK-3alpha or GSK-3beta isoforms both reduced cell motility. These data outline previously unidentified pathways and inhibitors that may be useful for the development of novel anti-invasive therapeutics for the treatment of brain tumors.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/enzimologia , Movimento Celular/efeitos dos fármacos , Glioma/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Cloreto de Lítio/farmacologia , Animais , Western Blotting , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Glioma/patologia , Quinase 3 da Glicogênio Sintase/efeitos dos fármacos , Humanos , Camundongos , Invasividade Neoplásica/fisiopatologia , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos
6.
J Histochem Cytochem ; 55(5): 461-76, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17189521

RESUMO

Cholinergic agents affect the light responses of many ganglion cells (GCs) in the mammalian retina by activating nicotinic acetylcholine receptors (nAChRs). Whereas retinal neurons that express beta2 subunit-containing nAChRs have been characterized in the rabbit retina, expression patterns of other nAChR subtypes remain unclear. Therefore, we evaluated the expression of alpha7 nAChRs in retinal neurons by means of single-, double-, and triple-label immunohistochemistry. Our data demonstrate that, in the rabbit retina, several types of bipolar cells, amacrine cells, and cells in the GC layer express alpha7 nAChRs. At least three different populations of cone bipolar cells exhibited alpha7 labeling, whereas glycine-immunoreactive amacrine cells comprised the majority of alpha7-positive amacrine cells. Some GABAergic amacrine cells also displayed alpha7 immunoreactivity; alpha7 labeling was never detected in rod bipolar cells or rod amacrine cells (AII amacrine cells). Our data suggest that activation of alpha7 nAChRs by acetylcholine (ACh) or choline may affect glutamate release from several types of cone bipolar cells, modulating GC responses. ACh-induced excitation of inhibitory amacrine cells might cause either inhibition or disinhibition of other amacrine and GC circuits. Finally, ACh may act on alpha7 nAChRs expressed by GCs themselves.


Assuntos
Células Amácrinas/metabolismo , Receptores Nicotínicos/biossíntese , Células Bipolares da Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Bungarotoxinas/metabolismo , Imuno-Histoquímica , Coelhos , Frações Subcelulares/metabolismo , Receptor Nicotínico de Acetilcolina alfa7
7.
J Comp Neurol ; 456(2): 167-75, 2003 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-12509873

RESUMO

The light-evoked release of acetylcholine (ACh) affects the responses of many retinal ganglion cells, in part via nicotinic acetylcholine receptors (nAChRs). nAChRs that contain beta2alpha3 neuronal nicotinic acetylcholine receptors have been identified and localized in the rabbit retina; these nAChRs are recognized by the monoclonal antibody mAb210. We have examined the expression of beta2alpha3 nAChRs by glycinergic amacrine cells in the rabbit retina and have identified different subpopulations of nicotinic cholinoceptive glycinergic cells using double and triple immunohistochemistry with quantitative analysis. Here we demonstrate that about 70% of the cholinoceptive amacrine cells in rabbit retina are glycinergic cells. At least three nonoverlapping subpopulations of mAb210 glycine-immunoreactive cells can be distinguished with antibodies against calretinin, calbindin, and gamma-aminobutyric acid (GABA)(A) receptors. The cholinergic cells in rabbit retina are thought to synapse only on other cholinergic cells and ganglion cells. Thus, the expression of beta2alpha3 nAChRs on diverse populations of glycinergic cells is puzzling. To explore this finding, the subcellular localization of beta2alpha3 was studied at the electron microscopic level. mAb210 immunoreactivity was localized on the dendrites of amacrines and ganglion cells throughout the inner plexiform layer, and much of the labeling was not associated with recognizable synapses. Thus, our findings indicate that ACh in the mammalian retina may modulate glycinergic circuits via extrasynaptic beta2alpha3 nAChRs.


Assuntos
Neurônios/citologia , Receptores Nicotínicos/biossíntese , Retina/citologia , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Animais , Biomarcadores/análise , Colina O-Acetiltransferase/biossíntese , Glicina/biossíntese , Proteínas da Membrana Plasmática de Transporte de Glicina , Imuno-Histoquímica , Microscopia Imunoeletrônica , Neurônios/metabolismo , Neurônios/ultraestrutura , Coelhos , Retina/metabolismo , Retina/ultraestrutura , Ácido gama-Aminobutírico/biossíntese
8.
J Comp Neurol ; 461(1): 76-90, 2003 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-12722106

RESUMO

Starburst amacrine cells in the macaque retina were studied by electron microscopic immunohistochemistry. We found that these amacrine cells make a type of synapse not described previously; they are presynaptic to axon terminals of bipolar cells. We also confirmed that starburst amacrine cells are presynaptic to ganglion cell dendrites and amacrine cell processes. In order to determine the functions of these synapses, we localized acetylcholine receptors using a monoclonal antibody (mAb210) that recognizes human alpha3- and alpha5-containing nicotinic receptors and also antisera against the five known subtypes of muscarinic receptors. The majority of the mAb210-immunoreactive perikarya were amacrine cells and ganglion cells, but a subpopulation of bipolar cells was also labeled. A subset of bipolar cells and a subset of horizontal cells were labeled with antibodies to M3 muscarinic receptors. A subset of amacrine cells, including those that contain cholecystokinin, were labeled with antibodies to M2 receptors. Taken together, these results suggest that acetylcholine can modulate the activity of retinal ganglion cells by multiple pathways.


Assuntos
Células Amácrinas/ultraestrutura , Macaca mulatta/fisiologia , Receptores Colinérgicos/metabolismo , Retina/ultraestrutura , Sinapses/ultraestrutura , Acetilcolina/metabolismo , Células Amácrinas/metabolismo , Animais , Olho/inervação , Imuno-Histoquímica , Interneurônios/fisiologia , Microscopia Eletrônica , Retina/metabolismo , Células Ganglionares da Retina/fisiologia , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Distribuição Tecidual , Ácido gama-Aminobutírico/metabolismo
9.
PLoS One ; 9(7): e102499, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25047810

RESUMO

Oncolytic viruses are genetically engineered viruses that are designed to kill cancer cells while doing minimal damage to normal healthy tissue. After being injected into a tumor, they infect cancer cells, multiply inside them, and when a cancer cell is killed they move on to spread and infect other cancer cells. Chondroitinase ABC (Chase-ABC) is a bacterial enzyme that can remove a major glioma ECM component, chondroitin sulfate glycosoamino glycans from proteoglycans without any deleterious effects in vivo. It has been shown that Chase-ABC treatment is able to promote the spread of the viruses, increasing the efficacy of the viral treatment. In this paper we develop a mathematical model to investigate the effect of the Chase-ABC on the treatment of glioma by oncolytic viruses (OV). We show that the model's predictions agree with experimental results for a spherical glioma. We then use the model to test various treatment options in the heterogeneous microenvironment of the brain. The model predicts that separate injections of OV, one into the center of the tumor and another outside the tumor will result in better outcome than if the total injection is outside the tumor. In particular, the injection of the ECM-degrading enzyme (Chase-ABC) on the periphery of the main tumor core need to be administered in an optimal strategy in order to infect and eradicate the infiltrating glioma cells outside the tumor core in addition to proliferative cells in the bulk of tumor core. The model also predicts that the size of tumor satellites and distance between the primary tumor and multifocal/satellite lesions may be an important factor for the efficacy of the viral therapy with Chase treatment.


Assuntos
Bactérias/enzimologia , Neoplasias Encefálicas/terapia , Condroitina ABC Liase/uso terapêutico , Glioma/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioma/patologia , Humanos , Modelos Biológicos
10.
Oncotarget ; 5(20): 9703-9, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25210852

RESUMO

SapC-DOPS is a novel nanotherapeutic that has been shown to target and induce cell death in a variety of cancers, including glioblastoma (GBM). GBM is a primary brain tumor known to frequently demonstrate resistance to apoptosis-inducing therapeutics. Here we explore the mode of action for SapC-DOPS in GBM, a treatment being developed by Bexion Pharmaceuticals for clinical testing in patients. SapC-DOPS treatment was observed to induce lysosomal dysfunction of GBM cells characterized by decreased glycosylation of LAMP1 and altered proteolytic processing of cathepsin D independent of apoptosis and autophagic cell death. We observed that SapC-DOPS induced lysosomal membrane permeability (LMP) as shown by LysoTracker Red and Acridine Orange staining along with an increase of sphingosine, a known inducer of LMP. Additionally, SapC-DOPS displayed strong synergistic interactions with the apoptosis-inducing agent TMZ. Collectively our data suggest that SapC-DOPS induces lysosomal cell death in GBM cells, providing a new approach for treating tumors resistant to traditional apoptosis-inducing agents.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Nanoestruturas/administração & dosagem , Fosfatidilserinas/farmacologia , Saposinas/farmacologia , Animais , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dacarbazina/administração & dosagem , Dacarbazina/farmacologia , Sinergismo Farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Lisossomos/efeitos dos fármacos , Camundongos , Camundongos Nus , Distribuição Aleatória , Saposinas/administração & dosagem , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Clin Cancer Res ; 17(6): 1362-72, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21177410

RESUMO

PURPOSE: The inhibitory role of secreted chondroitin sulfate proteoglycans on oncolytic viral (OV) therapy was examined. Chondroitinase ABC (Chase-ABC) is a bacterial enzyme that can remove chondroitin sulfate glycosaminoglycans from proteoglycans without any deleterious effects in vivo. We examined the effect of Chase-ABC on OV spread and efficacy. EXPERIMENTAL DESIGN: Three-dimensional glioma spheroids placed on cultured brain slices were utilized to evaluate OV spread. Replication-conditional OV-expressing Chase-ABC (OV-Chase) was engineered using HSQuik technology and tested for spread and efficacy in glioma spheroids. Subcutaneous and intracranial glioma xenografts were utilized to compare antitumor efficacy of OV-Chase, rHsvQ (control), and PBS. Titration of viral particles was performed from OV-treated subcutaneous tumors. Glioma invasion was assessed in collagen-embedded glioma spheroids in vitro and in intracranial tumors. All statistical tests were two sided. RESULTS: Treatment with Chase-ABC in cultured glioma cells significantly enhanced OV spread in glioma spheroids grown on brain slices (P < 0.0001). Inoculation of subcutaneous glioma xenografts with Chase-expressing OV significantly increased viral titer (>10 times, P = 0.0008), inhibited tumor growth, and significantly increased overall animal survival (P < 0.006) compared with treatment with parental rHsvQ virus. Single OV-Chase administration in intracranial xenografts also resulted in longer median survival of animals than rHsvQ treatment (32 vs. 21 days, P < 0.018). Glioma cell migration and invasion were not increased by OV-Chase treatment. CONCLUSIONS: We conclude that degradation of glioma extracellular matrix with OV-expressing bacterial Chase-ABC enhanced OV spread and antitumor efficacy.


Assuntos
Condroitina ABC Liase/metabolismo , Sulfatos de Condroitina/química , Animais , Antineoplásicos/farmacologia , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Deleção de Genes , Glioma , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Luminescentes/metabolismo , Camundongos , Transplante de Neoplasias , Vírus Oncolíticos , Proteoglicanas/química , Proteína Vermelha Fluorescente
12.
Vis Neurosci ; 24(4): 635-45, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17900379

RESUMO

A GABA- or glycine-induced increase in Cl(-) permeability can produce either a depolarization or hyperpolarization, depending on the Cl(-) equilibrium potential. It has been shown that retinal neurons express the chloride cotransporters, Na-K-2Cl (NKCC) and K-Cl (KCC), the primary molecular mechanisms that control the intracellular Cl(-) concentration. We thus studied (1) the localization of these cotransporters in the fish retina, and (2) how suppression of cotransporter activity in the fish retina affects function. Specific antibodies against NKCC and KCC2 revealed that both cotransporters were expressed in the outer and inner plexiform layers, and colocalized in many putative amacrine cells and in cells of the ganglion cell layer. However, the somata of putative horizontal cells displayed only NKCC immunoreactivity and many bipolar cells were only immunopositive for KCC2. In the outer retina, application of bumetanide, a specific inhibitor of NKCC activity, (1) increased the steady-state extracellular concentration of K+ ([K+](o)) and enhanced the light-induced decrease in the [K+](o), (2) increased the sPIII photoreceptor-dependent component of the ERG, and (3) reduced the extracellular space volume. In contrast, in the outer retina, application of furosemide, a specific inhibitor of KCC activity, decreased sPIII and the light-induced reduction in [K+](o), but had little effect on steady-state [K+](o). In the inner retina, bumetanide increased the sustained component of the light-induced increase in [K+](o). These findings thus indicate that NKCC and KCC2 control the [K+](o) and extracellular space volume in the retina in addition to regulating GABA- and glycine-mediated synaptic transmission. In addition, the anatomical and electrophysiological results together suggest that all of the major neuronal types in the fish retina are influenced by chloride cotransporter activity.


Assuntos
Carpa Dourada/fisiologia , Retina/fisiologia , Simportadores de Cloreto de Sódio-Potássio/fisiologia , Algoritmos , Animais , Interpretação Estatística de Dados , Eletrofisiologia , Eletrorretinografia , Espaço Extracelular/fisiologia , Imuno-Histoquímica , Estimulação Luminosa , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/fisiologia , Compostos de Amônio Quaternário/farmacologia , Simportadores de Cloreto de Sódio-Potássio/genética
13.
Vis Neurosci ; 19(4): 495-509, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12511082

RESUMO

The effects of intraocular injections of ethylcholine mustard aziridinium ion (AF64A), an irreversible inhibitor of choline uptake, on the rabbit retina were assessed electrophysiologically, pharmacologically, anatomically, and behaviorally. Survival times from 1 day to 30 days were investigated. After 24 h, the shortest time tested, the directional selectivity of On-Off responding ganglion cells having the characteristic morphology of On-Off directionally selective directionally selective (DS) ganglion cells, as revealed by intracellular dye injection, was significantly reduced, both by an apparent decrease of preferred direction responses and an increase in responses to null-direction movement. No toxin-mediated changes in the dendritic trees of these cells were noted. Cells in AF64A-affected retinas having the DS morphology did not respond significantly to GABAergic or cholinergic agents such as picrotoxin and eserine, but did respond to nicotine. Recordings from a small random sample of other ganglion cell classes in the same retinas yielded no obvious changes in response properties. The direct effects on starburst (cholinergic) amacrine cells, which were identified by intraocular injection of the fluorescent dye DAPI with the AF64A, were investigated by intracellular injections of Lucifer yellow, and by immunohistochemical staining with antibodies to choline acetyltransferase (ChAT). Although starburst amacrine cell somas survived the AF64A treatment for at least several days, the dendrites could not be visualized by fluorescent dye injection in affected retinas due to dye leakage of the injected fluorescent dye from either the soma or proximal dendritic region. ChAT staining revealed a sequence in which ChAT-positive cells were undetectable first in the inner nuclear layer, and then in the ganglion cell layer. Cholinergic amacrine cells in the central retina were also affected before those in the periphery. The electrophysiological changes observed typically preceded the loss of ChAT activity. Behavioral tests for optokinetic nystagmus responses also revealed a lack of such responses in the affected eyes.


Assuntos
Células Amácrinas/efeitos dos fármacos , Células Amácrinas/fisiologia , Aziridinas/farmacologia , Colina/análogos & derivados , Colina/farmacologia , Fibras Colinérgicas/efeitos dos fármacos , Fibras Colinérgicas/fisiologia , Retina/fisiologia , Toxinas Biológicas/farmacologia , Animais , Colina O-Acetiltransferase/metabolismo , Dendritos/ultraestrutura , Imuno-Histoquímica , Coelhos , Retina/ultraestrutura , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA