Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Traffic ; 16(12): 1239-53, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26373354

ABSTRACT

Na(+) /K(+) -ATPase (NKA) participates in setting electrochemical gradients, cardiotonic steroid signaling and cellular adhesion. Distinct isoforms of NKA are found in different tissues and subcellular localization patterns. For example, NKA α1 is widely expressed, NKA α3 is enriched in neurons and NKA α4 is a testes-specific isoform found in sperm flagella. In some tissues, ankyrin, a key component of the membrane cytoskeleton, can regulate the trafficking of NKA. In the retina, NKA and ankyrin-B are expressed in multiple cell types and immunostaining for each is striking in the synaptic layers. Labeling for NKA is also prominent along the inner segment plasma membrane (ISPM) of photoreceptors. NKA co-immunoprecipitates with ankyrin-B, but on a subcellular level colocalization of these two proteins varies dependent on the cell type. We used transgenic Xenopus laevis tadpoles to evaluate the subcellular trafficking of NKA in photoreceptors. GFP-NKA α3 and α1 are localized to the ISPM, but α4 is localized to outer segments (OSs). We identified a VxP motif responsible for the OS targeting by using a series of chimeric and mutant NKA constructs. This motif is similar to previously identified ciliary targeting motifs. Given the structural similarities between OSs and flagella, our findings shed light on the subcellular targeting of this testes-specific NKA isoform.


Subject(s)
Ankyrins/metabolism , Flagella/enzymology , Retina/enzymology , Retinal Photoreceptor Cell Inner Segment/enzymology , Retinal Photoreceptor Cell Outer Segment/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Motifs , Animals , Ankyrins/genetics , Cattle , Cell Membrane/enzymology , Green Fluorescent Proteins/genetics , Humans , Immunoprecipitation , In Vitro Techniques , Larva/enzymology , Mice, Inbred C57BL , Organisms, Genetically Modified , Protein Subunits , Protein Transport , Signal Transduction , Sodium-Potassium-Exchanging ATPase/genetics , Species Specificity , Xenopus laevis/genetics
2.
Cell Mol Life Sci ; 72(4): 833-43, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25142030

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels carry Ih, which contributes to neuronal excitability and signal transmission in the nervous system. Controlling the trafficking of HCN1 is an important aspect of its regulation, yet the details of this process are poorly understood. Here, we investigated how the C-terminus of HCN1 regulates trafficking by testing for its ability to redirect the localization of a non-targeted reporter in transgenic Xenopus laevis photoreceptors. We found that HCN1 contains an ER localization signal and through a series of deletion constructs, identified the responsible di-arginine ER retention signal. This signal is located in the intrinsically disordered region of the C-terminus of HCN1. To test the function of the ER retention signal in intact channels, we expressed wild type and mutant HCN1 in HEK293 cells and found this signal negatively regulates surface expression of HCN1. In summary, we report a new mode of regulating HCN1 trafficking: through the use of a di-arginine ER retention signal that monitors processing of the channel in the early secretory pathway.


Subject(s)
Arginine/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Animals, Genetically Modified/metabolism , Arginine/chemistry , HEK293 Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Molecular Sequence Data , Photoreceptor Cells/metabolism , Secretory Pathway , Sequence Alignment , Sequence Homology, Amino Acid , Xenopus laevis/metabolism
3.
PLoS One ; 15(10): e0240451, 2020.
Article in English | MEDLINE | ID: mdl-33052948

ABSTRACT

Cell differentiation and cell fate determination in sensory systems are essential for stimulus discrimination and coding of environmental stimuli. Color vision is based on the differential color sensitivity of retinal photoreceptors, however the developmental programs that control photoreceptor cell differentiation and specify color sensitivity are poorly understood. In Drosophila melanogaster, there is evidence that the color sensitivity of different photoreceptors in the compound eye is regulated by inductive signals between cells, but the exact nature of these signals and how they are propagated remains unknown. We conducted a genetic screen to identify additional regulators of this process and identified a novel mutation in the hibris gene, which encodes an irre cell recognition module protein (IRM). These immunoglobulin super family cell adhesion molecules include human KIRREL and nephrin (NPHS1). hibris is expressed dynamically in the developing Drosophila melanogaster eye and loss-of-function mutations give rise to a diverse range of mutant phenotypes including disruption of the specification of R8 photoreceptor cell diversity. We demonstrate that hibris is required within the retina, and that hibris over-expression is sufficient to disrupt normal photoreceptor cell patterning. These findings suggest an additional layer of complexity in the signaling process that produces paired expression of opsin genes in adjacent R7 and R8 photoreceptor cells.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Retina/growth & development , Animals , Cell Differentiation , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Imaginal Discs/metabolism , Mutation , Organ Specificity , Photoreceptor Cells, Invertebrate/cytology , Retina/metabolism
4.
Invest Ophthalmol Vis Sci ; 56(6): 3514-21, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26030105

ABSTRACT

PURPOSE: Hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels are widely expressed in the retina. In photoreceptors, the hyperpolarization-activated current (Ih) carried by HCN1 is important for shaping the light response. It has been shown in multiple systems that trafficking HCN1 channels to specific compartments is key to their function. The localization of HCN1 in photoreceptors is concentrated in the plasma membrane of the inner segment (IS). The mechanisms controlling this localization are not understood. We previously identified a di-arginine endoplasmic reticulum (ER) retention motif that negatively regulates the surface targeting of HCN1. In this study, we sought to identify a forward trafficking signal that could counter the function of the ER retention signal. METHODS: We studied trafficking of HCN1 and several mutants by imaging their subcellular localization in transgenic X. laevis photoreceptors. Velocity sedimentation was used to assay the assembly state of HCN1 channels. RESULTS: We found the HCN1 N-terminus can redirect a membrane reporter from outer segments (OS) to the plasma membrane of the IS. The sequence necessary for this behavior was mapped to a 20 amino acid region containing a leucine-based ER export motif. The ER export signal is necessary for forward trafficking but not channel oligomerization. Moreover, this ER export signal alone counteracted the di-arginine ER retention signal. CONCLUSIONS: We identified an ER export signal in HCN1 that functions with the ER retention signal to maintain equilibrium of HCN1 between the endomembrane system and the plasma membrane.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Immunohistochemistry , Models, Animal , Synapses/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL