Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Mol Cell Cardiol ; 186: 125-137, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38008210

RESUMEN

N-terminal cardiac myosin-binding protein C (cMyBP-C) domains (C0-C2) bind to thick (myosin) and thin (actin) filaments to coordinate contraction and relaxation of the heart. These interactions are regulated by phosphorylation of the M-domain situated between domains C1 and C2. In cardiomyopathies and heart failure, phosphorylation of cMyBP-C is significantly altered. We aimed to investigate how cMyBP-C interacts with myosin and actin. We developed complementary, high-throughput, C0-C2 FRET-based binding assays for myosin and actin to characterize the effects due to 5 HCM-linked variants or functional mutations in unphosphorylated and phosphorylated C0-C2. The assays indicated that phosphorylation decreases binding to both myosin and actin, whereas the HCM mutations in M-domain generally increase binding. The effects of mutations were greatest in phosphorylated C0-C2, and some mutations had a larger effect on actin than myosin binding. Phosphorylation also altered the spatial relationship of the probes on C0-C2 and actin. The magnitude of these structural changes was dependent on C0-C2 probe location (C0, C1, or M-domain). We conclude that binding can differ between myosin and actin due to phosphorylation or mutations. Additionally, these variables can change the mode of binding, affecting which of the interactions in cMyBP-C N-terminal domains with myosin or actin take place. The opposite effects of phosphorylation and M-domain mutations is consistent with the idea that cMyBP-C phosphorylation is critical for normal cardiac function. The precision of these assays is indicative of their usefulness in high-throughput screening of drug libraries for targeting cMyBP-C as therapy.


Asunto(s)
Citoesqueleto de Actina , Actinas , Proteínas Portadoras , Actinas/metabolismo , Fosforilación , Citoesqueleto de Actina/metabolismo , Miosinas/genética , Miosinas/metabolismo , Mutación
2.
J Biol Chem ; 299(12): 105369, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37865311

RESUMEN

Cardiac MyBP-C (cMyBP-C) interacts with actin and myosin to fine-tune cardiac muscle contractility. Phosphorylation of cMyBP-C, which reduces the binding of cMyBP-C to actin and myosin, is often decreased in patients with heart failure (HF) and is cardioprotective in model systems of HF. Therefore, cMyBP-C is a potential target for HF drugs that mimic its phosphorylation and/or perturb its interactions with actin or myosin. We labeled actin with fluorescein-5-maleimide (FMAL) and the C0-C2 fragment of cMyBP-C (cC0-C2) with tetramethylrhodamine (TMR). We performed two complementary high-throughput screens (HTS) on an FDA-approved drug library, to discover small molecules that specifically bind to cMyBP-C and affect its interactions with actin or myosin, using fluorescence lifetime (FLT) detection. We first excited FMAL and detected its FLT, to measure changes in fluorescence resonance energy transfer (FRET) from FMAL (donor) to TMR (acceptor), indicating binding. Using the same samples, we then excited TMR directly, using a longer wavelength laser, to detect the effects of compounds on the environmentally sensitive FLT of TMR, to identify compounds that bind directly to cC0-C2. Secondary assays, performed on selected modulators with the most promising effects in the primary HTS assays, characterized the specificity of these compounds for phosphorylated versus unphosphorylated cC0-C2 and for cC0-C2 versus C1-C2 of fast skeletal muscle (fC1-C2). A subset of identified compounds modulated ATPase activity in cardiac and/or skeletal myofibrils. These assays establish the feasibility of the discovery of small-molecule modulators of the cMyBP-C-actin/myosin interaction, with the ultimate goal of developing therapies for HF.


Asunto(s)
Proteínas Portadoras , Descubrimiento de Drogas , Insuficiencia Cardíaca , Miofibrillas , Bibliotecas de Moléculas Pequeñas , Humanos , Actinas/metabolismo , Descubrimiento de Drogas/métodos , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Miosinas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Evaluación Preclínica de Medicamentos , Miofibrillas/efectos de los fármacos , Proteínas Portadoras/metabolismo , Técnicas Biosensibles , Adenosina Trifosfatasas/metabolismo , Músculo Esquelético/metabolismo , Proteínas Recombinantes/metabolismo , Activación Enzimática/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia
3.
bioRxiv ; 2023 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-37066417

RESUMEN

Cardiac MyBP-C (cMyBP-C) interacts with actin-myosin to fine-tune cardiac muscle contractility. Phosphorylation of cMyBP-C, which reduces binding of cMyBP-C to actin or myosin, is often decreased in heart failure (HF) patients, and is cardioprotective in model systems for HF. Therefore, cMyBP-C is a potential target for HF drugs that mimic phosphorylation and/or perturb its interactions with actin or myosin. We labeled actin with fluorescein-5-maleimide (FMAL), and the C0-C2 fragment of cMyBP-C (cC0-C2) with tetramethyl rhodamine (TMR). We performed two complementary high-throughput screens (HTS) on an FDA-approved drug library, to discover small molecules that specifically bind to cMyBP-C and affect its interactions with actin or myosin, using fluorescence lifetime (FLT) detection. We first excited FMAL and detected its FLT, to measure changes in fluorescence resonance energy transfer (FRET) from FMAL (donor) to TMR (acceptor), indicating binding and/or structural changes in the protein complex. Using the same samples, we then excited TMR directly, using a longer wavelength laser, to detect the effects of compounds on the environmentally sensitive FLT of TMR, to identify compounds that bind directly to cC0-C2. Secondary assays, performed on selected modulators with the most promising effects in the primary HTS assays, characterized specificity of these compounds for phosphorylated versus unphosphorylated cC0-C2 and for cC0-C2 versus C1-C2 of fast skeletal muscle (fskC1-C2). A subset of identified compounds modulated ATPase activity in cardiac and/or skeletal myofibrils. These assays establish feasibility for discovery of small-molecule modulators of the cMyBP-C-actin/myosin interaction, with the ultimate goal of developing therapies for HF.

4.
J Gen Physiol ; 155(3)2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36633587

RESUMEN

Cardiac muscle contraction is regulated by Ca2+-induced structural changes of the thin filaments to permit myosin cross-bridge cycling driven by ATP hydrolysis in the sarcomere. In congestive heart failure, contraction is weakened, and thus targeting the contractile proteins of the sarcomere is a promising approach to therapy. However, development of novel therapeutic interventions has been challenging due to a lack of precise discovery tools. We have developed a fluorescence lifetime-based assay using an existing site-directed probe, N,N'-dimethyl-N-(iodoacetyl)-N'-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)ethylenediamine (IANBD) attached to human cardiac troponin C (cTnC) mutant cTnCT53C, exchanged into porcine cardiac myofibrils. We hypothesized that IANBD-cTnCT53C fluorescence lifetime measurements provide insight into the activation state of the thin filament. The sensitivity and precision of detecting structural changes in cTnC due to physiological and therapeutic modulators of thick and thin filament functions were determined. The effects of Ca2+ binding to cTnC and myosin binding to the thin filament were readily detected by this assay in mock high-throughput screen tests using a fluorescence lifetime plate reader. We then evaluated known effectors of altered cTnC-Ca2+ binding, W7 and pimobendan, and myosin-binding drugs, mavacamten and omecamtiv mecarbil, used to treat cardiac diseases. Screening assays were determined to be of high quality as indicated by the Z' factor. We conclude that cTnC lifetime-based probes allow for precise evaluation of the thin filament activation in functioning myofibrils that can be used in future high-throughput screens of small-molecule modulators of function of the thin and thick filaments.


Asunto(s)
Calcio , Troponina C , Humanos , Animales , Porcinos , Calcio/metabolismo , Fluorescencia , Troponina C/metabolismo , Miocardio/metabolismo , Contracción Miocárdica/fisiología
5.
J Mol Cell Cardiol ; 166: 116-126, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35227736

RESUMEN

Cardiac myosin-binding protein C (cMyBP-C) is a thick filament-associated protein of the sarcomere and a potential therapeutic target for treating contractile dysfunction in heart failure. Mimicking the structural dynamics of phosphorylated cMyBP-C by small-molecule drug binding could lead to therapies that modulate cMyBP-C conformational states, and thereby function, to improve contractility. We have developed a human cMyBP-C biosensor capable of detecting intramolecular structural changes due to phosphorylation and mutation. Using site-directed mutagenesis and time-resolved fluorescence resonance energy transfer (TR-FRET), we substituted cysteines in cMyBP-C N-terminal domains C0 through C2 (C0-C2) for thiol-reactive fluorescent probe labeling to examine C0-C2 structure. We identified a cysteine pair that upon donor-acceptor labeling reports phosphorylation-sensitive structural changes between the C1 domain and the tri-helix bundle of the M-domain that links C1 to C2. Phosphorylation reduced FRET efficiency by ~18%, corresponding to a ~11% increase in the distance between probes and a ~30% increase in disorder between them. The magnitude and precision of phosphorylation-mediated TR-FRET changes, as quantified by the Z'-factor, demonstrate the assay's potential for structure-based high-throughput screening of compounds for cMyBP-C-targeted therapies to improve cardiac performance in heart failure. Additionally, by probing C1's spatial positioning relative to the tri-helix bundle, these findings provide new molecular insight into the structural dynamics of phosphoregulation as well as mutations in cMyBP-C. Biosensor sensitivity to disease-relevant mutations in C0-C2 was demonstrated by examination of the hypertrophic cardiomyopathy mutation R282W. The results presented here support a screening platform to identify small molecules that regulate N-terminal cMyBP-C conformational states.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Insuficiencia Cardíaca , Proteínas Portadoras , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Humanos , Mutación , Fosforilación
6.
J Biol Chem ; 297(1): 100840, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34052227

RESUMEN

Cardiac myosin-binding protein C (cMyBP-C) interacts with actin and myosin to modulate cardiac muscle contractility. These interactions are disfavored by cMyBP-C phosphorylation. Heart failure patients often display decreased cMyBP-C phosphorylation, and phosphorylation in model systems has been shown to be cardioprotective against heart failure. Therefore, cMyBP-C is a potential target for heart failure drugs that mimic phosphorylation or perturb its interactions with actin/myosin. Here we have used a novel fluorescence lifetime-based assay to identify small-molecule inhibitors of actin-cMyBP-C binding. Actin was labeled with a fluorescent dye (Alexa Fluor 568, AF568) near its cMyBP-C binding sites; when combined with the cMyBP-C N-terminal fragment, C0-C2, the fluorescence lifetime of AF568-actin decreases. Using this reduction in lifetime as a readout of actin binding, a high-throughput screen of a 1280-compound library identified three reproducible hit compounds (suramin, NF023, and aurintricarboxylic acid) that reduced C0-C2 binding to actin in the micromolar range. Binding of phosphorylated C0-C2 was also blocked by these compounds. That they specifically block binding was confirmed by an actin-C0-C2 time-resolved FRET (TR-FRET) binding assay. Isothermal titration calorimetry (ITC) and transient phosphorescence anisotropy (TPA) confirmed that these compounds bind to cMyBP-C, but not to actin. TPA results were also consistent with these compounds inhibiting C0-C2 binding to actin. We conclude that the actin-cMyBP-C fluorescence lifetime assay permits detection of pharmacologically active compounds that affect cMyBP-C-actin binding. We now have, for the first time, a validated high-throughput screen focused on cMyBP-C, a regulator of cardiac muscle contractility and known key factor in heart failure.


Asunto(s)
Actinas/metabolismo , Proteínas Portadoras/metabolismo , Ensayos Analíticos de Alto Rendimiento , Miocardio/metabolismo , Actinas/química , Animales , Técnicas Biosensibles , Calorimetría , Fluorescencia , Transferencia Resonante de Energía de Fluorescencia , Humanos , Unión Proteica , Conejos , Sarcómeros/metabolismo , Factores de Tiempo
7.
J Gen Physiol ; 153(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33600558

RESUMEN

Binding properties of actin-binding proteins are typically evaluated by cosedimentation assays. However, this method is time-consuming, involves multiple steps, and has a limited throughput. These shortcomings preclude its use in screening for drugs that modulate actin-binding proteins relevant to human disease. To develop a simple, quantitative, and scalable F-actin-binding assay, we attached fluorescent probes to actin's Cys-374 and assessed changes in fluorescence lifetime upon binding to the N-terminal region (domains C0-C2) of human cardiac myosin-binding protein C (cMyBP-C). The lifetime of all five probes tested decreased upon incubation with cMyBP-C C0-C2, as measured by time-resolved fluorescence (TR-F), with IAEDANS being the most sensitive probe that yielded the smallest errors. The TR-F assay was compared with cosedimentation to evaluate in vitro changes in binding to actin and actin-tropomyosin arising from cMyBP-C mutations associated with hypertrophic cardiomyopathy (HCM) and tropomyosin binding. Lifetime changes of labeled actin with added C0-C2 were consistent with cosedimentation results. The HCM mutation L352P was confirmed to enhance actin binding, whereas PKA phosphorylation reduced binding. The HCM mutation R282W, predicted to disrupt a PKA recognition sequence, led to deficits in C0-C2 phosphorylation and altered binding. Lastly, C0-C2 binding was found to be enhanced by tropomyosin and binding capacity to be altered by mutations in a tropomyosin-binding region. These findings suggest that the TR-F assay is suitable for rapidly and accurately determining quantitative binding and for screening physiological conditions and compounds that affect cMyBP-C binding to F-actin for therapeutic discovery.


Asunto(s)
Citoesqueleto de Actina , Actinas , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Humanos , Unión Proteica
8.
J Biol Chem ; 294(44): 16228-16240, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31519753

RESUMEN

Cardiac myosin-binding protein C (cMyBP-C) is a thick filament-associated protein that influences actin-myosin interactions. cMyBP-C alters myofilament structure and contractile properties in a protein kinase A (PKA) phosphorylation-dependent manner. To determine the effects of cMyBP-C and its phosphorylation on the microsecond rotational dynamics of actin filaments, we attached a phosphorescent probe to F-actin at Cys-374 and performed transient phosphorescence anisotropy (TPA) experiments. Binding of cMyBP-C N-terminal domains (C0-C2) to labeled F-actin reduced rotational flexibility by 20-25°, indicated by increased final anisotropy of the TPA decay. The effects of C0-C2 on actin TPA were highly cooperative (n = ∼8), suggesting that the cMyBP-C N terminus impacts the rotational dynamics of actin spanning seven monomers (i.e. the length of tropomyosin). PKA-mediated phosphorylation of C0-C2 eliminated the cooperative effects on actin flexibility and modestly increased actin rotational rates. Effects of Ser to Asp phosphomimetic substitutions in the M-domain of C0-C2 on actin dynamics only partially recapitulated the phosphorylation effects. C0-C1 (lacking M-domain/C2) similarly exhibited reduced cooperativity, but not as reduced as by phosphorylated C0-C2. These results suggest an important regulatory role of the M-domain in cMyBP-C effects on actin structural dynamics. In contrast, phosphomimetic substitution of the glycogen synthase kinase (GSK3ß) site in the Pro/Ala-rich linker of C0-C2 did not significantly affect the TPA results. We conclude that cMyBP-C binding and PKA-mediated phosphorylation can modulate actin dynamics. We propose that these N-terminal cMyBP-C-induced changes in actin dynamics help explain the functional effects of cMyBP-C phosphorylation on actin-myosin interactions.


Asunto(s)
Actinas/metabolismo , Proteínas Portadoras/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Miosinas Cardíacas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Polarización de Fluorescencia/métodos , Humanos , Mediciones Luminiscentes/métodos , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Miofibrillas/metabolismo , Miosinas/metabolismo , Fosforilación , Unión Proteica/fisiología , Conejos , Rotación , Sarcómeros/metabolismo
9.
J Mol Cell Cardiol ; 125: 140-148, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30359561

RESUMEN

RATIONALE: Mutations in the gene encoding the sarcomeric protein cardiac myosin-binding protein C (cMyBP-C) are a leading cause of hypertrophic cardiomyopathy (HCM). Mouse models targeting cMyBP-C and use of recombinant proteins have been effective in studying its roles in contractile function and disease. Surprisingly, while the N-terminus of cMyBP-C is important to regulate myofilament binding and contains many HCM mutations, an incorrect sequence, lacking the N-terminal 8 amino acids has been used in many studies. OBJECTIVES: To determine the N-terminal cMyBP-C sequences in ventricles and investigate the roles of species-specific differences in cMyBP-C on myofilament binding. METHODS AND RESULTS: We determined cMyBP-C sequences in mouse and human by inspecting available sequence databases. N-terminal differences were confirmed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Cosedimentation assays with actin or myosin were used to examine binding in mouse, human and chimeric fusion proteins of cMyBP-C. Time-resolved FRET (TR-FRET) with site-directed probes on cMyBP-C was employed to measure structural dynamics. LC-MS/MS supported the sequencing data that mouse cMyBP-C contains an eight-residue N-terminal extension (NTE) not found in human. Cosedimentation assays revealed that cardiac myosin binding was strongly influenced by the presence of the NTE, which reduced binding by 60%. 75% more human C0-C2 than mouse bound to myosin. Actin binding of mouse C0-C2 was not affected by the NTE. 50% more human C0-C2 than mouse bound to actin. TR-FRET indicates that the NTE did not significantly affect structural dynamics across domains C0 and C1. CONCLUSIONS: Our functional results are consistent with the idea that cardiac myosin binding of N-terminal cMyBP-C is reduced in the mouse protein due to the presence of the NTE, which is proposed to interfere with myosin regulatory light chain (RLC) binding. The NTE is a critical component of mouse cMyBP-C, and should be considered in extrapolation of studies to cMyBP-C and HCM mechanisms in human.


Asunto(s)
Proteínas Portadoras/metabolismo , Miofibrillas/metabolismo , Actinas/metabolismo , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Cromatografía Liquida , Humanos , Ratones , Miosinas/metabolismo , Unión Proteica , Proteínas Recombinantes/metabolismo , Espectrometría de Masas en Tándem
10.
Phys Biol ; 13(6): 066005, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27848929

RESUMEN

Cysteine residues (Cys) in the membrane proximal region are common post-translational modification (PTM) sites in transmembrane proteins. Herein, the effects of a highly conserved membrane proximal α-subunit Cys1368 on the diffusion properties of αPS2CßPS integrins are reported. Sequence alignment shows that this cysteine is palmitoylated in human α3 and α6 integrin subunits. Replacing Cys1368 in wild-type integrins with valine (Val1368) putatively blocks a PTM site and alters integrins' ligand binding and diffusion characteristics. Both fluorescence recovery after photobleaching (FRAP) and single particle tracking (SPT) diffusion measurements show Val1368 integrins are more mobile compared to wild-type integrins. Approximately 33% and 8% more Val1368 integrins are mobile as measured by FRAP and SPT, respectively. The mobile Val1368 integrins also exhibit less time-dependent diffusion, as measured by FRAP. Tandem mass spectrometry data suggest that Cys1368 contains a redox or palmitoylation PTM in αPS2CßPS integrins. This membrane proximal Cys may play an important role in the diffusion of other alpha subunits that contain this conserved residue.


Asunto(s)
Cisteína/metabolismo , Cadenas alfa de Integrinas/química , Cadenas alfa de Integrinas/genética , Sustitución de Aminoácidos , Animales , Proteínas Bacterianas/genética , Bioquímica/métodos , Cisteína/química , Difusión , Drosophila/genética , Proteínas de Drosophila/química , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Cadenas alfa de Integrinas/metabolismo , Proteínas Luminiscentes/genética , Modificación Traduccional de las Proteínas , Valina/química , Valina/metabolismo
11.
Clin Cancer Res ; 18(9): 2429-35, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22415315

RESUMEN

Abnormal Hedgehog (Hh) pathway activity has been reported in many cancers, including basal cell carcinomas, medulloblastomas, rhabdomyosarcomas, glioblastomas, and breast and prostate cancers. For this reason, the Hh pathway is a flourishing area for development of anticancer drugs such as Hh ligand antagonists (e.g., 5E1 and robotnikinin), Smo inhibitors (e.g., GDC-0449 and IPI-926), and Gli transcriptional activity inhibitors (e.g., GANT58 and GANT61). It is now clear that primary cilia are required for activation of the Hh pathway in normal vertebrate cells. It is in the primary cilium that both positive and negative effectors of the Hh pathway are processed by posttranslational modifications. In many cancers, preliminary results suggest that primary cilia are lost. As drugs that inhibit different steps of the Hh pathway are developed, it will be important to consider how these drugs will function in the context of primary cilia in the tumor environment. Here, we discuss why some of the Hh inhibitors may be ineffective if primary cilia are lost on cancer cells. Understanding the relationships between clinical inhibitors of the Hh pathway and the presence or absence of primary cilia may turn out to be critical for targeting these therapeutics to the correct population of patients and improving their efficacy. Further work is needed in this area to maximize the potential of these exciting therapeutic targets.


Asunto(s)
Cilios/fisiología , Proteínas Hedgehog/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal , Animales , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Procesamiento Proteico-Postraduccional/efectos de los fármacos
12.
J Biol Chem ; 286(35): 30981-30993, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21757698

RESUMEN

We examined over 50 mutations in the Drosophila ßPS integrin subunit that alter integrin function in situ for their ability to bind a soluble monovalent ligand, TWOW-1. Surprisingly, very few of the mutations, which were selected for conditional lethality in the fly, reduce the ligand binding ability of the integrin. The most prevalent class of mutations activates the integrin heterodimer. These findings emphasize the importance of integrin affinity regulation and point out how molecular interactions throughout the integrin molecule are important in keeping the integrin in a low affinity state. Mutations strongly support the controversial deadbolt hypothesis, where the CD loop in the ß tail domain acts to restrain the I domain in the inactive, bent conformation. Site-directed mutations in the cytoplasmic domains of ßPS and αPS2C reveal different effects on ligand binding from those observed for αIIbß3 integrins and identify for the first time a cytoplasmic cysteine residue, conserved in three human integrins, as being important in affinity regulation. In the fly, we find that genetic interactions of the ßPS mutations with reduction in talin function are consistent with the integrin affinity differences measured in cells. Additionally, these genetic interactions report on increased and decreased integrin functions that do not result in affinity changes in the PS2C integrin measured in cultured cells.


Asunto(s)
Proteínas de Drosophila/genética , Matriz Extracelular/metabolismo , Cadenas alfa de Integrinas/genética , Cadenas beta de Integrinas/genética , Mutación , Alelos , Animales , Adhesión Celular , Drosophila , Humanos , Enlace de Hidrógeno , Ligandos , Mutagénesis Sitio-Dirigida , Unión Proteica , Proteínas/química , Talina/metabolismo
13.
Dev Biol ; 340(2): 504-17, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20152825

RESUMEN

Morphogenesis of the adult structures of holometabolous insects is regulated by ecdysteroids and juvenile hormones and involves cell-cell interactions mediated in part by the cell surface integrin receptors and their extracellular matrix (ECM) ligands. These adhesion molecules and their regulation by hormones are not well characterized. We describe the gene structure of a newly described ECM molecule, tenectin, and demonstrate that it is a hormonally regulated ECM protein required for proper morphogenesis of the adult wing and male genitalia. Tenectin's function as a new ligand of the PS2 integrins is demonstrated by both genetic interactions in the fly and by cell spreading and cell adhesion assays in cultured cells. Its interaction with the PS2 integrins is dependent on RGD and RGD-like motifs. Tenectin's function in looping morphogenesis in the development of the male genitalia led to experiments that demonstrate a role for PS integrins in the execution of left-right asymmetry.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Genitales Masculinos/fisiología , Alas de Animales/fisiología , Animales , Drosophila/genética , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/genética , Proteínas de la Matriz Extracelular/genética , Inmunohistoquímica , Hibridación in Situ , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/metabolismo , Larva/genética , Larva/metabolismo , Ligandos , Masculino , Morfogénesis/genética , Mutación , Transgenes , Alas de Animales/crecimiento & desarrollo , Alas de Animales/metabolismo
14.
J Biol Chem ; 285(3): 1841-9, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19917607

RESUMEN

Integrin alphaIIbbeta3 affinity regulation by talin binding to the cytoplasmic tail of beta3 is a generally accepted model for explaining activation of this integrin in Chinese hamster ovary cells and human platelets. Most of the evidence for this model comes from the use of multivalent ligands. This raises the possibility that the activation being measured is that of increased clustering of the integrin rather than affinity. Using a newly developed assay that probes integrins on the surface of cells with only monovalent ligands prior to fixation, I do not find increases in affinity of alphaIIbbeta3 integrins by talin head fragments in Chinese hamster ovary cells, nor do I observe affinity increases in human platelets stimulated with thrombin. Binding to a multivalent ligand does increase in both of these cases. This assay does report affinity increases induced by either Mn(2+), a cytoplasmic domain mutant (D723R) in the cytoplasmic domain of beta3, or preincubation with a peptide ligand. These results reconcile the previously observed differences between talin effects on integrin activation in Drosophila and vertebrate systems and suggest new models for talin regulation of integrin activity in human platelets.


Asunto(s)
Plaquetas/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Activación Enzimática , Humanos , Inmunoglobulina M/metabolismo , Ligandos , Ratones , Estructura Terciaria de Proteína , Talina/química , Talina/metabolismo , Trombina/metabolismo
15.
Mol Biol Cell ; 19(8): 3589-98, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18508915

RESUMEN

Integrin-mediated cell adhesion is essential for development of multicellular organisms. In worms, flies, and vertebrates, talin forms a physical link between integrin cytoplasmic domains and the actin cytoskeleton. Loss of either integrins or talin leads to similar phenotypes. In vertebrates, talin is also a key regulator of integrin affinity. We used a ligand-mimetic Fab fragment, TWOW-1, to assess talin's role in regulating Drosophila alphaPS2 betaPS affinity. Depletion of cellular metabolic energy reduced TWOW-1 binding, suggesting alphaPS2 betaPS affinity is an active process as it is for vertebrate integrins. In contrast to vertebrate integrins, neither talin knockdown by RNA interference nor talin head overexpression had a significant effect on TWOW-1 binding. Furthermore, replacement of the transmembrane or talin-binding cytoplasmic domains of alphaPS2 betaPS with those of human alphaIIb beta3 failed to enable talin regulation of TWOW-1 binding. However, substitution of the extracellular and transmembrane domains of alphaPS2 betaPS with those of alphaIIb beta3 resulted in a constitutively active integrin whose affinity was reduced by talin knockdown. Furthermore, wild-type alphaIIb beta3 was activated by overexpression of Drosophila talin head domain. Thus, despite evolutionary conservation of talin's integrin/cytoskeleton linkage function, talin is not sufficient to regulate Drosophila alphaPS2 betaPS affinity because of structural features inherent in the alphaPS2 betaPS extracellular and/or transmembrane domains.


Asunto(s)
Drosophila/metabolismo , Regulación de la Expresión Génica , Integrinas/metabolismo , Talina/química , Animales , Células CHO , Cricetinae , Cricetulus , Citoplasma/metabolismo , Citometría de Flujo , Modelos Biológicos , Fenotipo , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN
16.
Mol Biol Cell ; 18(10): 4190-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17699602

RESUMEN

The control of gene expression by the mitogen-activated protein (MAP) kinase extracellular signal-regulated kinase (ERK) requires its translocation into the nucleus. In Drosophila S2 cells nuclear accumulation of diphospho-ERK (dpERK) is greatly reduced by interfering double-stranded RNA against Drosophila importin-7 (DIM-7) or by the expression of integrin mutants, either during active cell spreading or after stimulation by insulin. In both cases, total ERK phosphorylation (on Westerns) is not significantly affected, and ERK accumulates in a perinuclear ring. Tyrosine phosphorylation of DIM-7 is reduced in cells expressing integrin mutants, indicating a mechanistic link between these components. DIM-7 and integrins localize to the same actin-containing peripheral regions in spreading cells, but DIM-7 is not concentrated in paxillin-positive focal contacts or stable focal adhesions. The Corkscrew (SHP-2) tyrosine phosphatase binds DIM-7, and Corkscrew is required for the cortical localization of DIM-7. These data suggest a model in which ERK phosphorylation must be spatially coupled to integrin-mediated DIM-7 activation to make a complex that can be imported efficiently. Moreover, dpERK nuclear import can be restored in DIM-7-deficient cells by Xenopus Importin-7, demonstrating that ERK import is an evolutionarily conserved function of this protein.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Cadenas alfa de Integrinas/metabolismo , Carioferinas/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/enzimología , Citoplasma/efectos de los fármacos , Citoplasma/enzimología , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/enzimología , Adhesiones Focales/efectos de los fármacos , Insulina/farmacología , Modelos Biológicos , Mutación/genética , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Xenopus
17.
Dev Biol ; 308(2): 294-308, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17618618

RESUMEN

The Drosophila alphaPS2betaPS integrin is required for diverse development events, including muscle attachment. We characterized six unusual mutations in the alphaPS2 gene that cause a subset of the null phenotype. One mutation changes a residue in alphaPS2 that is equivalent to the residue in alphaV that contacts the arginine of RGD. This change severely reduced alphaPS2betaPS affinity for soluble ligand, abolished the ability of the integrin to recruit laminin to muscle attachment sites in the embryo and caused detachment of integrins and talin from the ECM. Three mutations that alter different parts of the alphaPS2 beta-propeller, plus a fourth that eliminated a late phase of alphaPS2 expression, all led to a strong decrease in alphaPS2betaPS at muscle ends, but, surprisingly, normal levels of talin were recruited. Thus, although talin recruitment requires alphaPS2betaPS, talin levels are not simply specified by the amount of integrin at the adhesive junction. These mutations caused detachment of talin and actin from integrins, suggesting that the integrin-talin link is weaker than the ECM-integrin link.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila/genética , Drosophila/fisiología , Genes de Insecto , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/fisiología , Mutación , Actinas/metabolismo , Uniones Adherentes/genética , Uniones Adherentes/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Drosophila/embriología , Proteínas de Drosophila/química , Matriz Extracelular/fisiología , Cadenas alfa de Integrinas/química , Ligandos , Masculino , Microscopía Fluorescente , Modelos Moleculares , Datos de Secuencia Molecular , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Fenotipo , Conformación Proteica , Homología de Secuencia de Aminoácido , Talina/metabolismo
18.
Anal Chem ; 79(8): 3142-7, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17346031

RESUMEN

A method for measuring the microclustering of a class of cell surface receptors called integrins is reported. Integrins are proteins involved in bidirectional signaling across the cell membrane and are important in cell adhesion, growth, and survival. Their activity is regulated by changes in protein conformation and protein clustering. The developed in vivo clustering assay uses fluorescence resonance energy transfer (FRET) and has the benefit of requiring a single cloning step to generate FRET donors and acceptors that can be used to measure the clustering of a series of integrin mutants. The FRET reporters contain extracellular donor or acceptor fluorescent protein attached to native integrin cytoplasmic and transmembrane domains, and these are expressed along with wild-type or mutant integrins. Expression of the FRET reporters has no affect on the ligand binding properties of coexpressed integrins. FRET values are calculated for cell lines spreading on ligand coated surfaces, and these values are independent of fluorescent protein expression. No FRET is observed in cell lines expressing the reporters in the absence of integrins. Integrin-dependent FRET values increase approximately 2-3-fold when the integrins contain mutations that result in increased ligand binding affinities.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Integrinas/análisis , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Células Cultivadas , Clonación Molecular , Drosophila , Integrinas/genética , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Plásmidos/química , Plásmidos/genética
19.
J Cell Biochem ; 102(1): 211-23, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17372926

RESUMEN

The Drosophila alphaPS2 integrin subunit is found in two isoforms. alphaPS2C contains 25 residues not found in alphaPS2m8, encoded by the alternative eighth exon. Previously, it was shown that cells expressing alphaPS2C spread more effectively than alphaPS2m8 cells on fragments of the ECM protein Tiggrin, and that alphaPS2C-containing integrins are relatively insensitive to depletion of Ca(2+). Using a ligand mimetic probe for Tiggrin affinity (TWOW-1), we show that the affinity of alphaPS2CbetaPS for this ligand is much higher than that of alphaPS2m8betaPS. However, the two isoforms become more similar in the presence of activating levels of Mn(2+). Modeling indicates that the exon 8-encoded residues replace the third beta strand of the third blade of the alpha subunit beta-propeller structure, and generate an exaggerated loop between this and the fourth strand. alphaPS2 subunits with the extra loop structure but with an m8-like third strand, or subunits with a C-like strand but an m8-like short loop, both fail to show alphaPS2C-like affinity for TWOW-1. Surprisingly, a single C > m8-like change at the third strand-loop transition point is sufficient to make alphaPS2C require Ca(2+) for function, despite the absence of any known cation binding site in this region. These data indicate that alternative splicing in integrin alpha subunit extracellular domains may affect ligand affinity via relatively subtle alterations in integrin conformation. These results may have relevance for vertebrate alpha6 and alpha7, which are alternatively spliced at the same site.


Asunto(s)
Empalme Alternativo , Proteínas de Drosophila/química , Cadenas alfa de Integrinas/química , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Células Cultivadas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/metabolismo , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Isoformas de Proteínas/química , Estructura Secundaria de Proteína , Subunidades de Proteína/química , Alineación de Secuencia
20.
J Biol Chem ; 281(8): 5050-7, 2006 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-16371365

RESUMEN

We developed a ligand-mimetic antibody Fab fragment specific for Drosophila alphaPS2betaPS integrins to probe the ligand binding affinities of these invertebrate receptors. TWOW-1 was constructed by inserting a fragment of the extracellular matrix protein Tiggrin into the H-CDR3 of the alphavbeta3 ligand-mimetic antibody WOW-1. The specificity of alphaPS2betaPS binding to TWOW-1 was demonstrated by numerous tests used for other integrin-ligand interactions. Binding was decreased in the presence of EDTA or RGD peptides and by mutation of the TWOW-1 RGD sequence or the betaPS metal ion-dependent adhesion site (MIDAS) motif. TWOW-1 binding was increased by mutations in the alphaPS2 membrane-proximal cytoplasmic GFFNR sequence or by exposure to Mn2+. Although Mn2+ is sometimes assumed to promote maximal integrin activity, TWOW-1 binding in Mn2+ could be increased further by the alphaPS2 GFFNR --> GFANA mutation. A mutation in the betaPS I domain (betaPS-b58; V409D) greatly increased ligand binding affinity, explaining the increased cell spreading mediated by alphaPS2betaPS-b58. Further mutagenesis of this residue suggested that Val-409 normally stabilizes the closed head conformation. Mutations that potentially reduce interaction of the integrin beta subunit plexin-semaphorin-integrin (PSI) and stalk domains have been shown to have activating properties. We found that complete deletion of the betaPS PSI domain enhanced TWOW-1 binding. Moreover the PSI domain is dispensable for at least some other integrin functions because betaPS-DeltaPSI displayed an enhanced ability to mediate cell spreading. These studies establish a means to evaluate mechanisms and consequences of integrin affinity modulation in a tractable model genetic system.


Asunto(s)
Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Cadenas alfa de Integrinas/química , Cadenas alfa de Integrinas/genética , Secuencias de Aminoácidos , Animales , Citoplasma/metabolismo , Relación Dosis-Respuesta a Droga , Drosophila , Ácido Edético/química , Integrina alfaVbeta3/metabolismo , Integrinas/química , Integrinas/metabolismo , Ligandos , Manganeso/química , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis , Mutación , Oligopéptidos/química , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA