GRIF-1–kinesin-1 interactions: a confocal microscopy study

2006 ◽  
Vol 34 (1) ◽  
pp. 48-50 ◽  
Author(s):  
K. Pozo ◽  
F.A. Stephenson

GRIF-1 [GABAA (γ-aminobutyric acidA) receptor interacting factor-1] is a member of a coiled-coil family of proteins thought to function as adaptors in the anterograde trafficking of organelles utilizing the kinesin-1 motor proteins to synapses. To study in more detail the molecular interaction between GRIF-1 and the kinesin-1 family member KIF5C, fluorescent yellow- and fluorescent cyan-tagged GRIF-1, KIF5C, the KIF5C MD (motor domain) and the KIF5C NMD (non-motor domain) fusion proteins were generated. Each was characterized with respect to size and ability to co-associate by immunoprecipitation following expression in HEK-293 (human embryonic kidney 293) cells. Further, their distribution in transfected HEK-293 and transformed African green monkey kidney (COS-7) cells was analysed by confocal microscopy. The fluorescent GRIF-1 and KIF5C fusion proteins were all found to behave as wild-type. Double GRIF-1/KIF5C transfectants revealed co-localization. The GRIF-1/KIF5C and GRIF-1/KIF5C NMD double transfectants showed different subcellular distributions compared with single GRIF-1, KIF5C or KIF5C NMD transfections. These studies confirm the association between GRIF-1 and kinesin-1 NMDs. Fluorescence resonance energy transfer studies are ongoing to characterize this interaction in more detail.

2019 ◽  
Vol 20 (14) ◽  
pp. 3532 ◽  
Author(s):  
Noura Al-Zamel ◽  
Suleiman Al-Sabah ◽  
Yunus Luqmani ◽  
Lobna Adi ◽  
Siby Chacko ◽  
...  

Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are important regulators of metabolism, making their receptors (GLP-1R and GIPR) attractive targets in the treatment of type 2 diabetes mellitus (T2DM). GLP-1R agonists are used clinically to treat T2DM but the use of GIPR agonists remains controversial. Recent studies suggest that simultaneous activation of GLP-1R and GIPR with a single peptide provides superior glycemic control with fewer adverse effects than activation of GLP-1R alone. We investigated the signaling properties of a recently reported dual-incretin receptor agonist (P18). GLP-1R, GIPR, and the closely related glucagon receptor (GCGR) were expressed in HEK-293 cells. Activation of adenylate cyclase via Gαs was monitored using a luciferase-linked reporter gene (CRE-Luc) assay. Arrestin recruitment was monitored using a bioluminescence resonance energy transfer (BRET) assay. GLP-1, GIP, and glucagon displayed exquisite selectivity for their receptors in the CRE-Luc assay. P18 activated GLP-1R with similar potency to GLP-1 and GIPR with higher potency than GIP. Interestingly, P18 was less effective than GLP-1 at recruiting arrestin to GLP-1R and was inactive at GCGR. These data suggest that P18 can act as both a dual-incretin receptor agonist, and as a G protein-biased agonist at GLP-1R.


2006 ◽  
Vol 4 (1) ◽  
pp. nrs.04021 ◽  
Author(s):  
Kristen L. Koterba ◽  
Brian G. Rowan

Bioluminescent resonance energy transfer (BRET2) is a recently developed technology for the measurement of protein-protein interactions in a live, cell-based system. BRET2 is characterized by the efficient transfer of excited energy between a bioluminescent donor molecule (Renilla luciferase) and a fluorescent acceptor molecule (a mutant of Green Fluorescent Protein (GFP2)). The BRET2 assay offers advantages over fluorescence resonance energy transfer (FRET) because it does not require an external light source thereby eliminating problems of photobleaching and autoflourescence. The absence of contamination by light results in low background that permits detection of very small changes in the BRET2 signal. BRET2 is dependent on the orientation and distance between two fusion proteins and therefore requires extensive preliminary standardization experiments to conclude a positive BRET2 signal independent of variations in protein titrations and arrangement in tertiary structures. Estrogen receptor (ER) signaling is modulated by steroid receptor coactivator 1 (SRC-1). To establish BRET2 in a ligand inducible system we used SRC-1 as the donor moiety and ER as the acceptor moiety. Expression and functionality of the fusion proteins were assessed by transient transfection in HEK-293 cells followed by Western blot analysis and measurement of ER-dependent reporter gene activity. These preliminary determinations are required prior to measuring nuclear receptor protein-protein interactions by BRET2. This article describes in detail the BRET2 methodology for measuring interaction between full-length ER and coregulator proteins in real-time, in an in vivo environment.


2021 ◽  
Author(s):  
Nilesh Umakant Deshpande ◽  
Mishika Virmani ◽  
Manickam Jayakannan

We report aggregation induced emission (AIE) driven polysaccharide polymersome as fluorescence resonance energy transfer (FRET) nanoprobes to study their intracellular enzyme-responsive delivery by real-time live-cell confocal microscopy bio-imaging techniques. AIE...


Hypertension ◽  
2014 ◽  
Vol 64 (suppl_1) ◽  
Author(s):  
Daniel C Villela ◽  
Anke Teichmann ◽  
Sebastian Kirsch ◽  
Maibritt Mardahl ◽  
Lisa M Münter ◽  
...  

The angiotensin AT2-receptor (AT2R) and the receptor MAS share a strinkingly similar spectrum of signaling mechanisms and protective, physiological actions. Furthermore, cross-inhibition by the respective receptor antagonists has been observed. Therefore we hypothesised that a physical interaction between these two receptors may exist. HEK-293 cells were transfected with vectors encoding MAS or AT2R fused in the C-terminus with the fluorophores CFP or YFP for FRET and GFP or mCherry for FCCS. FRET with photobleaching was used to detect, whether MAS and AT2R are localised in very close proximity (1-10nm) in cell membranes thus indicating dimerisation. FCCS was used to follow simultaneously occurring fluctuations in fluorescence intensity of both labeled molecules. Several controls were applied such as co-transfection of equal amounts of fused and non-fused MAS/AT2R expression vectors for competition, co-tranfection of coding and uncoding pcDNA vectors or co-transfection with an unrelated transmembrane receptor. Experiments were conducted under baseline conditions and in cells treated with AT2R/MAS agonists and antagonists Significant FRET efficiency of 10.8±0.8% was measured for AT2-YFP/MAS-CFP strongly indicating heterodimerisation. FRET efficiency was not altered by AT2R or MAS agonists or antagonists. Non-fluorescent MAS and AT2R competed with fluorescent receptors as indicated by a 50% reduction in FRET efficiency (6.0±0.6%), while empty vectors did not compete (9.6±0.6%). No FRET efficiency was observed with an unrelated transmembrane receptor (0.44±1.44%) indicating specificity of receptor interactions. Both, MAS and AT2R also formed homodimers (7.4±0.8% for MAS, 9.2±0.8% for AT2R). Hetero- and homodimerisations were absent if amino acid C35 of the AT2R was mutated (3,9 ± 1,2%). FCCS corroborated the FRET results and revealed a significantly enhanced cross correlation in cells tranfected with fluorophore-tagged MAS/AT2R when compared to vectors only expressing the fluorophores (8.5±3% vs 11.1±4%; p<0.0001). Our data strongly suggest that MAS and the AT2R form homo- and heterodimers. Studies to investigate the physiological relevance of MAS/AT2R dimerisation are currently being conducted.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2028-2028
Author(s):  
Abdelouahid El Khattouti ◽  
Volker R. Stoldt ◽  
Rüdiger E. Scharf

Abstract Abstract 2028 Background and Objectives: The HPA-1 polymorphism of αIIbβ3 arises from a Leu→Pro exchange at residue 33 of the β3 subunit resulting in HPA-1a (Leu33) or HPA-1b (Pro33). We have documented that patients with coronary artery disease who are carriers of HPA-1b (Pro33) experience their myocardial infarction 5.2 years earlier than HPA-1a/1a (Leu33) patients (J Thromb Haemost 2005; 3: 1522). Based on these observations, it has been postulated that HPA-1b (Pro33) is a prothrombotic variant of αIIbβ3. We have now generated a model overexpressing fluorescent proteins fused with αIIbβ3 in transfected HEK293 cells. Methods: :A yellow protein (YFP) and a cyan fluorescent protein (CFP) were cloned to the C-termini of the β3 and αIIb subunits prior to transfection of HEK293 cells, subsequently expressing the fusion proteins of both HPA-1 isoforms. Using flow cytometry, Western blotting and specific antibodies directed against αIIb or β3, we identified 12 HPA-1a and 11 HPA-1b positive clones. For further experiments only those cell lines expressing equal amounts of fluorescent fusion proteins, i.e. a 140 kD αIIb-CFP and a 113 kD β3-YFP, were used. Results: Functional integrity of both integrin variants and proper membrane insertion were documented by intact activation of transfected HEK293 cells through G protein-coupled receptors with organic acid (1-stearoyl-2-arachidonoyl-sn-glycerol) or direct phorbol 12-myristate 13-acetate-induced stimulation of protein kinase C and by specific binding of Alexa488 fibrinogen to αIIbβ3 in response to inside-out signaling. In the presencence of pertussis toxin or abciximab, activation or ligand binding of αIIbβ3 were completely (>98%) inhibited in both isoforms. Activation of αIIbβ3 stimulates the tyrosine kinase Src, constitutively associated with the the β-subunit of the integrin. To determine whether αIIbβ3-dependent outside-in signaling is responsible for a polymorphism-related modulation, we performed adhesion experiments under static conditions with fibrinogen (50 μg/ml) in the absence or presence of Mn2+ (0.5 mM). Specific activation of the phosphotyrosine motif (Src-pY418), as determined by Western blotting and quantified by densitometry (ratio of Src-pY418/total Src), was 15 + 1.5% higher in HPA-1b than HPA-1a cells in the presence of Mn2+ (n=6 independent experiments, p<0.01). To explore the molecular nature of this difference in terms of putative changes in the allostery of integrin αIIbβ3 with regard to the HPA-1 polymorphism, dynamic measurements were performed using fluorescence resonance energy transfer (FRET). The relative decrease in FRET signal, indicating spatial separation of the cytoplasmic tails of the α- and β-subunit as a consequence of integrin activation, was recorded every minute over 0.5 hrs in transfected HEK293 cells adherent onto fibrinogen. At every time point, the kinetic measurements revealed a significantly faster and more distict (> 5%) decrease in HPA-1b than in HPA-1a cells under static adhesion (p<0.009). Upon exposure of adherent HEK293 cells to increasing shear rates (stepwise elevation from 50 to 1600 sec-1 by doubling the initial shear rate every minute), the spatial separation of the integrin subunits occurred significantly faster and more distinct (> 10%) in HPA-1b (Pro33) than HPA-1a (Leu33) cells in response to shear (p<0.0014). Under the same conditions, the rate of HPA-1b cells still adherent onto immobilized fibrinogen was 80%, while the relative number of residual HPA-1a cells decreased to 20% upon exposure to 1600 sec-1 (p<0.0001). These displacement experiments suggest that the HPA-1b (Pro33) variant is more resistant to biomechanical stress than the HPA-1a (Leu33) isoform. Conclusions: Our findings suggest that the HPA-1 polymorphism can have a significant impact on the activation of αIIbβ3. This is evident from a higher outside-in signaling and a higher resistance to biomechanical stress upon exposure to increasing shear of HPA-1b (Pro33) in comparison with HPA-1a (Leu33) transfectants. The difference in spatial separation of the cytoplasmic tails of the integrin in response to activation, as demonstrated by FRET analyses under static and flow dynamic conditions, reflects allosteric changes that may contribute to the prothrombotic phenotype of the HPA-1b (Pro33) variant. Disclosures: No relevant conflicts of interest to declare.


1998 ◽  
Vol 72 (11) ◽  
pp. 8586-8596 ◽  
Author(s):  
Xiao Huan Liang ◽  
Linda K. Kleeman ◽  
Hui Hui Jiang ◽  
Gerald Gordon ◽  
James E. Goldman ◽  
...  

ABSTRACT bcl-2, the prototypic cellular antiapoptotic gene, decreases Sindbis virus replication and Sindbis virus-induced apoptosis in mouse brains, resulting in protection against lethal encephalitis. To investigate potential mechanisms by which Bcl-2 protects against central nervous system Sindbis virus infection, we performed a yeast two-hybrid screen to identify Bcl-2-interacting gene products in an adult mouse brain library. We identified a novel 60-kDa coiled-coil protein, Beclin, which we confirmed interacts with Bcl-2 in mammalian cells, using fluorescence resonance energy transfer microscopy. To examine the role of Beclin in Sindbis virus pathogenesis, we constructed recombinant Sindbis virus chimeras that express full-length human Beclin (SIN/beclin), Beclin lacking the putative Bcl-2-binding domain (SIN/beclinΔBcl-2BD), or Beclin containing a premature stop codon near the 5′ terminus (SIN/beclinstop). The survival of mice infected with SIN/beclin was significantly higher (71%) than the survival of mice infected with SIN/beclinΔBcl-2BD (9%) or SIN/beclinstop (7%) (P < 0.001). The brains of mice infected with SIN/beclin had fewer Sindbis virus RNA-positive cells, fewer apoptotic cells, and lower viral titers than the brains of mice infected with SIN/beclinΔBcl-2BD or SIN/beclinstop. These findings demonstrate that Beclin is a novel Bcl-2-interacting cellular protein that may play a role in antiviral host defense.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Aliza T. Ehrlich ◽  
Meriem Semache ◽  
Pierre Couvineau ◽  
Stefan Wojcik ◽  
Hiroyuki Kobayashi ◽  
...  

AbstractThe atypical chemokine receptor 3, ACKR3, is a G protein-coupled receptor, which does not couple to G proteins but recruits βarrestins. At present, ACKR3 is considered a target for cancer and cardiovascular disorders, but less is known about the potential of ACKR3 as a target for brain disease. Further, mouse lines have been created to identify cells expressing the receptor, but there is no tool to visualize and study the receptor itself under physiological conditions. Here, we engineered a knock-in (KI) mouse expressing a functional ACKR3-Venus fusion protein to directly detect the receptor, particularly in the adult brain. In HEK-293 cells, native and fused receptors showed similar membrane expression, ligand induced trafficking and signaling profiles, indicating that the Venus fusion does not alter receptor signaling. We also found that ACKR3-Venus enables direct real-time monitoring of receptor trafficking using resonance energy transfer. In ACKR3-Venus knock-in mice, we found normal ACKR3 mRNA levels in the brain, suggesting intact gene transcription. We fully mapped receptor expression across 14 peripheral organs and 112 brain areas and found that ACKR3 is primarily localized to the vasculature in these tissues. In the periphery, receptor distribution aligns with previous reports. In the brain there is notable ACKR3 expression in endothelial vascular cells, hippocampal GABAergic interneurons and neuroblast neighboring cells. In conclusion, we have generated Ackr3-Venus knock-in mice with a traceable ACKR3 receptor, which will be a useful tool to the research community for interrogations about ACKR3 biology and related diseases.


2019 ◽  
Vol 116 (10) ◽  
pp. 4275-4284 ◽  
Author(s):  
Stefan Niekamp ◽  
Jongmin Sung ◽  
Walter Huynh ◽  
Gira Bhabha ◽  
Ronald D. Vale ◽  
...  

Light microscopy is a powerful tool for probing the conformations of molecular machines at the single-molecule level. Single-molecule Förster resonance energy transfer can measure intramolecular distance changes of single molecules in the range of 2 to 8 nm. However, current superresolution measurements become error-prone below 25 nm. Thus, new single-molecule methods are needed for measuring distances in the 8- to 25-nm range. Here, we describe methods that utilize information about localization and imaging errors to measure distances between two different color fluorophores with ∼1-nm accuracy at distances >2 nm. These techniques can be implemented in high throughput using a standard total internal reflection fluorescence microscope and open-source software. We applied our two-color localization method to uncover an unexpected ∼4-nm nucleotide-dependent conformational change in the coiled-coil “stalk” of the motor protein dynein. We anticipate that these methods will be useful for high-accuracy distance measurements of single molecules over a wide range of length scales.


2012 ◽  
Vol 303 (6) ◽  
pp. F800-F811 ◽  
Author(s):  
Abdel A. Alli ◽  
Hui-Fang Bao ◽  
Alia A. Alli ◽  
Yasir Aldrugh ◽  
John Z. Song ◽  
...  

Phosphatidylinositol phosphates (PIPs) are known to regulate epithelial sodium channels (ENaC). Lipid binding assays and coimmunoprecipitation showed that the amino-terminal domain of the β- and γ-subunits of Xenopus ENaC can directly bind to phosphatidylinositol 4,5-bisphosphate (PIP2), phosphatidylinositol 3,4,5-trisphosphate (PIP3), and phosphatidic acid (PA). Similar assays demonstrated various PIPs can bind strongly to a native myristoylated alanine-rich C-kinase substrate (MARCKS), but weakly or not at all to a mutant form of MARCKS. Confocal microscopy demonstrated colocalization between MARCKS and PIP2. Confocal microscopy also showed that MARCKS redistributes from the apical membrane to the cytoplasm after PMA-induced MARCKS phosphorylation or ionomycin-induced intracellular calcium increases. Fluorescence resonance energy transfer studies revealed ENaC and MARCKS in close proximity in 2F3 cells when PKC activity and intracellular calcium concentrations are low. Transepithelial current measurements from Xenopus 2F3 cells treated with PMA and single-channel patch-clamp studies of Xenopus 2F3 cells treated with a PKC inhibitor altered Xenopus ENaC activity, which suggest an essential role for MARCKS in the regulation of Xenopus ENaC activity.


Sign in / Sign up

Export Citation Format

Share Document