Inverse Effects of EDTA on αIIbβ3-Mediated Adhesion to Immobilized Fibrinogen Versus the D98 Plasmin Fragment of Fibrinogen

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1036-1036
Author(s):  
Hina Zafar ◽  
Jihong Li ◽  
George A David ◽  
Barry S. Coller

Abstract αIIbβ3 is known to mediate adhesion of platelets to immobilized fibrinogen through its interaction with the C-terminal γ chain dodecapeptide (γ12) and EDTA inhibits the adhesion by binding divalent cations required for ligand interaction with the β3 metal ion-dependent adhesion site (MIDAS) cation. Studies by several groups, however, suggest that αIIβb3 can also interact with other sites on fibrin(ogen). To identify potential additional sites of interaction between fibrinogen and αIIbβ3, we studied the adhesion of HEK293 cells expressing αIIbβ3 (αIIbβ3-HEK) to the D98 plasmin fragment of fibrinogen, which lacks the γ12 peptide. The D98 fragment did not contain the γ12 peptide as judged by both immunoblotting with mAb 7E9 (anti-γ12) and mass spectroscopy. αIIbβ3-HEK did not bind to immobilized D98 (10 µg/ml coating concentration) in the presence of 2 mM Ca2+/1 mM Mg2+, but they did bind to immobilized intact fibrinogen (10 µg/ml coating concentration) and the adhesion was inhibited by mAbs 10E5 (anti-αIIbβ3), 7E3 (anti-αIIbβ3 + αVβ3), and 7E9. Adhesion of αIIbβ3-HEK to fibrinogen was nearly eliminated by 10 mM EDTA [13,007 ± 3,676 vs 304 ± 331 arbitrary fluorescence intensity units (AFU); n=9; p<0.001]. Unexpectedly, and in dramatic contrast, 10 mM EDTA increased adhesion of αIIbβ3-HEK to D98 nearly 25-fold, from 458 ± 601 to 10,718 ± 3,106 AFU (n=9; p=0.001). The adhesion to D98 in the presence of EDTA was not inhibited by mAb 7E9 or mAb LM609 (anti-αVβ3), and was inhibited by mAb 7E3 by less than 10%. EDTA-dependent adhesion was, however, inhibited by mAb 10E5, which binds to the αIIb cap domain and inhibits fibrinogen binding to αIIbβ3, by 85% ± 4% (n=7; p=0.003). Dose-response experiments demonstrated that 3 mM EDTA was sufficient to block adhesion to fibrinogen and 3-4 mM EDTA was required to enhance adhesion to D98. Adding the β3 D119 mutation to αIIbβ3-HEK (αIIbβ3-D119-HEK), which disrupts the MIDAS, eliminated adhesion to fibrinogen (αIIbβ3-HEK: 17,342 ± 6,148 vs. αIIbβ3-D119-HEK: 0 ± 241 AFU; n=3; p=0.006), but had little or no effect on the binding to D98 in the presence of EDTA (αIIbβ3-HEK: 11,363 ± 3,700 vs. αIIbβ3-D119-HEK: 9,026 ± 3,252 AFU; n=3; p=0.054). However, unlike EDTA-dependent adhesion of αIIbβ3-HEK to D98, the adhesion of αIIbβ3-D119-HEK was inhibited by mAb 10E5 by only 20% ± 19% (n=3; p=0.247). We conclude that EDTA exposes one or more sites on αIIbβ3 that bind(s) to a site(s) on immobilized D98 that is either not accessible or not expressed on intact fibrinogen. These data are consistent with the known effect of EDTA in altering the conformation of αIIbβ3 as judged by its exposing "ligand-induced" binding sites recognized by mAbs such as AP5 and PMI-1, even in the absence of ligand, and the ability of αIIbβ3 to mediate interactions with fibrin to support clot retraction even in the presence of EDTA. EDTA-treated αIIbβ3 may, therefore, provide insights into potential ancillary sites of interaction between αIIbβ3 and fibrin(ogen). Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1035-1035
Author(s):  
Hina Zafar ◽  
Jihong Li ◽  
George A David ◽  
Barry S. Coller

Abstract The interaction of the fibrinogen γ chain dodecapeptide (HHLGGAKQAGDV; γ12) with the αIIbβ3 binding pocket is required for fibrinogen binding to αIIbβ3 and the binding of other αIIbβ3 ligands that contain RGD sequences such as von Willebrand factor and fibronectin. Since the strength of fibrinogen binding to αIIbβ3 increases over time, it is possible that there are additional sites of interaction between αIIbβ3 and fibrinogen, and several studies have proposed potential candidate ancillary sites on fibrinogen, including γ316-322 and γ370-381 (Podolnikova et al. JBC 289;2371,2014 and Remijn et al. Br J Haematol 117;650,2002). If such fibrinogen sites interact with αIIbβ3 at regions separate from the γ12 (RGD) binding site, these αIIbβ3 sites may be attractive targets for developing pure αIIbβ3 antagonists that are specific for fibrinogen. To identify potential additional interaction sites on fibrinogen and αIIbβ3, we studied adhesion of HEK293 cells expressing normal αIIbβ3 (αIIbβ3-HEK) to either fibrinogen or the D98 plasmin cleavage fragment of fibrinogen (which lacks γ12) in the presence of 2 mM Ca2+/1 mM Mg2+. The D98 fragment did not contain the γ12 peptide as judged by both immunoblotting with mAb 7E9 (anti-γ12) and mass spectroscopy. We also studied HEK293 cells expressing αIIbβ3 containing: 1. The double αIIb F992A + F993A mutations (αIIb-FF), which causes constitutive activation and ligand binding 2. Other activating mutations of αIIbβ3, including αIIb truncation at 991, β3 N339S, and β3 truncation at 717, 3. The β3 D119A mutation, which disrupts the β3 metal ion-dependent adhesion site (MIDAS) and eliminates adhesion of cells expressing normal αIIbβ3 to fibrinogen, or 4. Combined αIIb FF and β3 D119 mutations (FF+D119). αIIbβ3-HEK and αIIb-FF both bound to immobilized fibrinogen (10 µg/ml coating concentration). αIIbβ3-HEK did not adhere to immobilized D98 (10 µg/ml coating concentration), whereas αIIb-FF did adhere [αIIbβ3-HEK: 201 ± 295 vs. αIIb-FF: 8,221 ± 1,585 arbitrary fluorescence intensity units (AFU); n=7; p=0.003]. HEK cells expressing the other activating mutations also adhered to both fibrinogen and D98. HEK cells expressing the D119 mutation did not adhere to fibrinogen or D98 and adding the D119 mutation to the αIIb FF mutant led to loss of adhesion to both fibrinogen and D98. Adhesion of αIIb-FF to D98 was inhibited by mAb 10E5, which inhibits fibrinogen binding to αIIbβ3 and binds to the αIIb cap domain (89% ± 18%; n=7; p=0.003) and by mAb 7E3, which inhibits fibrinogen binding and binds to the β3 subunits of both αIIbβ3 and αVβ3 (95% ± 10%; n=7; p=0.006), but not by mAb 7E9 (28% ± 29%; n=7; p=0.299). Since cells expressing activated, but not unactivated αIIbβ3 were able to adhere to D98, our data are consistent with a model in which the initial interaction between αIIbβ3 and fibrinogen is mediated by γ12 binding to the αIIbβ3 ligand binding site followed by a conformational change that exposes additional site(s) on αIIbβ3 for another region or regions of fibrinogen. Refrences: Podolnikova NP, Yakovlev S, Yakubenko VP, Wang X, Gorkun OV, Ugarova TP. The interaction of integrin alphaIIbbeta3 with fibrin occurs through multiple binding sites in the alphaIIb beta-propeller domain. J Biol Chem. 2014;289:2371-2383. Remijn JA, Ijsseldijk MJ, van Hemel BM, Galanakis DK, Hogan KA, Lounes KC, Lord ST, Sixma JJ, de Groot PG. Reduced platelet adhesion in flowing blood to fibrinogen by alterations in segment gamma316-322, part of fibrin-specific region. Br J Haematol 2002;117:650-657. Disclosures No relevant conflicts of interest to declare.


1995 ◽  
Vol 305 (1) ◽  
pp. 151-158 ◽  
Author(s):  
P Spencer ◽  
P M Jordan

Experiments are described in which the individual properties of the two 5-aminolaevulinic acid (ALA) binding sites, the A-site and the P-site, of 5-aminolaevulinic acid dehydratase (ALAD) have been investigated. The ALA binding affinity at the A-site is greatly enhanced (at least 10-fold) on the binding of the catalytic metal ion (bound at the alpha-site). The nature of the catalytic metal ion, Mg2+ or Zn2+, also gave major variations in the substrate Km, P-site affinity for ALA, the effect of potassium and phosphate ions and the pH-dependence of substrate binding. Modification of the P-site by reaction of the enzyme-substrate Schiff base with NaBH4 and analysis of the reduced adduct by electro-spray mass spectrometry indicated a maximum of 1 mol of substrate incorporated/mol of subunit, correlating with a linear loss of enzyme activity. The reduced Schiff-base adduct was used to investigate substrate binding at the A-site by using rate-of-dialysis analysis. The affinity for ALA at the A-site of Mg alpha Zn beta ALAD was found to determine the Km for the reaction and was pH-dependent, with its affinity increasing from 1 mM at pH 6 to 70 microM at pH 8.5. The affinity of ALA at the P-site of Zn alpha An beta ALAD is proposed to limit the Km at pH values above 7, since the measured Kd for ALA at the A-site in 45 microM Tris, pH 8, was well below the observed Km (600 microM) under the same conditions. The amino group of the ALA molecule bound at the P-site was identified as a critical binding component for the A-site, explaining why ALA binding to ALAD is ordered, with the P-site ALA binding first. Structural requirements for ALA binding at the A- and P-sites have been identified: the P-site requires the carbonyl and carboxylate groups, whereas the A-site requires the amino, carbonyl and carboxylate groups of the substrate.


Blood ◽  
1996 ◽  
Vol 88 (10) ◽  
pp. 3824-3830 ◽  
Author(s):  
TH Mondoro ◽  
CD Wall ◽  
MM White ◽  
LK Jennings

Ligand-induced binding sites (LIBS) are neoantigenic regions of glycoprotein (GP)IIb-IIIa that are exposed upon interaction of the receptor with the ligand fibrinogen or the ligand recognition sequence (RGDS). LIBS have been suggested to contribute to postreceptor occupancy events such as full-scale platelet aggregation, adhesion to collagen, and clot retraction. This study examined the induction requirements of a GPIIIa LIBS with regard to ligand specificity. Through the use of the anti-LIBS D3, we report that this complex- activating antibody induces fibrinogen-and von Willebrand factor-binding to GPIIb-IIIa on intact platelets. Bound ligand was detected by flow cytometric analysis and platelet aggregation assays. These bound ligands increased the number of D3-binding sites and altered the affinity of D3 for GPIIb-IIIa on platelets. In contrast, activation of platelet GPIIb-IIIa by D3 did not increase the binding of another RGD- containing ligand, vitronectin. Furthermore, bound vitronectin on thrombin-stimulated platelets did not cause the expression of the D3 LIBS epitope. We conclude direct activation of GPIIb-IIIa in the absence of platelet activation results in selective ligand interaction and that D3 LIBS induction requires the binding of the multivalent ligands, fibrinogen or von Willebrand factor. Thus, the region of GPIIIa recognized by D3 may be an important regulatory domain in ligand- receptor interactions that directly mediate platelet aggregation.


1981 ◽  
Author(s):  
Elizabeth Kornecki ◽  
Stefan Niewiarowski

Fibrinogen and calcium are required for the aggregation of platelets stimulated by ADP or pre-treated with proteolytic enzymes. Specific platelet surface fibrinogen binding sites (receptors) are exposed after platelets are stimulated by ADP or pre-treated with Chymotrypsin or pronase. It has previously been shown in our laboratory that an intact, symmetrical fibrinogen molecule is essential for fibrinogen binding and fibrinogen-induced aggregation of both ADP-stimulated and proteolytically-treated platelets. Here we propose that the mechanism by which fibrinogen and calcium aggregate platelets is by forming inter-platelet bridges linking the fibrinogen receptors of adjacent platelets together. In support of this proposition are the following new lines of evidence: 1) The fibrinogen-induced aggregations of ADP-stfiliulated or proteolytically-treated platelets are inhibited by high concentrations of fibrinogen (Ki=2.6 and 8.5 × 10 5M, respectively). The fibrinogen binding sites on adjacent platelets, at these concentrations, would be saturated by fibrinogen and therefore no inter-platelet fibrinogen bridges could be formed to hold the platelets together. 2) ADP-stimulated or chymotrypsin-treated platelets aggregated by fibrinogen are deaggregated by Chymotrypsin or pronase and this deaggregation coincides with the loss of 125I-fibrinogen from the platelet surface. 3) Preincubation of platelets with EDTA results in inhibition of both platelet aggregation and 125I-fibrinogen binding. Following the aggregations of ADP-stimulated or of chymotrypsin-treated platelets by fibrinogen, the addition of EDTA to the platelet aggregates results in both their deaggregation and their loss of bound 125I-fibrinogen. Thus it appears that divalent cations, especially calcium, are essential for the formation of fibrinogen-linked platelet aggregates.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 994-994
Author(s):  
Akihiro Tamura ◽  
Hideyo Hirai ◽  
Asumi Yokota ◽  
Atsushi Sato ◽  
Tsukimi Shoji ◽  
...  

Abstract Currently, monocytes are classified into at least two subsets. Classical monocytes, also known as inflammatory monocytes (a Ly6C+ subset in mice and a CD14+ CD16− subset in human), are involved in innate immune responses. On the other hand, patrolling monocytes (a Ly6C− subset in mice and a CD14− CD16+ subset in human) have been recently identified. Ly6C− monocytes are found attached on the luminal side of endothelium and scavenge microparticles. Developmentally, Ly6C+ and Ly6C− monocytes share common monocyte progenitors (cMoPs), or Ly6C− monocytes might be converted from Ly6C+ monocytes. Although involvement of Ly6C− monocytes in various kinds of diseases has been reported, molecular mechanisms which regulate the homeostasis of Ly6C− monocytes are largely unknown. CCAAT/Enhancer Binding Protein β (C/EBPβ) is a leucine zipper type transcription factor. We and others have previously shown that C/EBPβ is required for stress-induced granulopoiesis (Hirai et al. Nat Immunol, 2006, Satake et al. J Immunol, 2012, Hayashi et al. Leukemia 2013). However, its roles in steady state hematopoiesis remain relatively unknown. We have recently found that peripheral blood monocytes are significantly reduced in Cebpb−/− mice (Tamura et al. Biochem Biophys Res Commun, 2015). In addition, last year in this meeting, we have reported that Cebpb mRNA is highly upregulated during differentiation from myeloid progenitors or Ly6C+ monocytes to Ly6C− monocytes, and that Ly6C− monocytes are almost completely absent in Cebpb−/− mice due to enhanced cell death [Abstract #224]. Here, we further investigated the molecular mechanisms underlying C/EBPβ-dependent survival of Ly6C− monocytes. In this study, we focused on the regulation of Csf1r (also known as M-CSF receptor). Csf1r is an essential molecule for the development and survival of monocytes. To determine the developmental stages at which Csf1r plays critical roles, we measured the expressions of Csf1r mRNA in hematopoietic stem/progenitor cells and monocyte subsets obtained from wild-type (WT) mice. Csf1r mRNA was expressed at at low levels in hematopoietic stem/progenitors including macrophage dendritic precursors (MDPs) and cMoPs. Csf1r mRNA started to be upregulated in Ly6C+ monocytes, followed by a drastic increase in Ly6C− monocytes. These expression patterns were quite similar to those of Cebpb, suggesting the close relationship between Csf1r and C/EBPβ. Interestingly, such drastic increase of Csf1r mRNA in Ly6C− monocytes was blunted in Cebpb−/− mice, and protein levels of Csf1r in Cebpb−/− Ly6C− monocytes were significantly lower than those in WT Ly6C− monocytes. In order to evaluate the effect of C/EBPβ overexpression on Csf1r expression, EML cells, a mouse hematopoietic stem cell line, were engineered to express C/EBPβ-estrogen receptor (ER) fusion protein or ER alone. Nuclear translocation of C/EBPβ-ER in the presence of tamoxifen resulted in significantly increased levels of Csf1r mRNA and protein when compared to nuclear translocation of ER alone. Previous reports have demonstrated that a combination of a promoter sequence and an enhancer region located in the first intron of Csf1r gene (Fms intronic regulatory element: FIRE) is enough to recapitulate the endogenous Csf1r expression and that these elements contained consensus binding sites for C/EBP transcription factors. Then, we hypothesized that C/EBPβ binds to these sites, activates transcription of Csf1r gene and promotes survival of Ly6C- monocytes. To evaluate this hypothesis, we utilized an expression vector, in which green fluorescent protein (GFP) is driven by a combination of the Csf1r promoter and FIRE sequences (Csf1r-EGFP-FIRE) (a kind gift from Drs Clare P and David A Hume, University of Edinburgh). When a C/EBPβexpression vector was co-transfected with the vector containing Csf1r-EGFP-FIRE into HEK293 cells, the frequencies of GFP positive cells were significantly higher when compared to a control vector (C/EBPβ vs control; 4.6±0.6 vs 1.6±1.0, p=0.01), suggesting that C/EBPβ regulates Csf1r expression through these elements. We are currently evaluating the significance of C/EBP consensus binding sites in the promoter and the enhancer. ChIP PCR is also in progress to further verify our hypothesis. Collectively, these results suggest that Csf1r is a critical downstream target of C/EBPβ in Ly6C- monocytes. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 260-260
Author(s):  
Tetsuji Kamata ◽  
Makoto Handa ◽  
Yohko Kawai ◽  
Yasuo Ikeda ◽  
Sadakazu Aiso

Abstract Abstract 260 Platelet αIIbβ3 integrin undergoes structural rearrangements to increase the affinity for ligands. Two major changes in integrin three-dimensional structure are critical for this activation process. One is the straightening of integrin legs (extension), and the other is the swing out movement of the β3 hybrid domain (swing-out). In this study, we have found that this structural requirement for activation differs depending upon the divalent cations present. We previously reported that the swing-out-defective mutant αIIbD319C/β3V359C did not bind fibrinogen in the presence of Ca2+/Mg2+. However, the same mutant bound fibrinogen in the presence of Mn2+, when activated by monoclonal antibody PT25-2 or by forcing the integrin legs to extend by concomitant αIIbQ595NTT mutation. We engineered another mutation that was designed specifically to prevent the swing-out. Likewise, this β3G327C/β3V419C mutation blocked fibrinogen binding in the presence of Ca2+/Mg2+, but not in the presence of Mn2+. On the other hand, when integrin extension was specifically prevented by αIIbD464C/αIIbS728C mutation, fibrinogen binding was observed neither in the presence of Ca2+/Mg2+ nor in Mn2+. Thus, swing-out is not essential for activation in Mn2+, as long as integrin legs are in extended state, while both swing-out and extension are required in Ca2+/Mg2+. To explore the underlying mechanism originating this difference, three distinct cation-binding sites in the βA domain were mutated. Among the three sites, MIDAS is occupied by Mg2+, while ADMIDAS and LIMBS/SyMBS are occupied by Ca2+ in Ca2+/Mg2+ condition. When amino acid residues composing these sites were mutated to Ala, none of them bound fibrinogen, except for D126A and D127A in the presence of Ca2+/Mg2+. Notably, these mutations restored fibrinogen binding in the swing-out-defective mutant. Amino acid residues D126 and D127 compose ADMIDAS together with S123 and M335. Swing-out of the hybrid domain disrupts M335 from ADMIDAS. This allows Ca2+ to move toward MIDAS, bringing the β1-α1 loop closer to ligand to provide contact site. However, similar rearrangement of the cation/ligand binding sites has been shown to take place in Mn2+ without the swing-out. The results suggest partial disruption of ADMIDAS is the key event for activation in Ca2+/Mg2+, while it is not so in Mn2+. Our results may reconcile the dispute over the apparently contradicting findings in the crystal structure that integrin with a closed head with Mn2+ represents an active conformation as one with an open head with Ca2+/Mg2+. Disclosures: Kawai: Daiichi Sankyo: Consultancy; Bayer: Consultancy; Toyama Chemical: Consultancy.


1979 ◽  
Author(s):  
E.I. Peerschke ◽  
R.A. Grant ◽  
M.B. Zucker

Since calcium and fibrinogen are essential cofactors for ADP-induced aggregation, their mechanisms of action were investigated. Aspirin-treated platelets were filtered through Sepharose 2B equilibrated with cation-poor Tyrode’s solution. After adding the radioactive compounds at 22, platelets were centrifuged through silicone oil. Trapping was assessed in separate samples with 14C-sorbitoi. Calcium binding was maximal at 1 hr and with 200 uH CaCl2. Two binding sites could be demonstrated on normal and thrombasthenic platelets and on platelets which had lost their ability to aggregate (but not to change shape or promote clot retraction) after treatment with EDTA (8 min, 37°, pH 7.8). ADP did not alter calcium binding in the presence or absence of fibrinogen. Fibrinogen, however, bound to normal gel filtered platelets in the presence of ADP and ionized calcium “ but not to thrombasthenic or EDTA-treated platelets or to normal platelets in the presence of EDTA or at pH 6.5. Binding of fibrinogen increased with concentration but saturation was not observed even at physiologic levels. Fibrinogen binding was similar in gel filtered platelets and citrated piatelet-rich plasma. These studies indicate that stimulation of platelets with ADP under conditions favorable to aggregation is associated with binding of fibrinogen but not of additional calcium.


Blood ◽  
2000 ◽  
Vol 95 (11) ◽  
pp. 3380-3386 ◽  
Author(s):  
Wei Zhang ◽  
Robert W. Colman

Abstract The activity of phosphodiesterase (PDE)3A requires divalent cations. Putative metal-binding sites are expected at 2 highly conserved metal-binding motifs, HXXXH(X)25E. A functional truncated recombinant PDE3A containing the catalytic domain (PDE3A▵1) and mutant proteins were expressed in a baculovirus/Sf9 cell system. All the mutant proteins had decreased catalytic efficiency (kcat/Km). Mutants H752A, H756A, and E825A had kcat of less than 0.0008 s−1 to 0.0475 s−1 compared to PDE3A▵1, with 1.86 second−1, with unchanged Km. Although E866A had a kcat of 0.235 s−1, the Kmfor cyclic adenosine monophosphate (cAMP) was increased 11-fold and the Ki for cyclic guanosine monophosphate (cGMP) was 27-fold higher than PDE3A▵1. The Ki of H836A for cGMP was 177-fold higher than that of PDE3A▵1. The Km for E971A was 5-fold higher than PDE3A▵1. These results suggest that the cAMP and cGMP binding sites are overlapping, but not identical, involving both common and different amino acids. Mutants E825A, H836A, and E866A showed low activity in a metal ion-free assay; however, their enzymatic activities were increased 4- to 10-fold in buffers containing Mn2+, Mg2+, or Co2+. This observation indicates that conserved amino acids in the second metal-binding motif might not be involved in binding divalent cations but may serve other functions such as substrate or inhibitor binding in PDE3A.


2005 ◽  
Vol 126 (2) ◽  
pp. 137-150 ◽  
Author(s):  
Jianmin Jiang ◽  
Mingjiang Li ◽  
Lixia Yue

TRPM7 is unique in being both an ion channel and a protein kinase. It conducts a large outward current at +100 mV but a small inward current at voltages ranging from −100 to −40 mV under physiological ionic conditions. Here we show that the small inward current of TRPM7 was dramatically enhanced by a decrease in extracellular pH, with an ∼10-fold increase at pH 4.0 and 1–2-fold increase at pH 6.0. Several lines of evidence suggest that protons enhance TRPM7 inward currents by competing with Ca2+ and Mg2+ for binding sites, thereby releasing blockade of divalent cations on inward monovalent currents. First, extracellular protons significantly increased monovalent cation permeability. Second, higher proton concentrations were required to induce 50% of maximal increase in TRPM7 currents when the external Ca2+ and Mg2+ concentrations were increased. Third, the apparent affinity for Ca2+ and Mg2+ was significantly diminished at elevated external H+ concentrations. Fourth, the anomalous-mole fraction behavior of H+ permeation further suggests that protons compete with divalent cations for binding sites in the TRPM7 pore. Taken together, it appears that at physiological pH (7.4), Ca2+ and Mg2+ bind to TRPM7 and inhibit the monovalent cationic currents; whereas at high H+ concentrations, the affinity of TRPM7 for Ca2+ and Mg2+ is decreased, thereby allowing monovalent cations to pass through TRPM7. Furthermore, we showed that the endogenous TRPM7-like current, which is known as Mg2+-inhibitable cation current (MIC) or Mg nucleotide–regulated metal ion current (MagNuM) in rat basophilic leukemia (RBL) cells was also significantly potentiated by acidic pH, suggesting that MIC/MagNuM is encoded by TRPM7. The pH sensitivity represents a novel feature of TRPM7 and implies that TRPM7 may play a role under acidic pathological conditions.


Sign in / Sign up

Export Citation Format

Share Document