scholarly journals Suppression of integrin activation by the membrane-distal sequence of the integrin alphaIIb cytoplasmic tail

2004 ◽  
Vol 379 (2) ◽  
pp. 317-323 ◽  
Author(s):  
Jun YAMANOUCHI ◽  
Takaaki HATO ◽  
Tatsushiro TAMURA ◽  
Shigeru FUJITA

Integrin cytoplasmic tails regulate integrin activation including an increase in integrin affinity for ligands. Although there is ample evidence that the membrane-proximal regions of the α and β tails interact with each other to maintain integrins in a low-affinity state, little is known about the role of the membrane-distal region of the α tail in regulation of integrin activation. We report a critical sequence for regulation of integrin activation in the membranedistal region of the αIIb tail. Alanine substitution of the RPP residues in the αIIb tail rendered αIIbβ3 constitutively active in a metabolic energy-dependent manner. Although an αIIb/α6Aβ3 chimaeric integrin, in which the αIIb tail was replaced by the α6A tail, was in an energy-dependent active state to bind soluble ligands, introduction of the RPP sequence into the α6A tail inhibited binding of an activation-dependent antibody PAC1. In αIIb/α6Aβ3, deleting the TSDA sequence from the α6A tail or single amino acid substitutions of the TSDA residues inhibited αIIb/α6Aβ3 activation and replacing the membrane-distal region of the αIIb tail with TSDA rendered αIIbβ3 active, suggesting a stimulatory role of TSDA in energy-dependent integrin activation. However, adding TSDA to the αIIb tail containing the RPP sequence of the membrane-distal region failed to activate αIIbβ3. These results suggest that the RPP sequence after the GFFKR motif of the αIIb tail suppresses energy-dependent αIIbβ3 activation. These findings provide a molecular basis for the regulation of energy-dependent integrin activation by α subunit tails.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1545-1545
Author(s):  
Kelly Aylward ◽  
Marc Devocelle ◽  
Niamh Moran

Abstract The platelet-specific integrin αIIbβ3 plays a critical role in platelet aggregation and pathological thrombosis. Integrin affinity and ligand binding are regulated by the highly conserved αIIb membrane-proximal motif 989KVGFFKR995. We have recently shown that this motif is dependent on the presence of two phenylalanines (FF) for its activity. In order to investigate the role of KVGFFKR on integrin transmembrane signaling we used two parallel systems: (1) stable Chinese Hamster Ovary (CHO) cells expressing mutant αIIbβ3 integrins and (2) platelets treated with synthetic palmitylated (pal) peptides corresponding to the seven amino acid motif. In CHO cells, we chose cytoskeletal reorganization as a means to explore outside-in signaling. Alanine substitutions were introduced to the α-subunit KVGFFKR domain and co-expressed with wildtype β3. Cells were stably transfected with wildtype αIIb(992FF993)/β3, αIIb(992AA993)/β3 and αIIb (992AF993)/β3 to produce the FF, AA and AF cells respectively. Their ability to reorganize their cytoskeleton upon adhesion to fibrinogen was then determined. Even though double alanine substitution produced a constitutively activated integrin, the AA cells were unable to give rise to cytoskeletal reassembly as seen in the FF and AF cells. Using phalloidin as a marker, the AA cells displayed polymerized F-actin but failed to show the elaborate elongated stress fibers formed in the FF and AF cells. To further investigate the role of the KVGFFKR motif on downstream signaling events, we focused on using pal-peptides in platelets. We have shown that in addition to stimulating platelet aggregation presumably by facilitating the spatial separation of the integrin cytoplasmic tails, pal-KVGFFKR (pal-FF) induced tyrosine phosphorylation even in the absence of ligand (EDTA:5mM) or (ReoPro:10μg/ml). The tyrosine phosphoproteome associated with alanine-substituted peptides pal-KVGAFKR (pal-AF) and pal-KVGFAKR (pal-FA) was similar to that of pal-FF. However there is a remarkable absence of a specific 100kDa band (probably α-actinin) in the phosphoprotein profile in response to pal-KVGAAKR (pal-AA) both with peptide treatment alone and in the presence of TRAP. A closer look at ppFAK125 revealed that its tyrosine phosphorylation is also inhibited by pal-AA. Since α-actinin and ppFAK125 phosphorylation are closely linked events it supports α-actinin as the 100kDa missing phosphoprotein. However, pal-AA did not inhibit ppSyk72or ppSrc60 activation. Moreover pal-AA was identified as a potent antagonist, inhibiting platelet aggregation, PAC-1 binding and tyrosine phosphorylation. In summary, a double alanine substitution of the α-subunit membrane proximal domain disturbs cytoskeletal reorganization downstream, even though this substitution produces a constitutively activated integrin. This suggests a signaling role for the conserved α-integrin motif in addition to regulating integrin affinity. Furthermore in platelets, pal-FF peptide, by mimicking the endogenous αIIb KVGFFKR sequence can both activate the integrin and contribute to an intracellular signaling response even when ligand binding is absent. Taken together, both the stable cell system and pal-peptides in platelets support a role for the KVGFFKR domain in outside-in signaling. Also since pal-AA is an antagonist of integrin function it highlights the complexity of the proximal regulation of αIIbβ3 activation and suggests a dual role for this motif in integrin activation and intracellular signaling.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1849-1849 ◽  
Author(s):  
Seiji Tadokoro ◽  
Masamichi Shiraga ◽  
Hirokazu Kashiwagi ◽  
Tsuyoshi Kamae ◽  
Masao Akiyama ◽  
...  

Abstract Integrin activation is regulated by many different biochemical signaling pathways through the integrin cytoplasmic tails. Multiprotein complexes assembled around the integrin cytoplasmic tail are linked to the actin cytoskeleton. Binding of the cytoskeletal proteins to integrin cytoplasmic tails leads to the conformational rearrangements of integrin extracellular domains that modulate their affinity. Talin-1 or Kindlin-3 has been identified as integrin activator complex proteins. α-Actinin also links the cytoplasmic domains of integrin β tails to actin filaments. We report here a new role for α-actinin in inside-out integrin activation. To explore the role of α-actinin in inside-out signaling, platelets were stimulated with protease-activated receptor (PAR) - activating peptides (AP) under non-stirring condition for up to 20 min. Immunoprecipitation with anti-αIIbβ3 followed by immnoblotting with anti-α-actinin revealed that in resting platelets α-actinin was constitutively associated with αIIbβ3. When platelets were stimulated by PAR1-AP, α-actinin was dissociated from αIIbβ3 as an initial step. Interestingly α-actinin re-bound to αIIbβ3 at 20 min after PAR1-AP stimulation. In contrast to PAR1-AP stimulation, the α-actinin dissociation from αIIbβ3 induced by PAR4-AP was long-lasting. To reveal the dynamic changes in αIIbβ3 activation, we recently developed initial velocity analysis for PAC1 binding. In brief, FITC-PAC1 was added to the activated platelets at indicated time points after stimulation and incubated for only 30 seconds to get the PAC1 binding velocity at the time points in question. The velocity of PAC1 binding reflects the relative numbers of activated αIIbβ3 at the time points. This initial velocity analysis more clearly revealed that PAR1-AP stimulation induced only transient αIIbβ3 activation, whereas PAR4-AP induced long-lasting αIIbβ3 activation. Moreover, the dissociation of α-actinin from αIIbβ3 appears to correlate with the time-dependent changes in the number of activated αIIbβ3. The kinetics of α-actinin-αIIbβ3 interaction was synchronized with tyrosine phosphorylation of α-actinin. When stimulated with PAR1-AP, α-actinin was de-phosphorylated rapidly and re-phosphorylated in late phase. PAR4-AP induced more prolonged de-phosphorylation of α-actinin than PAR1-AP. Thus, these results suggest that the interaction between α-actinin and αIIbβ3 may correlate with inside-out signaling induced by PAR1-AP and PAR4-AP. In platelets from a patient with Glanzmann thrombasthenia the phosphotyrosine profile of α-actinin was almost the same as that of control platelets in both PAR1-AP and PAR4-AP stimulation, confirming that these changes are not mediated αIIbβ3 outside-in signaling. In sharp contrast PAR4-AP stimulation failed to induce the sustained de-phosphorylation of α-actinin in P2Y12-ADP receptor deficient platelets. The blockade of P2Y12 with AR-C69931MX impaired the levels of activated αIIbβ3 induced by PAR4-AP, which correlated with the re-association of α-actinin. To further examine the role for α-actinin in integrin activation, α-actinin was overexpressed in human megakaryoblastic CMK cells and PAR1- AP induced PAC-1 binding to αIIbβ3 was assessed. Initial velocity analysis on CMK cells showed that overexpressed α-actinin inhibited PAR1-AP induced αIIbβ3 activation. These data imply that the binding of α-actinin to αIIbβ3 may regulate the levels of αIIbβ3 activation. Our observations may provide a new molecular framework for understanding the functions of β3 integrins in platelets.


2005 ◽  
Vol 288 (2) ◽  
pp. H936-H945 ◽  
Author(s):  
C. Radel ◽  
V. Rizzo

To identify the role of caveolin-1 in integrin mechanotransduction, we exposed bovine aortic endothelial cells to 10 dyn/cm2 of laminar shear stress. Caveolin-1 was acutely and transiently phosphorylated with shear, occurring downstream of β1-integrin activation as the β1-integrin blocking antibody JB1A was inhibitory. In manipulating Src family kinase (SFK) activity with knockdown of Csk or type 1 protein phosphatase (PP1) treatment, we observed coordinate increase and decrease in shear-induced caveolin-1 phosphorylation, respectively. Hence, shear-stimulated caveolin-1 phosphorylation is regulated by SFKs. Shear-induced recruitment and phosphorylation of caveolin-1 occurred at β1-integrin sites in a β1-integrin- and SFK-dependent manner. Csk, described to interact with pY14-caveolin-1 and integrins, bound to an increased pool of phosphorylated caveolin-1 after shear corresponding with elevated Csk at β1-integrin sites. Like caveolin-1, treatment with JB1A and PP1 attenuated shear-induced Csk association with β1-integrins. Csk function was assayed with transfection of a caveolin-1 phosphorylation domain peptide. The peptide attenuated shear-induced association of Csk at β1-integrin sites, as well as colocalization of Csk with paxillin and phosphorylated caveolin-1. Because integrin and Csk activity regulate cytoskeletal reorganization, we evaluated the role of this mechanism in shear-induced myosin light chain (MLC) phosphorylation. Knockdown of Csk expression was sufficient to reduce MLC diphosphorylation due to shear. Disruption of Csk-integrin association by peptide treatment was also inhibitory of the MLC diphosphorylation response. Together these data indicate that integrin activation with shear stress results in SFK-regulated caveolin-1 phosphorylation that, in turn, mediates Csk association at integrin sites, where it plays a role in downstream, shear-stimulated MLC diphosphorylation.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1431-1431
Author(s):  
Jieqing Zhu ◽  
Jiafu Liu ◽  
Yan-Qing Ma ◽  
Zhengli Wang

Abstract Integrin inside-out activation is essential for platelet aggregation mediated by αIIbβ3 and leukocytes migration and arresting mediated by αLβ2. How integrin is activated by the inside-out stimulation is not completely understood. Integrin activation from inside the cell is regulated through the transmembrane and cytoplasmic domains. Mutagenesis and structural studies revealed that the inactive integrin conformation is maintained by the specific interactions at the transmembrane and cytoplasmic domains. Inside-out signals impinging on integrin cytoplasmic domain disturb the transmembrane and cytoplasmic associations, resulting in conformational change of extracellular domain that is required for binding ligands. Studies on the mechanism of integrin inside-out activation have been focused on β cytoplasmic tail that is relatively conserved and bears binding sites for the common intracellular activators including talin and kindlin. The integrin α cytoplasmic tails only share a conserved GFFKR motif at the membrane-proximal region that forms specific interface with the membrane-proximal region of β cytoplasmic tail. The membrane-distal regions after the GFFKR motif are diverse significantly both in length and sequence. Their roles in integrin activation have not been well characterized. In this study, by comprehensive mutagenesis, we defined the role of the membrane-distal region of α integrin cytoplasmic tail in maintaining integrin in the resting state and in integrin inside-out activation. We found that complete deletion of the αIIb cytoplasmic membrane-distal region greatly enhances αIIbβ3 activation induced by the active mutations such as β3-K716A and β3-G708L, indicating that the missing of membrane-distal region facilitates integrin activation, i.e. the αIIb membrane-distal region contributes to the inactive integrin conformation. On the other hand, complete deletion of the αIIb membrane-distal region abolished integrin activation induced by the active mutations of αIIb-R995 and β3-D723, indicating that the αIIb membrane-distal region also contributes to integrin inside-out activation. We demonstrated that deletion of the membrane-distal region of αIIb, αV, or αL integrin greatly diminished ligand binding induced by overexpression of talin-1 head and/or kindlin-2 or -3 in 293FT cells. We further confirmed the effect of α cytoplasmic membrane-distal region on integrin inside-out activation in K562 cells. In the absence of αIIb cytoplasmic membrane-distal region, PMA failed to induce ligand binding to αIIbβ3 integrin expressed in K562 cells. This effect was due to the lack of talin-1-head and kindlin-induced integrin conformational change (ectodomain extension and headpiece opening) in the absence of α cytoplasmic membrane-distal region as reported by the conformation-dependent monoclonal antibodies. Structural superposition of αIIbβ3 transmembrane-cytoplasmic heterodimer and talin-1-head/β-tail complex reveals steric clashes between talin-1 head and the αIIb membrane-distal residues (NR997) immediately follow the GFFKR motif, which has been suggested to play a role in talin-mediated integrin activation. To test this possibility, we retained two native residues, NR997 for the αIIb membrane-distal region and found that it partially restores talin-1-head-induced integrin activation. Replacing the NR997 with small amino acids, GG997 or AA997 has little effect, while with the bulky residues YY997 significantly reduced talin-1-head-induced αIIbβ3 activation. Interestingly, retaining two native residues for the membrane-distal region of αV or αL integrin failed to restore talin-1-head-induced αVβ3 or αLβ2 activation. Retaining as long as 8 native residues for the αL membrane-distal region is not sufficient to restore talin-1-head-induced αLβ2 activation to the level of intact αL. These data demonstrate that a steric clash might play a role but is not the sole mechanism by which the α cytoplasmic membrane-distal region participates in integrin inside-out activation. A proper length and amino acids of the membrane-distal region is required for talin-induced integrin activation. Our data established an essential role of the α integrin cytoplasmic membrane-distal region in integrin activation and provide new insight of how talin and kindlin induce the high affinity integrin conformation that is required for fully functional integrins. Disclosures No relevant conflicts of interest to declare.


2000 ◽  
Vol 116 (6) ◽  
pp. 795-824 ◽  
Author(s):  
S. Nikonov ◽  
T.D. Lamb ◽  
E.N. Pugh

We investigated the kinetics and sensitivity of photocurrent responses of salamander rods, both in darkness and during adaptation to steady backgrounds producing 20–3,000 photoisomerizations per second, using suction pipet recordings. The most intense backgrounds suppressed 80% of the circulating dark current and decreased the flash sensitivity ∼30-fold. To investigate the underlying transduction mechanism, we expressed the responses as a fraction of the steady level of cGMP-activated current recorded in the background. The fractional responses to flashes of any fixed intensity began rising along a common trajectory, regardless of background intensity. We interpret these invariant initial trajectories to indicate that, at these background intensities, light adaptation does not alter the gain of any of the amplifying steps of phototransduction. For subsaturating flashes of fixed intensity, the fractional responses obtained on backgrounds of different intensity were found to “peel off” from their common initial trajectory in a background-dependent manner: the more intense the background, the earlier the time of peeling off. This behavior is consistent with a background-induced reduction in the effective lifetime of at least one of the three major integrating steps in phototransduction; i.e., an acceleration of one or more of the following: (1) the inactivation of activated rhodopsin (R*); (2) the inactivation of activated phosphodiesterase (E*, representing the complex Gα–PDE of phosphodiesterase with the transducin α-subunit); or (3) the hydrolysis of cGMP, with rate constant β. Our measurements show that, over the range of background intensities we used, β increased on average to ∼20 times its dark-adapted value; and our theoretical analysis indicates that this increase in β is the primary mechanism underlying the measured shortening of time-to-peak of the dim-flash response and the decrease in sensitivity of the fractional response.


Cells ◽  
2021 ◽  
Vol 10 (4) ◽  
pp. 825
Author(s):  
Katarzyna Bialkowska ◽  
Jun Qin ◽  
Edward F. Plow

Integrins serve as conduits for the transmission of information between cells and their extracellular environment. Signaling across integrins is bidirectional, transducing both inside-out and outside-signaling. Integrin activation, a transition from a low affinity/avidity state to a high affinity/avidity state for cognate ligands, is an outcome of inside-signaling. Such activation is particularly important for the recognition of soluble ligands by blood cells but also influences cell-cell and cell-matrix interactions. Integrin activation depends on a complex series of interactions, which both accelerate and inhibit their interconversion from the low to the high affinity/avidity state. There are three components regarded as being most proximately involved in integrin activation: the integrin cytoplasmic tails, talins and kindlins. The participation of each of these molecules in integrin activation is highly regulated by post-translation modifications. The importance of targeted phosphorylation of integrin cytoplasmic tails and talins in integrin activation is well-established, but much less is known about the role of post-translational modification of kindlins. The kindlins, a three-member family of 4.1-ezrin-radixin-moesin (FERM)-domain proteins in mammals, bind directly to the cytoplasmic tails of integrin beta subunits. This commentary provides a synopsis of the emerging evidence for the role of kindlin phosphorylation in integrin regulation.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3453-3453
Author(s):  
Aye Myat Myat Thinn ◽  
Jieqing Zhu

Abstract Integrins are α/β heterodimeric cell adhesion receptors with each subunit comprising of a large extracellular domain, a single-spanning transmembrane domain, and usually a short cytoplasmic tail. Different combinations of 18 α and 8 β subunits make up 24 integrin members that recognize diverse extracellular ligands important in numerous biological functions such as immune responses, maintenance of hemostasis, and development. Abnormal activation of integrin is associated with many pathological conditions including thrombosis, inflammatory diseases, as well as tumor-driven cell growth, metastasis, and angiogenesis. Therefore, tight regulation is crucial in integrin activation. Recent structural and functional studies have shown that integrin activation is regulated by the cytoplasmic tails. Studies on the mechanism of integrin activation from inside the cell (namely inside-out activation) have been focused on the β cytoplasmic tail that is relatively conserved and bears binding sites for the common intracellular activators such as talin and kindlin. However, the role of α cytoplasmic tail in integrin activation remains elusive. The integrin α cytoplasmic tails share a conserved GFFKR motif at the membrane-proximal region that forms a specific interface with the membrane-proximal region of the β cytoplasmic tail. In contrast, the membrane-distal (MD) regions following the GFFKR motif are diverse significantly both in length, sequence and structure when reported, and their roles in integrin activation have not been well characterized. Our recent studies demonstrated that the α-MD region is required for talin and kindlin-induced activation of αIIb, αV, and αL integrins and suggest that the sequence diversity of the α-MD region might play a role in the regulation of integrin activation. In this study, we further examined the role of α-MD regions in integrin inside-out activation using αIIb, αL, and α5 integrins as platforms. Each MD region of αIIb, αL, and α5 was replaced with those of other α subunits that heterodimerize with β3, β2, and β1 integrins, respectively. β3 subunit forms heterodimers with αIIb and αV integrins. β2 subunit forms heterodimers with αL, αM, αD, and αX integrins. β1 subunit forms heterodimers with α1, α2, α3, α4, α5, α6, α7, α8, α9, α10, α11, and αV integrins. Thus, using these integrin α-chimeras, we were able to systemically study the role of 17 α-MD regions in integrin inside-out activation while retaining the native association of α and β subunits at the cytoplasmic domains. Ligand-mimetic mAb PAC-1, intercellular adhesion molecule-1 (ICAM-1), and human fibronectin were used to measure the talin-head-induced activation of αIIb, αL, and α5 chimeras co-expressed in HEK293FT cells with β3, β2, and β1 integrins, respectively. Conformation-specific monoclonal antibodies were used to report integrin conformational activation. The endogenous α5β1 integrin of HEK293FT cells were knocked out by the CRISPR/Cas9 technology. Our data showed that the chimeric α integrins had different levels of inside-out activation when compared with their corresponding wild-type integrins. Some chimeras such as αIIb-αV, αL-αX, αL-αD, αL-αM, α5-α2, α5-α4, and α5-α9 showed lower integrin activation than the wild types, while other chimeras such as α5-α7 and α5-α10 rendered α5β1 integrin more active than wild type. As a control, the αIIb-α1 and αIIb-αL chimeras all showed higher inside-out activation than wild-type αIIb. Our results suggest that specific amino acids of the α-MD region that immediately follow the GFFKR motif might contribute to integrin inside-out activation, probably through regulating the conformational change of the integrin α transmembrane and cytoplasmic domains. Our study demonstrates an important role of the α-MD region in integrin activation and indicates that structure and sequence diversity of the α-MD region might contribute to the diverse functions of integrins, which are determined by different integrin α subunits. Disclosures No relevant conflicts of interest to declare.


2014 ◽  
Vol 34 (suppl_1) ◽  
Author(s):  
Feng Ye ◽  
Ang Li ◽  
Qiang Guo ◽  
Weiming Hu

Increases in ligand binding to integrins (“activation”) play critical roles in platelet and leukocyte function. Integrin activation requires talin and kindlin binding to integrin β cytoplasmic tails. Much research has focused on the conserved GFFKR motif in integrin αIIb tails for its importance in keeping integrins inactive and integrin β cytoplasmic tails and their interacting partners. However, the roles of αIIb tail distal of GFFKR motif are unexplored. Here, we examine the role of αIIb tail distal of GFFKR in talin-mediated inside-out integrin activation, αIIbβ3 outside-in signaling and αIIbβ3-talin interactions. Deletion of amino acid residues after the GFFKR motif in αIIb tail abolished talin-induced inside-out αIIbβ3 activation without affecting αIIbβ3-talin interaction or outside-in αIIbβ3 signaling in modeled cell systems, measured by cell spreading and Src phosphorylation. Addition of non-homologous or non-specific amino acids to the GFFKR motif restored the capacity of talin to activate αIIbβ3 in modeled cells. Moreover, thrombin-stimulated αIIbβ3 activation in megakaryocytic leukemia cells (CMK cells) are similarly abolished by truncation after GFFKR and restored by adding non-specific sequences. Furthermore, Molecular modeling indicates that β3-bound talin sterically clashes with the αIIb tail in the αIIbβ3 complexes, potentially disfavoring the α-β interactions that keep αIIbβ3 inactive. Thus, the αIIb tail sequences distal of GFFKR participate in talin-mediated inside-out αIIbβ3 activation through its steric clashes with β3-bound talin.


2002 ◽  
Vol 184 (16) ◽  
pp. 4520-4528 ◽  
Author(s):  
Angela H. Finney ◽  
Robert J. Blick ◽  
Katsuhiko Murakami ◽  
Akira Ishihama ◽  
Ann M. Stevens

ABSTRACT During quorum sensing in Vibrio fischeri, the luminescence, or lux, operon is regulated in a cell density-dependent manner by the activator LuxR in the presence of an acylated homoserine lactone autoinducer molecule [N-(3-oxohexanoyl) homoserine lactone]. LuxR, which binds to the lux operon promoter at a position centered at −42.5 relative to the transcription initiation site, is thought to function as an ambidextrous activator making multiple contacts with RNA polymerase (RNAP). The specific role of the α-subunit C-terminal domain (αCTD) of RNAP in LuxR-dependent transcriptional activation of the lux operon promoter has been investigated. The effects of 70 alanine substitution variants of the α subunit were determined in vivo by measuring the rate of transcription of the lux operon via luciferase assays in recombinant Escherichia coli. The mutant RNAPs from strains exhibiting at least twofold-increased or -decreased activity in comparison to the wild type were further examined by in vitro assays. Since full-length LuxR has not been purified, an autoinducer-independent N-terminally truncated form of LuxR, LuxRΔN, was used for in vitro studies. Single-round transcription assays were performed using reconstituted mutant RNAPs in the presence of LuxRΔN, and 14 alanine substitutions in the αCTD were identified as having negative effects on the rate of transcription from the lux operon promoter. Five of these 14 α variants were also involved in the mechanisms of both LuxR- and LuxRΔN-dependent activation in vivo. The positions of these residues lie roughly within the 265 and 287 determinants in α that have been identified through studies of the cyclic AMP receptor protein and its interactions with RNAP. This suggests a model where residues 262, 265, and 296 in α play roles in DNA recognition and residues 290 and 314 play roles in α-LuxR interactions at the lux operon promoter during quorum sensing.


2003 ◽  
Vol 376 (2) ◽  
pp. 449-456 ◽  
Author(s):  
Alexander DIETRICH ◽  
Alexander SCHEER ◽  
Daria ILLENBERGER ◽  
Yoel KLOOG ◽  
Yoav I. HENIS ◽  
...  

The α and βγ subunits of heterotrimeric G-proteins contain specific lipid modifications, which are required for their biological function. However, the relevance of these modifications to the interactions within the heterotrimeric G-protein is not fully understood. In order to explore the role of the S-prenyl moiety of the isoprenylated βγ dimer of retinal transducin, βγt, in the formation of the heterotrimeric complex with the corresponding N-acylated α subunit, αt, we employed purified fully processed subunits, which are soluble in aqueous solutions without detergents. Pertussis-toxin-mediated [32P]ADP-ribosylation of αt is strongly stimulated (≈10-fold) in the presence of βγt and can thus serve as a measure for heterotrimer formation. Using this assay, preincubation of αt with S-prenyl analogues containing farnesyl or geranylgeranyl moieties was found to inhibit heterotrimer formation in a dose-dependent manner. The inhibition was competitive and reversible, as indicated by its reversal upon increase of the βγt dimer concentration or by removal of the S-prenyl analogue using gel filtration. The competitive nature of the inhibition is supported by the marked attenuation of the inhibition when the S-prenyl analogue was added to αt together with or after βγt. The inhibition does not involve interaction with the αt acyl group, since an S-prenyl analogue inhibited the [32P]ADP-ribosylation of an unlipidated αt mutant. These data suggest the existence of a hitherto unrecognized S-prenyl-binding site in αt, which is critical for its interaction with prenylated βγt.


Sign in / Sign up

Export Citation Format

Share Document