Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Physiol ; 2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-38340081

RESUMO

We determined the role played by the transient receptor potential canonical 6 (TRPC6) channel in evoking the mechanical component of the exercise pressor reflex in male decerebrated Sprague-Dawley rats. TRPC6 channels were identified by quadruple-labelled (DiI, TRPC6, neurofilament-200 and peripherin) immunohistochemistry in dorsal root ganglion (DRG) cells innervating the triceps surae muscles (n = 12). The exercise pressor reflex was evoked by statically contracting the triceps surae muscles before and after injection of the TRPC6 antagonist BI-749327 (n = 11; 12 µg kg-1 ) or SAR7334 (n = 11; 7 µg kg-1 ) or the TRPC6 positive modulator C20 (n = 11; 18 µg kg-1 ). Similar experiments were conducted while the muscles were passively stretched (n = 8-12), a manoeuvre that isolated the mechanical component of the reflex. Blood pressure, tension, renal sympathetic nerve activity (RSNA) and blood flow were recorded. Of the DRG cells innervating the triceps surae muscles, 85% stained positive for the TRPC6 antigen, and 45% of those cells co-expressed neurofilament-200. Both TRPC6 antagonists decreased the reflex pressor responses to static contraction (-32 to -42%; P < 0.05) and to passive stretch (-35 to -52%; P < 0.05), whereas C20 increased these responses (55-65%; P < 0.05). In addition, BI-749327 decreased the peak and integrated RSNA responses to both static contraction (-39 to -43%; P < 0.05) and passive stretch (-56 to -62%; P < 0.05), whereas C20 increased the RSNA to passive stretch only. The onset latency of the decrease or increase in RSNA occurred within 2 s of the onset of the manoeuvres (P < 0.05). Collectively, our results show that TRPC6 plays a key role in evoking the mechanical component of the exercise pressor reflex. KEY POINTS: The exercise pressor reflex plays a key role in the sympathetic and haemodynamic responses to exercise. This reflex is composed of two components, namely the mechanoreflex and the metaboreflex. The receptors responsible for evoking the mechanoreflex are poorly documented. A good candidate for this function is the transient receptor potential canonical 6 (TRPC6) channel, which is activated by mechanical stimuli and expressed in dorsal root ganglia of rats. Using two TRPC6 antagonists and one positive modulator, we investigated the role played by TRPC6 in evoking the mechanoreflex in decerebrated rats. Blocking TRPC6 decreased the renal sympathetic and the pressor responses to both contraction and stretch, the latter being a manoeuvre that isolates the mechanoreflex. In contrast, the positive modulator increased the pressor reflex to contraction and stretch, in addition to the sympathetic response to stretch. Our results provide strong support for a role played by the TRPC6 channel in evoking the mechanoreflex.

2.
Mol Pharmacol ; 105(3): 250-259, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38182431

RESUMO

Opioid analgesics are frequently associated with gastrointestinal side effects, including constipation, nausea, dysphagia, and reduced gastric motility. Though it has been shown that stimulation of opioid receptors expressed in enteric motor neurons contributes to opioid-induced constipation, it remains unclear whether activation of opioid receptors in gastric-projecting nodose ganglia neurons contributes to the reduction in gastric motility and emptying associated with opioid use. In the present study, whole-cell patch-clamp recordings were performed to determine the mechanism underlying opioid receptor-mediated modulation of Ca2+ currents in acutely isolated gastric vagal afferent neurons. Our results demonstrate that CaV2.2 channels provide the majority (71% ± 16%) of Ca2+ currents in gastric vagal afferent neurons. Furthermore, we found that application of oxycodone, U-50488, or deltorphin II on gastric nodose ganglia neurons inhibited Ca2+ currents through a voltage-dependent mechanism by coupling to the Gα i/o family of heterotrimeric G-proteins. Because previous studies have demonstrated that the nodose ganglia expresses low levels of δ-opioid receptors, we also determined the deltorphin II concentration-response relationship and assessed deltorphin-mediated Ca2+ current inhibition following exposure to the δ-opioid receptor antagonist ICI 174,864 (0.3 µM). The peak mean Ca2+ current inhibition following deltorphin II application was 47% ± 24% (EC50 = 302.6 nM), and exposure to ICI 174,864 blocked deltorphin II-mediated Ca2+ current inhibition (4% ± 4% versus 37% ± 20%). Together, our results suggest that analgesics targeting any opioid receptor subtype can modulate gastric vagal circuits. SIGNIFICANCE STATEMENT: This study demonstrated that in gastric nodose ganglia neurons, agonists targeting all three classical opioid receptor subtypes (µ, δ, and κ) inhibit voltage-gated Ca2+ channels in a voltage-dependent mechanism by coupling to Gαi/o. These findings suggest that analgesics targeting any opioid receptor subtype would modulate gastric vagal circuits responsible for regulating gastric reflexes.


Assuntos
Analgésicos Opioides , Receptores Opioides kappa , Humanos , Analgésicos Opioides/farmacologia , Receptores Opioides mu/fisiologia , Constipação Intestinal , Neurônios Aferentes , Receptores Opioides , Analgésicos/farmacologia
3.
Neurogastroenterol Motil ; 35(11): e14646, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37480186

RESUMO

BACKGROUND: Neurogenic bowel is a dysmotility disorder following spinal cord injury (SCI) that negatively impacts quality of life, social integration, and physical health. Colonic transit is directly modulated by the enteric nervous system. Interstitial Cells of Cajal (ICC) distributed throughout the small intestine and colon serve as specialized pacemaker cells, generating rhythmic electrical slow waves within intestinal smooth muscle, or serve as an interface between smooth muscle cells and enteric motor neurons of the myenteric plexus. Interstitial Cells of Cajal loss has been reported for other preclinical models of dysmotility, and our previous experimental SCI study provided evidence of reduced excitatory and inhibitory enteric neuronal count and smooth muscle neural control. METHODS: Immunohistochemistry for the ICC-specific marker c-Kit was utilized to examine neuromuscular remodeling of the distal colon in male and female rats with experimental SCI. KEY RESULTS: Myenteric plexus ICC (ICC-MP) exhibited increased cell counts 3 days following SCI in male rats, but did not significantly increase in females until 3 weeks after SCI. On average, ICC-MP total primary arborization length increased significantly in male rats at 3-day, 3-week, and 6-week time points, whereas in females, this increase occurred most frequently at 6 weeks post-SCI. Conversely, circular muscle ICC (ICC-CM) did not demonstrate post-SCI changes. CONCLUSIONS AND INFERENCES: These data demonstrate resiliency of the ICC-MP in neurogenic bowel following SCI, unlike seen in other related disease states. This plasticity underscores the need to further understand neuromuscular changes driving colonic dysmotility after SCI in order to advance therapeutic targets for neurogenic bowel treatment.


Assuntos
Sistema Nervoso Entérico , Intestino Neurogênico , Traumatismos da Medula Espinal , Ratos , Masculino , Feminino , Animais , Qualidade de Vida , Plexo Mientérico , Colo , Neurônios Motores , Traumatismos da Medula Espinal/complicações
4.
Cells ; 10(3)2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33809186

RESUMO

Neurodegenerative retinal diseases, such as glaucoma and diabetic retinopathy, involve a gradual loss of neurons in the retina as the disease progresses. Central nervous system neurons are not able to regenerate in mammals, therefore, an often sought after course of treatment for neuronal loss follows a neuroprotective or regenerative strategy. Neuroprotection is the process of preserving the structure and function of the neurons that have survived a harmful insult; while regenerative approaches aim to replace or rewire the neurons and synaptic connections that were lost, or induce regrowth of damaged axons or dendrites. In order to test the neuroprotective effectiveness or the regenerative capacity of a particular agent, a robust experimental model of retinal neuronal damage is essential. Zebrafish are being used more often in this type of study because their eye structure and development is well-conserved between zebrafish and mammals. Zebrafish are robust genetic tools and are relatively inexpensive to maintain. The large array of functional and behavioral tests available in zebrafish makes them an attractive model for neuroprotection studies. Some common insults used to model retinal disease and study neuroprotection in zebrafish include intense light, chemical toxicity and mechanical damage. This review covers the existing retinal neuroprotection and regeneration literature in the zebrafish and highlights their potential for future studies.


Assuntos
Degeneração Neural , Regeneração Nervosa/efeitos dos fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Neurogênese/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Doenças Retinianas/tratamento farmacológico , Neurônios Retinianos/efeitos dos fármacos , Peixe-Zebra , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Retinianas/genética , Doenças Retinianas/metabolismo , Doenças Retinianas/patologia , Neurônios Retinianos/metabolismo , Neurônios Retinianos/patologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
5.
Front Cell Neurosci ; 14: 600777, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33335476

RESUMO

Feedback inhibition by horizontal cells regulates rod and cone photoreceptor calcium channels that control their release of the neurotransmitter glutamate. This inhibition contributes to synaptic gain control and the formation of the center-surround antagonistic receptive fields passed on to all downstream neurons, which is important for contrast sensitivity and color opponency in vision. In contrast to the plasmalemmal GABA transporter found in non-mammalian horizontal cells, there is evidence that the mechanism by which mammalian horizontal cells inhibit photoreceptors involves the vesicular release of the inhibitory neurotransmitter GABA. Historically, inconsistent findings of GABA and its biosynthetic enzyme, L-glutamate decarboxylase (GAD) in horizontal cells, and the apparent lack of surround response block by GABAergic agents diminished support for GABA's role in feedback inhibition. However, the immunolocalization of the vesicular GABA transporter (VGAT) in the dendritic and axonal endings of horizontal cells that innervate photoreceptor terminals suggested GABA was released via vesicular exocytosis. To test the idea that GABA is released from vesicles, we localized GABA and GAD, multiple SNARE complex proteins, synaptic vesicle proteins, and Cav channels that mediate exocytosis to horizontal cell dendritic tips and axonal terminals. To address the perceived relative paucity of synaptic vesicles in horizontal cell endings, we used conical electron tomography on mouse and guinea pig retinas that revealed small, clear-core vesicles, along with a few clathrin-coated vesicles and endosomes in horizontal cell processes within photoreceptor terminals. Some small-diameter vesicles were adjacent to the plasma membrane and plasma membrane specializations. To assess vesicular release, a functional assay involving incubation of retinal slices in luminal VGAT-C antibodies demonstrated vesicles fused with the membrane in a depolarization- and calcium-dependent manner, and these labeled vesicles can fuse multiple times. Finally, targeted elimination of VGAT in horizontal cells resulted in a loss of tonic, autaptic GABA currents, and of inhibitory feedback modulation of the cone photoreceptor Cai, consistent with the elimination of GABA release from horizontal cell endings. These results in mammalian retina identify the central role of vesicular release of GABA from horizontal cells in the feedback inhibition of photoreceptors.

6.
Neuroreport ; 31(1): 22-28, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31789762

RESUMO

Light-induced retinal degeneration (LIRD) models are used to recapitulate the pathologies of retinal diseases that affect photoreceptors. Current LIRD models use a dark-adaptation period of 7-14 days followed by high-intensity light exposure. The purpose of this study was to determine whether photoreceptor damage and death would occur in pigmented zebrafish using a short period of dark-adaptation. Zebrafish were dark-adapted for 24 h and then exposed to constant high-intensity light for 48 h. Immunohistochemical analysis was performed on vertical retinal sections to assess damage and apoptosis. Photoreceptors exhibited structural damage, apoptosis, and cell loss after 24 and 48 h of light exposure as previously reported in studies using 7-14 day dark-adaption. Also, photoreceptors lost following light damage were regenerated after 28 days. These results suggest that a short period of dark-adaptation is sufficient for a LIRD model in pigmented zebrafish.


Assuntos
Modelos Animais de Doenças , Luz/efeitos adversos , Células Fotorreceptoras de Vertebrados/patologia , Degeneração Retiniana/patologia , Animais , Adaptação à Escuridão , Feminino , Masculino , Regeneração Nervosa/fisiologia , Degeneração Retiniana/etiologia , Peixe-Zebra
7.
Purinergic Signal ; 15(3): 327-342, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31273575

RESUMO

Adenosine is an endogenous nucleoside in the central nervous system that acts on adenosine receptors. These are G protein-coupled receptors that have four known subtypes: A1, A2A, A2B, and A3 receptors. In the present study, we aimed to map the location of the adenosine receptor subtypes in adult wild-type zebrafish retina using in situ hybridization and immunohistochemistry. A1R, A2AR, and A2BR mRNA were detected in the ganglion cell layer (GCL), the inner nuclear layer (INL), the outer nuclear layer (ONL), and the outer segment (OS). A3R mRNA was detected in the GCL, ONL, and OS. A1R-immunoreactivity was expressed as puncta in the INL and in the outer plexiform layer (OPL). A1Rs were located within the cone pedicle and contiguous to horizontal cell tips in the OPL. A2AR-immunoreactivity was expressed as puncta in the GCL, inner plexiform layer (IPL), INL, and outer retina. A2AR puncta in the outer retina were situated around the ellipsoids and nuclei of cones, and weakly around the rod nuclei. A1Rs and A2ARs were clustered around ON cone bipolar cell terminals and present in the OFF lamina of the INL but were not expressed on mixed rod/cone response bipolar cell terminals. A2BR-immunoreactivity was mainly localized to the Müller cells, while A3Rs were found to be expressed in retinal ganglion cells of the GCL, INL, ONL, and OS. In summary, all four adenosine receptor subtypes were localized in the zebrafish retina and are in agreement with expression patterns shown in retinas from other species.


Assuntos
Receptores Purinérgicos P1/metabolismo , Retina/metabolismo , Animais , Peixe-Zebra
8.
Neurosci Lett ; 706: 182-188, 2019 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-31085293

RESUMO

The nodose ganglion (NG) is the main parasympathetic ganglion conveying sensory signals to the CNS from numerous visceral organs including digestive signals such as gastric distension or the release the gastrointestinal peptides. The response characteristics of NG neurons to ATP and ADP and pharmacological interrogation of purinergic receptor subtypes have been previously investigated but often in NG cells of undetermined visceral origin. In this study, we confirmed the presence of P2X3 and P2Y1 receptors and characterized P2X and P2Y responses in gastric-innervating NG neurons. Application of ATP-evoked large inward currents and cytosolic Ca2+ increases in gastric-innervating NG neurons. Despite the expression of P2Y1 receptors, ADP elicited only minor modulation of voltage-gated Ca2+ channels. Considering the sensitivity of NG neurons to comorbidities associated with disease or neural injury, purinergic modulation of gastric NG neurons in disease- or injury-states is worthy of further investigation.


Assuntos
Gânglio Nodoso/metabolismo , Receptores Purinérgicos/metabolismo , Células Receptoras Sensoriais/metabolismo , Estômago/inervação , Nervo Vago/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Masculino , Gânglio Nodoso/efeitos dos fármacos , Ratos , Ratos Wistar , Células Receptoras Sensoriais/efeitos dos fármacos , Estômago/efeitos dos fármacos , Nervo Vago/efeitos dos fármacos
9.
Neuroreport ; 29(2): 118-122, 2018 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-29251688

RESUMO

Retinal ganglion cells (RGCs) that express the photopigment melanopsin (mRGCs) are photosensitive and initiate the non-image-forming pathway, where the majority of their axons terminate in the suprachiasmatic nucleus (SCN). RGCs only make up approximately half of the cells in the ganglion cell layer of the retina; therefore, it is important to be able to distinguish them from other cell types. The transgenic Thy-1 YFP mouse line 16 (Thy-1 YFP-16) expresses yellow-fluorescent protein (YFP) in projection neurons, including RGCs. Our objective was to determine whether mRGCs are labeled with YFP in Thy-1 YFP-16 transgenic mice. Paraformaldehyde-fixed retinal wholemounts and frozen vertical sections were prepared from Thy-1 YFP-16 mice and fluorescently labeled with rabbit anti-melanopsin and guinea-pig anti-RNA binding protein with multiple splicing to identify mRGCs and total RGCs, respectively. Thy-1 YFP-16 mouse brains were sectioned coronally and imaged to view RGC axonal projections to the SCN. Confocal images of retinal preparations show that the majority (∼89%) of mRGCs are not YFP-positive in Thy-1 YFP-16 mice, where ∼11% expressed a weak fluorescent signal. In addition, there are almost no YFP-positive axons present in the SCN of coronal brain sections. We conclude that the majority of mRGC somas and axons are not labeled with YFP in the transgenic Thy-1 YFP-16 mouse line; therefore, this mouse model may not suitable for research involving mRGC visual pathways.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Luminescentes/metabolismo , Camundongos Transgênicos/anatomia & histologia , Camundongos Transgênicos/metabolismo , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo , Opsinas de Bastonetes/metabolismo , Animais , Proteínas de Bactérias/genética , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Proteínas Luminescentes/genética , Masculino , Microscopia Confocal , Microscopia de Fluorescência , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Vias Visuais/citologia , Vias Visuais/metabolismo
10.
J Biol Chem ; 291(41): 21335-21349, 2016 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-27539848

RESUMO

Protein-tyrosine phosphatase receptor type G (RPTPγ/PTPRG) interacts in vitro with contactin-3-6 (CNTN3-6), a group of glycophosphatidylinositol-anchored cell adhesion molecules involved in the wiring of the nervous system. In addition to PTPRG, CNTNs associate with multiple transmembrane proteins and signal inside the cell via cis-binding partners to alleviate the absence of an intracellular region. Here, we use comprehensive biochemical and structural analyses to demonstrate that PTPRG·CNTN3-6 complexes share similar binding affinities and a conserved arrangement. Furthermore, as a first step to identifying PTPRG·CNTN complexes in vivo, we found that PTPRG and CNTN3 associate in the outer segments of mouse rod photoreceptor cells. In particular, PTPRG and CNTN3 form cis-complexes at the surface of photoreceptors yet interact in trans when expressed on the surfaces of apposing cells. Further structural analyses suggest that all CNTN ectodomains adopt a bent conformation and might lie parallel to the cell surface to accommodate these cis and trans binding modes. Taken together, these studies identify a PTPRG·CNTN complex in vivo and provide novel insights into PTPRG- and CNTN-mediated signaling.


Assuntos
Contactinas , Complexos Multiproteicos , Proteínas do Tecido Nervoso , Tecido Nervoso/metabolismo , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores , Transdução de Sinais/fisiologia , Animais , Contactinas/química , Contactinas/genética , Contactinas/metabolismo , Humanos , Camundongos , Modelos Biológicos , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/química , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/metabolismo
11.
PLoS One ; 7(9): e43463, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22984429

RESUMO

Photoreceptor terminals contain post-synaptic density (PSD) proteins e.g., PSD-95/PSD-93, but their role at photoreceptor synapses is not known. PSDs are generally restricted to post-synaptic boutons in central neurons and form scaffolding with multiple proteins that have structural and functional roles in neuronal signaling. The Shank family of proteins (Shank 1-3) functions as putative anchoring proteins for PSDs and is involved in the organization of cytoskeletal/signaling complexes in neurons. Specifically, Shank 1 is restricted to neurons and interacts with both receptors and signaling molecules at central neurons to regulate plasticity. However, it is not known whether Shank 1 is expressed at photoreceptor terminals. In this study we have investigated Shank 1A localization in the outer retina at photoreceptor terminals. We find that Shank 1A is expressed presynaptically in cone pedicles, but not rod spherules, and it is absent from mice in which the Shank 1 gene is deleted. Shank 1A co-localizes with PSD-95, peanut agglutinin, a marker of cone terminals, and glycogen phosphorylase, a cone specific marker. These findings provide convincing evidence for Shank 1A expression in both the inner and outer plexiform layers, and indicate a potential role for PSD-95/Shank 1 complexes at cone synapses in the outer retina.


Assuntos
Mamíferos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Animais , Proteína 4 Homóloga a Disks-Large , Deleção de Genes , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos , Aglutinina de Amendoim/metabolismo , Ligação Proteica , Células Fotorreceptoras Retinianas Cones/citologia , Sinapses/metabolismo , Aglutininas do Germe de Trigo/metabolismo
12.
J Biol Chem ; 286(37): 32563-74, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21705333

RESUMO

The choroid plexus lining the four ventricles in the brain is where the majority of cerebrospinal fluid (CSF) is produced. The secretory function of the choroid plexus is mediated by specific transport systems that allow the directional flux of nutrients and ions into the CSF and the removal of toxins. Normal CSF dynamics and chemistry ensure that the environment for neural function is optimal. Here, we report that targeted disruption of the Slc4a5 gene encoding the electrogenic sodium bicarbonate cotransporter NBCe2 results in significant remodeling of choroid plexus epithelial cells, including abnormal mitochondrial distribution, cytoskeletal protein expression, and ion transporter polarity. These changes are accompanied by very significant abnormalities in intracerebral ventricle volume, intracranial pressure, and CSF electrolyte levels. The Slc4a5(-/-) mice are significantly more resistant to induction of seizure behavior than wild-type controls. In the retina of Slc4a5(-/-) mice, loss of photoreceptors, ganglion cells, and retinal detachment results in visual impairment assessed by abnormal electroretinogram waveforms. Our findings are the first demonstration of the fundamental importance of NBCe2 in the biology of the nervous system.


Assuntos
Plexo Corióideo/metabolismo , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Equilíbrio Hidroeletrolítico , Animais , Plexo Corióideo/patologia , Pressão Intracraniana/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/patologia , Proteínas do Tecido Nervoso/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Descolamento Retiniano/líquido cefalorraquidiano , Descolamento Retiniano/genética , Simportadores de Sódio-Bicarbonato/genética
13.
J Comp Neurol ; 518(10): 1647-69, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20235161

RESUMO

Gamma-aminobutyric acid (GABA) is likely expressed in horizontal cells of all species, although conflicting physiological findings have led to considerable controversy regarding its role as a transmitter in the outer retina. This study has evaluated key components of the GABA system in the outer retina of guinea pig, an emerging retinal model system. The presence of GABA, its rate-limiting synthetic enzyme glutamic acid decarboxylase (GAD(65) and GAD(67) isoforms), the plasma membrane GABA transporters (GAT-1 and GAT-3), and the vesicular GABA transporter (VGAT) was evaluated by using immunohistochemistry with well-characterized antibodies. The presence of GAD(65) mRNA was also evaluated by using laser capture microdissection and reverse transcriptase-polymerase chain reaction. Specific GABA, GAD(65), and VGAT immunostaining was localized to horizontal cell bodies, as well as to their processes and tips in the outer plexiform layer. Furthermore, immunostaining of retinal whole mounts and acutely dissociated retinas showed GAD(65) and VGAT immunoreactivity in both A-type and B-type horizontal cells. However, these cells did not contain GAD(67), GAT-1, or GAT-3 immunoreactivity. GAD(65) mRNA was detected in horizontal cells, and sequencing of the amplified GAD(65) fragment showed approximately 85% identity with other mammalian GAD(65) mRNAs. These studies demonstrate the presence of GABA, GAD(65), and VGAT in horizontal cells of the guinea pig retina, and support the idea that GABA is synthesized from GAD(65), taken up into synaptic vesicles by VGAT, and likely released by a vesicular mechanism from horizontal cells.


Assuntos
Glutamato Descarboxilase/metabolismo , Isoenzimas/metabolismo , Células Horizontais da Retina/enzimologia , Células Horizontais da Retina/fisiologia , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Sequência de Bases , Calbindinas , Feminino , Glutamato Descarboxilase/genética , Cobaias , Humanos , Imuno-Histoquímica , Isoenzimas/genética , Masculino , Camundongos , Microdissecção/métodos , Dados de Sequência Molecular , Neuroglia/citologia , Neuroglia/metabolismo , Células Horizontais da Retina/citologia , Proteína G de Ligação ao Cálcio S100/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética
14.
Neuroreport ; 20(10): 923-9, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19491713

RESUMO

In the retina, adenosine is released in the dark and has been shown to inhibit Ca2+ influx through voltage-gated Ca2+ channels in cones. Therefore, we tested whether adenosine can inhibit exocytosis from isolated cone photoreceptors. Simultaneous measurements of membrane exocytosis and Ca2+ were made from cones using the activity-dependent dye, Synaptored-C2, and the Ca2+ indicator dye, Fluo-4. Adenosine suppressed exocytosis in cones, indicating that transmitter release is also reduced from cone terminals, and further supports an inhibitory mechanism for modulating transmitter release onto second-order neurons. Furthermore, this raises the possibility that adenosine might be neuroprotective for photoreceptors and second-order neurons by suppressing Ca2+ levels in cones and reducing exocytosis of L-glutamate, respectively.


Assuntos
Adenosina/metabolismo , Ambystoma/metabolismo , Exocitose/fisiologia , Terminações Pré-Sinápticas/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Transmissão Sináptica/fisiologia , Adenosina/farmacologia , Ambystoma/anatomia & histologia , Compostos de Anilina , Animais , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Citoproteção/efeitos dos fármacos , Citoproteção/fisiologia , Exocitose/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Indicadores e Reagentes , Cinética , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Neurotransmissores/metabolismo , Técnicas de Cultura de Órgãos , Potássio/metabolismo , Potássio/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Compostos de Piridínio , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Transmissão Sináptica/efeitos dos fármacos , Visão Ocular/efeitos dos fármacos , Visão Ocular/fisiologia , Xantenos
15.
PLoS One ; 4(6): e6009, 2009 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-19554099

RESUMO

BACKGROUND: The global disparity in cancer incidence remains a major public health problem. We focused on prostate cancer since microscopic disease in men is common, but the incidence of clinical disease varies more than 100 fold worldwide. Ca(2+) signaling is a central regulator of cell proliferation, but has received little attention in cancer prevention. We and others have reported a strong dose-dependent reduction in the incidence of prostate and lung cancer within populations exposed to boron (B) in drinking water and food; and in tumor and cell proliferation in animal and cell culture models. METHODS/PRINCIPAL FINDINGS: We examined the impact of B on Ca(2+) stores using cancer and non-cancer human prostate cell lines, Ca(2+) indicators Rhod-2 AM and Indo-1 AM and confocal microscopy. In DU-145 cells, inhibition of Ca(2+) release was apparent following treatment with Ringers containing RyR agonists cADPR, 4CmC or caffeine and respective levels of BA (50 microM), (1, 10 microM) or (10, 20, 50,150 microM). Less aggressive LNCaP cancer cells required 20 microM BA and the non-tumor cell line PWR1E required 150 microM BA to significantly inhibit caffeine stimulated Ca(2+) release. BA (10 microM) and the RyR antagonist dantroline (10 microM) were equivalent in their ability to inhibit ER Ca(2+) loss. Flow cytometry and confocal microscopy analysis showed exposure of DU-145 cells to 50 microM BA for 1 hr decreased stored [Ca(2+)] by 32%. CONCLUSION/SIGNIFICANCE: We show B causes a dose dependent decrease of Ca(2+) release from ryanodine receptor sensitive stores. This occurred at BA concentrations present in blood of geographically disparate populations. Our results suggest higher BA blood levels lower the risk of prostate cancer by reducing intracellular Ca(2+) signals and storage.


Assuntos
Ácidos Bóricos/farmacologia , Cálcio/metabolismo , Neoplasias da Próstata/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Indóis/farmacologia , Masculino
16.
Cell Biol Toxicol ; 25(4): 309-20, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18516691

RESUMO

Boron (B) is a developmental and reproductive toxin. It is also essential for some organisms. Plants use uptake and efflux transport proteins to maintain homeostasis, and in humans, boron has been reported to reduce prostate cancer. Ca2+ signaling is one of the primary mechanisms used by cells to respond to their environment. In this paper, we report that boric acid (BA) inhibits NAD+ and NADP+ as well as mechanically induced release of stored Ca2+ in growing DU-145 prostate cancer cells. Cell proliferation was inhibited by 30% at 100 microM, 60% at 250 microM, and 97% at 1,000 microM BA. NAD+-induced Ca2+ transients were partly inhibited at 250 microM BA and completely at 1,000 microM BA, whereas both NADP+ and mechanically induced transients were inhibited by 1,000 microM BA. Expression of CD38 protein increased in proportion to BA exposure (0-1,000 microM). In vitro mass spectrometry analysis showed that BA formed adducts with the CD38 products and Ca2+ channel agonists cyclic adenosine diphosphate ribose (cADPR) and nicotinic acid adenine dinucleotide phosphate (NAADP). Vesicles positive for the Ca2+ fluorophore fluo-3 acetoxymethyl ester accumulated in cells exposed to 250 and 1,000 microM BA. The BA analog, methylboronic acid (MBA; 250 and 1,000 microM), did not inhibit cell proliferation or NAD+, NADP+, or mechanically stimulated Ca2+ store release. Nor did MBA increase CD38 expression or cause the formation of intracellular vesicles. Thus, mammalian cells can distinguish between BA and its synthetic analog MBA and exhibit graded concentration-dependent responses. Based on these observations, we hypothesize that toxicity of BA stems from the ability of high concentrations to impair Ca2+ signaling.


Assuntos
Ácidos Bóricos/toxicidade , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , ADP-Ribosil Ciclase 1/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular Tumoral , ADP-Ribose Cíclica/metabolismo , Humanos , Masculino , NAD/metabolismo , NADP/metabolismo
17.
J Comp Neurol ; 512(1): 6-26, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18975268

RESUMO

Plasmalemmal and vesicular gamma-aminobutyric acid (GABA) transporters influence neurotransmission by regulating high-affinity GABA uptake and GABA release into the synaptic cleft and extracellular space. Postnatal expression of the plasmalemmal GABA transporter-1 (GAT-1), GAT-3, and the vesicular GABA/glycine transporter (VGAT) were evaluated in the developing mouse retina by using immunohistochemistry with affinity-purified antibodies. Weak transporter immunoreactivity was observed in the inner retina at postnatal day 0 (P0). GAT-1 immunostaining at P0 and at older ages was in amacrine and displaced amacrine cells in the inner nuclear layer (INL) and ganglion cell layer (GCL), respectively, and in their processes in the inner plexiform layer (IPL). At P10, weak GAT-1 immunostaining was in Müller cell processes. GAT-3 immunostaining at P0 and older ages was in amacrine cells and their processes, as well as in Müller cells and their processes that extended radially across the retina. At P10, Müller cell somata were observed in the middle of the INL. VGAT immunostaining was present at P0 and older ages in amacrine cells in the INL as well as processes in the IPL. At P5, weak VGAT immunostaining was also observed in horizontal cell somata and processes. By P15, the GAT and VGAT immunostaining patterns appear similar to the adult immunostaining patterns; they reached adult levels by about P20. These findings demonstrate that GABA uptake and release are initially established in the inner retina during the first postnatal week and that these systems subsequently mature in the outer retina during the second postnatal week.


Assuntos
Membrana Celular/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Retina/crescimento & desenvolvimento , Retina/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Encéfalo/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Retina/citologia
18.
J Neurosci Res ; 87(5): 1107-14, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19006083

RESUMO

Galanin activates three receptors, the galanin receptor 1 (GalR1), GalR2, and GalR3. In the gastrointestinal tract, GalR1 mediates the galanin inhibition of cholinergic transmission to the longitudinal muscle and reduction of peristalsis efficiency in the small intestine. Galanin has also been shown to inhibit depolarization-evoked Ca2+ increases in cultured myenteric neurons. Because GalR1 immunoreactivity is localized to cholinergic myenteric neurons, we hypothesized that this inhibitory action of galanin on myenteric neurons is mediated by GalR1. We investigated the effect of galanin 1-16, which has high affinity for GalR1 and GalR2, in the presence or absence of the selective GalR1 antagonist, RWJ-57408, and of galanin 2-11, which has high affinity for GalR2 and GalR3, on Ca2+ influx through voltage-dependent Ca2+ channels in cultured myenteric neurons. Myenteric neurons were loaded with fluo-4 and depolarized by high K+ concentration to activate voltage-dependent Ca2+ channels. Intracellular Ca2+ levels were quantified with confocal microscopy. Galanin 1-16 (0.01-1 microM) inhibited the depolarization-evoked Ca2+ increase in a dose-dependent manner with an EC(50) of 0.172 microM. The selective GalR1 antagonist, RWJ-57408 (10 microM), blocked the galanin 1-16 (1 microM)-mediated inhibition of voltage-dependent Ca2+ channel. By contrast, the GalR2/GalR3 agonist, galanin 2-11 did not affect the K+-evoked Ca2+ influx in myenteric neurons. GalR1 immunoreactivity was localized solely to myenteric neurons in culture, as previously observed in intact tissue. These findings indicate that the inhibition of depolarization-evoked Ca2+ influx in myenteric neurons in culture is mediated by GalR1 and confirm the presence of functional GalR1 in the myenteric plexus. This is consonant with the hypothesis that GalR1 mediates galanin inhibition of transmitter release from myenteric neurons.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Galanina/metabolismo , Plexo Mientérico/metabolismo , Neurônios/metabolismo , Receptor Tipo 1 de Galanina/metabolismo , Compostos de Anilina , Animais , Células Cultivadas , Feminino , Masculino , Potenciais da Membrana , Microscopia Confocal , Plexo Mientérico/citologia , Fragmentos de Peptídeos/metabolismo , Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Galanina/antagonistas & inibidores , Receptor Tipo 2 de Galanina/agonistas , Receptor Tipo 3 de Galanina/agonistas , Xantenos
19.
Brain Res ; 1215: 20-9, 2008 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-18482716

RESUMO

Glutamate is the major excitatory neurotransmitter in the retina, and most glutamatergic neurons express one of the three known vesicular glutamate transporters (VGLUT1, 2, or 3). However, the expression profiles of these transporters vary greatly in the retina. VGLUT1 is expressed by photoreceptor and bipolar cell terminals, and VGLUT2 appears to be predominately expressed by ganglion cells, and perhaps Müller cells, cone photoreceptor terminals, and horizontal cells in some species. The discovery of a third vesicular glutamate transporter, VGLUT3, has brought about speculation concerning its role and function based on its expression in amacrine cells. To address this we studied the postnatal development of VGLUT3 from day 0 through adult in the rat retina, and compared this with the expression patterns of VGLUT1 and VGLUT2. VGLUT3 expression was restricted to a population of amacrine cells. Expression of VGLUT3 was first observed at postnatal day 10 (P10) in the soma and some processes, which extensively arborized in both the ON and OFF sublamina of the IPL by P15. In contrast, VGLUT1 and VGLUT2 expression appeared earlier than VGLUT3; with VGLUT1 initially detected at P5 in photoreceptor terminals and P6 in bipolar terminals, and VGLUT2 immunoreactivity initially detected at P0 in ganglion cell bodies, and remained prominent throughout all stages of development. Interestingly, VGLUT3 has extensive somatic expression throughout development, which could be involved in non-synaptic modulation by glutamate in developing retina, and could influence trophic and extra-synaptic neuronal signaling by glutamate in the inner retina.


Assuntos
Células Amácrinas/metabolismo , Retina/metabolismo , Células Bipolares da Retina/metabolismo , Membranas Sinápticas/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Células Fotorreceptoras/metabolismo , Ratos , Ratos Sprague-Dawley , Retina/citologia , Retina/crescimento & desenvolvimento , Distribuição Tecidual , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/metabolismo
20.
J Neurosci Res ; 85(5): 1126-37, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17304584

RESUMO

Endogenous adenosine has already been shown to inhibit transmitter release from the rod synapse by suppressing Ca(2+) influx through voltage-gated Ca(2+) channels. However, it is not clear how adenosine modulates the cone synapse. Cone photoreceptors, like rod photoreceptors, also possess L-type Ca(2+) channels that regulate the release of L-glutamate. To assess the impact of adenosine on Ca(2+) influx though voltage-gated Ca(2+) channels in cone terminals, whole-cell perforated-patch clamp recording and Ca(2+) imaging with fluo-4 were used on isolated cones and salamander retinal slices. Synaptic markers (VAMP and piccolo) and activity-dependent dye labeling revealed that tiger salamander cone terminals contain a broad, vesicle-filled cytoplasmic extension at the base of the somatic compartment, which is unlike rod terminals that contain one or more thin axons, each terminating in a large bulbous synaptic terminal. The spatiotemporal Ca(2+) responses of the cone terminals do not differ significantly from the Ca(2+) responses of the soma or inner segment like that observed in rods. Whole-cell recording of cone I(Ca) and Ca(2+) imaging of synaptic terminals in cones demonstrate that adenosine inhibited both I(Ca) and the depolarization-evoked Ca(2+) increase in cone terminals in a dose-dependent manner from 1 to 50 muM. These results indicate that, as in rods, adenosine's ability to suppress voltage-dependent Ca(2+) channels at the cone synapse will limit the amount of L-glutamate released. Therefore, adenosine has an inhibitory effect on L-glutamate release at the first synapse, which likely favors elevated adenosine levels in the dark or during dark-adapted conditions.


Assuntos
Adenosina/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Terminações Pré-Sinápticas/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Transmissão Sináptica/fisiologia , Adenosina/farmacologia , Ambystoma , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Canais de Cálcio/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Adaptação à Escuridão/fisiologia , Relação Dose-Resposta a Droga , Corantes Fluorescentes , Ácido Glutâmico/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neuropeptídeos/metabolismo , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/ultraestrutura , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura , Proteína 1 Associada à Membrana da Vesícula/metabolismo , Visão Ocular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...