Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 372
Filtrar
1.
Langmuir ; 40(13): 7060-7066, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38513212

RESUMEN

Nanosheet arrays with stable signal output have become promising photoactive materials for photoelectrochemical (PEC) immunosensors. However, an essential concern is the facile recombination of carriers in one-component nanoarrays, which cannot be readily prevented, ultimately resulting in weak photocurrent signals. In this study, an immunosensor using gold nanoparticle-anchored BiOI/Bi2S3 nanosheet arrays (BiOI/Bi2S3/Au) as a signal converter was fabricated for sensitive detection of cardiac troponin I (cTnI). The ternary nanosheet arrays were prepared by a simple method in which Bi2S3 was well-coated on the BiOI surface by in situ growth, whereas the addition of Au further improved the photoelectric conversion efficiency and could link more antibodies. The three-dimensional (3D) ordered sheet-like network array structure and BiOI/Bi2S3/Au ternary nanosheet arrays showed stable and high photoelectric signal output and no significant difference in signals across different batches under visible light excitation. The fabricated immunosensor has a sensitive response to the target detection marker cTnI in a wide linear range of 500 fg/mL to 50 ng/mL, and the detection limit was 32 fg/mL, demonstrating good stability and selectivity. This work not only shows the great application potential of ternary heterojunction arrays in the field of PEC immunosensors but also provides a useful exploration for improving the stability of immunosensors.


Asunto(s)
Técnicas Biosensibles , Nanopartículas del Metal , Troponina I , Técnicas Biosensibles/métodos , Técnicas Electroquímicas/métodos , Oro/química , Inmunoensayo/métodos , Límite de Detección , Troponina I/química , Troponina I/inmunología , Bismuto/química
2.
Anal Chem ; 95(31): 11687-11694, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37506038

RESUMEN

Porphyrins easily aggregate due to unfavorable π-π accumulation, causing luminescent quenching in the aqueous phase and subsequently reducing luminescent efficiency. It is a feasible way to immobilize porphyrin molecules through metal-organic framework materials (MOFs). In this study, 5,10,15,20-tetrakis (4-carboxyphenyl) porphyrin (TCPP) was introduced into the metal-organic skeleton (PCN-224) as a ligand. The result showed that the electrochemiluminescence (ECL) and photoluminescence (PL) efficiency of the MOF skeleton was 8.2 and 6.5 times higher than TCPP, respectively. Impressively, the periodic distribution of porphyrin molecules in the MOF framework can overcome the bottleneck of porphyrin aggregation, resulting in the organic ligand TCPP participating in the electron transfer reaction. Herein, based on the PCN-224, a sandwich-type ECL immunosensor was constructed for the determination of cardiac troponin I (cTnI). It provided sensitive detection of cTnI in the range of 1 fg/mL to 10 ng/mL with a detection limit of 0.34 fg/mL. This work not only innovatively exploited a disaggregation ECL (DIECL) strategy via the crystalline framework of MOF to enhance the PL and ECL efficiency of porphyrin but also provided a promising ECL platform for the ultrasensitive monitoring of cTnI.


Asunto(s)
Mediciones Luminiscentes , Glicósidos/química , Estructuras Metalorgánicas/química , Mediciones Luminiscentes/métodos , Troponina I/química , Límite de Detección , Técnicas Biosensibles/métodos , Inmunoensayo/métodos
3.
J Chem Inf Model ; 63(11): 3534-3543, 2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-37261389

RESUMEN

The cardiac thin filament comprises F-actin, tropomyosin, and troponin (cTn). cTn is composed of three subunits: troponin C (cTnC), troponin I (cTnI), and troponin T (cTnT). To computationally study the effect of the thin filament on cTn activation events, we employed targeted molecular dynamics followed by umbrella sampling using a model of the thin filament to measure the thermodynamics of cTn transition events. Our simulations revealed that the thin filament causes an increase in the free energy required to open the cTnC hydrophobic patch and causes a more favorable interaction between this region and the cTnI switch peptide. Mutations to the cTn complex can lead to cardiomyopathy, a collection of diseases that present clinically with symptoms of hypertrophy or dilation of the cardiac muscle, leading to impairment of the heart's ability to function normally and ultimately myocardial infarction or heart failure. Upon introduction of cardiomyopathic mutations to R145 of cTnI, we observed a general decrease in the free energy of opening the cTnC hydrophobic patch, which is on par with previous experimental results. These mutations also exhibited a decrease in electrostatic interactions between cTnI-R145 and actin-E334. After introduction of a small molecule to the wild-type cTnI-actin interface to intentionally disrupt intersubunit contacts, we successfully observed similar thermodynamic consequences and disruptions to the same protein-protein contacts as observed with the cardiomyopathic mutations. Computational studies utilizing the cTn complex in isolation would have been unable to observe these effects, highlighting the importance of using a more physiologically relevant thin-filament model to investigate the global consequences of cardiomyopathic mutations to the cTn complex.


Asunto(s)
Actinas , Troponina I , Troponina I/genética , Troponina I/química , Actinas/genética , Mutación , Termodinámica , Péptidos/genética , Calcio
4.
Am J Physiol Cell Physiol ; 323(6): C1728-C1739, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36280392

RESUMEN

Stretch activation is defined as a delayed increase in force after rapid stretches. Although there is considerable evidence for stretch activation in isolated cardiac myofibrillar preparations, few studies have measured mechanisms of stretch activation in mammalian skeletal muscle fibers. We measured stretch activation following rapid step stretches [∼1%-4% sarcomere length (SL)] during submaximal Ca2+ activations of rat permeabilized slow-twitch skeletal muscle fibers before and after protein kinase A (PKA), which phosphorylates slow myosin binding protein-C. PKA significantly increased stretch activation during low (∼25%) Ca2+ activation and accelerated rates of delayed force development (kef) during both low and half-maximal Ca2+ activation. Following the step stretches and subsequent force development, fibers were rapidly shortened to original sarcomere length, which often elicited a shortening-induced transient force overshoot. After PKA, step shortening-induced transient force overshoot increased ∼10-fold following an ∼4% SL shortening during low Ca2+ activation levels. kdf following step shortening also increased after PKA during low and half-maximal Ca2+ activations. We next investigated thin filament regulation of stretch activation. We tested the interplay between cardiac troponin I (cTnI) phosphorylation at the canonical PKA and novel tyrosine kinase sites on stretch activation. Native slow-skeletal Tn complexes were exchanged with recombinant human cTn complex with different human cTnI N-terminal pseudo-phosphorylation molecules: 1) nonphosphorylated wild type (WT), 2) the canonical S22/23D PKA sites, 3) the tyrosine kinase Y26E site, and 4) the combinatorial S22/23D + Y26E cTnI. All three pseudo-phosphorylated cTnIs elicited greater stretch activation than WT. Following stretch activation, a new, elevated stretch-induced steady-state force was reached with pseudo-phosphorylated cTnI. Combinatorial S22/23D + Y26E pseudo-phosphorylated cTnI increased kdf. These results suggest that slow-skeletal myosin binding protein-C (sMyBP-C) phosphorylation modulates stretch activation by a combination of cross-bridge recruitment and faster cycling kinetics, whereas cTnI phosphorylation regulates stretch activation by both redundant and synergistic mechanisms; and, taken together, these sarcomere phosphoproteins offer precision targets for enhanced contractility.


Asunto(s)
Calcio , Miofibrillas , Ratas , Humanos , Animales , Miofibrillas/metabolismo , Calcio/metabolismo , Sarcómeros/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Troponina I/química , Fosforilación , Miosinas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Miocardio/metabolismo , Contracción Miocárdica/fisiología , Mamíferos/metabolismo
5.
Biochemistry ; 61(11): 1103-1112, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35522994

RESUMEN

The C-terminal 14-16 residues of human troponin T are required for full inactivation, and they prevent full activation at saturating Ca2+. Basic residues within that C-terminal region of TnT are essential for its function, but the mechanism of action is unknown. That region of TnT is natively disordered and does not appear in reconstructions of the troponin structure. We used Förster resonance energy transfer to determine if the C-terminal basic region of TnT alters transitions of TnI or if it operates independently. We also examined Ca2+-dependent changes in the C-terminal region of TnT itself. Probes on TnI-143 (inhibitory region) and TnI-159 (switch region) moved away from sites on actin and tropomyosin and toward TnC-84 at high Ca2+. Ca2+ also displaced C-terminal TnT from actin-tropomyosin but without movement toward TnC. Deletion of C-terminal TnT produced changes in TnI-143 like those effected by Ca2+, but effects on TnI-159 were muted; there was no effect on the distance of the switch region to TnC-84. Substituting Ala for basic residues within C-terminal TnT displaced C-terminal TnT from actin-tropomyosin. The results suggest that C-terminal TnT stabilizes tropomyosin in the inactive position on actin. Removal of basic residues from C-terminal TnT produced a Ca2+-like state except that the switch region of TnI was not bound to TnC. Addition of Ca2+ caused more extreme displacement from actin-tropomyosin as the active state became more fully occupied as in the case of wild-type TnT in the presence of both Ca2+ and bound rigor myosin S1.


Asunto(s)
Troponina I , Troponina T , Actinas/metabolismo , Calcio/metabolismo , Humanos , Músculo Esquelético/metabolismo , Tropomiosina/química , Troponina C/química , Troponina C/genética , Troponina I/química , Troponina T/química , Troponina T/genética
6.
J Biol Chem ; 296: 100228, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33814345

RESUMEN

The conserved C-terminal end segment of troponin I (TnI) plays a critical role in regulating muscle relaxation. This function is retained in the isolated C-terminal 27 amino acid peptide (residues 184-210) of human cardiac TnI (HcTnI-C27): When added to skinned muscle fibers, HcTnI-C27 reduces the Ca2+-sensitivity of activated myofibrils and facilitates relaxation without decreasing the maximum force production. However, the underlying mechanism of HcTnI-C27 function is unknown. We studied the conformational preferences of HcTnI-C27 and a myopathic mutant, Arg192His, (HcTnI-C27-H). Both peptides were mainly disordered in aqueous solution with a nascent helix involving residues from Trp191 to Ile195, as shown by NMR analysis and molecular dynamics simulations. The population of nascent helix was smaller in HcTnI-C27-H than in HcTnI-C27, as shown by circular dichroism (CD) titrations. Fluorescence and isothermal titration calorimetry (ITC) showed that both peptides bound tropomyosin (αTm), with a detectably higher affinity (∼10 µM) of HcTnI-C27 than that of HcTnI-C27-H (∼15 µM), consistent with an impaired Ca2+-desensitization effect of the mutant peptide on skinned muscle strips. Upon binding to αTm, HcTnI-C27 acquired a weakly stable helix-like conformation involving residues near Trp191, as shown by transferred nuclear Overhauser effect spectroscopy and hydrogen/deuterium exchange experiments. With the potent Ca2+-desensitization effect of HcTnI-C27 on skinned cardiac muscle from a mouse model of hypertrophic cardiomyopathy, the data support that the C-terminal end domain of TnI can function as an isolated peptide with the intrinsic capacity of binding tropomyosin, providing a promising therapeutic approach to selectively improve diastolic function of the heart.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Fibras Musculares Esqueléticas/metabolismo , Miofibrillas/metabolismo , Péptidos/química , Tropomiosina/metabolismo , Troponina I/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Calcio/metabolismo , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/prevención & control , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Cinética , Ratones , Simulación del Acoplamiento Molecular , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/patología , Relajación Muscular , Mutación , Miofibrillas/efectos de los fármacos , Miofibrillas/patología , Péptidos/genética , Péptidos/metabolismo , Péptidos/farmacología , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Tropomiosina/química , Tropomiosina/genética , Troponina I/genética , Troponina I/metabolismo
7.
J Mol Evol ; 90(1): 30-43, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34966949

RESUMEN

Troponin-based Ca2+ regulation of striated muscle contraction emerged approximately 700 million years ago with largely conserved functions during evolution. Troponin I (TnI) is the inhibitory subunit of troponin and has evolved into three muscle type-specific isoforms in vertebrates. Cardiac TnI is specifically expressed in the adult heart and has a unique N-terminal extension implicating a specific value during natural selection. The N-terminal extension of cardiac TnI in higher vertebrates contains ß-adrenergic-regulated protein kinase A (PKA) phosphorylation sites as a mechanism to enhance cardiac muscle relaxation and facilitate ventricular filling. Phylogenic studies showed that the N-terminal extension of cardiac TnI first emerged in the genomes of early tetrapods as well as primordial lobe-finned fishes such as the coelacanth whereas it is absent in ray-finned fish. This apparently rapid evolution of ß-adrenergic regulation of cardiac function suggests a high selection value for the heart of vertebrate animals on land to work under higher metabolic demands. Sequencing and PKA phosphorylation data showed that lungfish cardiac TnI has evolved with an amphibian-like N-terminal extension with prototype PKA phosphorylation sites while its overall structure remained fish like. The data demonstrate that the submolecular structure of TnI may evolve ahead of the whole protein for cardiac muscle contractility to adapt to new environmental conditions. Understanding the evolution of the ß-adrenergic regulation of TnI and cardiac adaptation to the increased energetic demands of life on land adds knowledge for the treatment of human heart diseases and failure.


Asunto(s)
Corazón , Miocardio , Troponina I , Adrenérgicos/metabolismo , Animales , Peces , Miocardio/metabolismo , Fosforilación , Troponina I/química , Troponina I/genética , Troponina I/metabolismo
8.
Arch Biochem Biophys ; 725: 109282, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35577070

RESUMEN

Tropomyosin, controlled by troponin-linked Ca2+-binding, regulates muscle contraction by a macromolecular scale steric-mechanism that governs myosin-crossbridge-actin interactions. At low-Ca2+, C-terminal domains of troponin-I (TnI) trap tropomyosin in a position on thin filaments that interferes with myosin-binding, thus causing muscle relaxation. Steric inhibition is reversed at high-Ca2+ when TnI releases from F-actin-tropomyosin as Ca2+ and the TnI switch-peptide bind to the N-lobe of troponin-C (TnC). The opposite end of cardiac TnI contains a phosphorylation-sensitive ∼30 residue-long N-terminal peptide that is absent in skeletal muscle, and likely modifies these interactions in hearts. Here, PKA-dependent phosphorylation of serine 23 and 24 modulates Ca2+ and possibly switch-peptide binding to TnC, causing faster relaxation during the cardiac-cycle (lusitropy). The cardiac-specific N-terminal TnI domain is not captured in crystal structures of troponin or in cryo-EM reconstructions of thin filaments; thus, its global impact on thin filament structure and function is uncertain. Here, we used protein-protein docking and molecular dynamics simulation-based protocols to build a troponin model that was guided by and hence consistent with the recent seminal Yamada structure of Ca2+-activated thin filaments. We find that when present on thin filaments, phosphorylated Ser23/24 along with adjacent polar TnI residues interact closely with both tropomyosin and the N-lobe of TnC during our simulations. These interactions would likely bias tropomyosin to an off-state positioning on actin. In situ, such enhanced relaxation kinetics would promote cardiac lusitropy.


Asunto(s)
Tropomiosina , Troponina I , Actinas/metabolismo , Calcio/metabolismo , Simulación de Dinámica Molecular , Péptidos/metabolismo , Tropomiosina/química , Troponina C/metabolismo , Troponina I/química
9.
Mol Cell Biochem ; 477(6): 1803-1815, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35316461

RESUMEN

The cardiac isoform of troponin I has a unique N-terminal extension (~ 1-30 amino acids), which contributes to the modulation of cardiac contraction and relaxation. Hearts of various species including humans produce a truncated variant of cardiac troponin I (cTnI-ND) deleting the first ~ 30 amino acids as an adaption in pathophysiological conditions. In this study, we investigated the impact of cTnI-ND chronic expression in transgenic mouse hearts compared to wildtype (WT) controls (biological n = 8 in each group). We aimed to determine the global phosphorylation effects of cTnI-ND on the cardiac proteome, thereby determining the signaling pathways that have an impact on cardiac function. The samples were digested and isobarically labeled and equally mixed for relative quantification via nanoLC-MS/MS. The peptides were then enriched for phospho-peptides and bioinformatic analysis was done with Ingenuity Pathway Analysis (IPA). We found approximately 77% replacement of the endogenous intact cTnI with cTnI-ND in the transgenic mouse hearts with 1674 phospho-proteins and 2971 non-modified proteins. There were 73 significantly altered phospho-proteins; bioinformatic analysis identified the top canonical pathways as associated with integrin, protein kinase A, RhoA, and actin cytoskeleton signaling. Among the 73 phospho-proteins compared to controls cTnI-ND hearts demonstrated a significant decrease in paxillin and YAP1, which are known to play a role in cell mechano-sensing pathways. Our data indicate that cTnI-ND modifications in the sarcomere are sufficient to initiate changes in the phospho-signaling profile that may underly the chronic-adaptive response associated with cTnI cleavage in response to stressors by modifying mechano-sensitive signaling pathways.


Asunto(s)
Espectrometría de Masas en Tándem , Troponina I , Aminoácidos , Animales , Calcio/metabolismo , Ratones , Ratones Transgénicos , Contracción Miocárdica , Miocardio/metabolismo , Péptidos , Fosforilación , Transducción de Señal , Troponina I/química , Troponina I/genética , Troponina I/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(14): 6969-6974, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30886088

RESUMEN

Sudden unexpected death of an infant (SUDI) is a devastating occurrence for families. To investigate the genetic pathogenesis of SUDI, we sequenced >70 genes from 191 autopsy-negative SUDI victims. Ten infants sharing a previously unknown variant in troponin I (TnI) were identified. The mutation (TNNI1 R37C+/-) is in the fetal/neonatal paralog of TnI, a gene thought to be expressed in the heart up to the first 24 months of life. Using phylogenetic analysis and molecular dynamics simulations, it was determined that arginine at residue 37 in TNNI1 may play a critical functional role, suggesting that the variant may be pathogenic. We investigated the biophysical properties of the TNNI1 R37C mutation in human reconstituted thin filaments (RTFs) using fluorometry. RTFs reconstituted with the mutant R37C TnI exhibited reduced Ca2+-binding sensitivity due to an increased Ca2+ off-rate constant. Furthermore, we generated TNNI1 R37C+/- mutants in human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) using CRISPR-Cas9. In monolayers of hiPSC-CMs, we simultaneously monitored voltage and Ca2+ transients through optical mapping and compared them to their isogenic controls. We observed normal intrinsic beating patterns under control conditions in TNNI1 R37C+/- at stimulation frequencies of 55 beats/min (bpm), but these cells showed no restitution with increased stimulation frequency to 65 bpm and exhibited alternans at >75 bpm. The WT hiPSC-CMs did not exhibit any sign of arrhythmogenicity even at stimulation frequencies of 120 bpm. The approach used in this study provides critical physiological and mechanistic bases to investigate sarcomeric mutations in the pathogenesis of SUDI.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Simulación de Dinámica Molecular , Mutación Missense , Miocitos Cardíacos/metabolismo , Muerte Súbita del Lactante/genética , Troponina I , Calcio/química , Calcio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/patología , Recién Nacido , Contracción Miocárdica/genética , Miocitos Cardíacos/patología , Sarcómeros/genética , Sarcómeros/metabolismo , Sarcómeros/patología , Muerte Súbita del Lactante/patología , Troponina I/química , Troponina I/genética , Troponina I/metabolismo
11.
J Mol Cell Cardiol ; 155: 50-57, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33647310

RESUMEN

The cardiac thin filament is regulated in a Ca2+-dependent manner through conformational changes of troponin and tropomyosin (Tm). It has been generally understood that under conditions of low Ca2+ the inhibitory peptide domain (IP) of troponin I (TnI) binds to actin and holds Tm over the myosin binding sites on actin to prevent crossbridge formation. More recently, evidence that the C-terminal mobile domain (MD) of TnI also binds actin has made for a more complex scenario. This study uses a computational model to investigate the consequences of assuming that TnI regulates Tm movement via two actin-binding domains rather than one. First, a 16-state model of the cardiac thin filament regulatory unit was created with TnI-IP as the sole regulatory domain. Expansion of this to include TnI-MD formed a 24-state model. Comparison of these models showed that assumption of a second actin-binding site allows the individual domains to have a lower affinity for actin than would be required for IP acting alone. Indeed, setting actin affinities of the IP and MD to 25% of that assumed for the IP in the single-site model was sufficient to achieve precisely the same degree of Ca2+ regulation. We also tested the 24-state model's ability to represent steady-state experimental data in the case of disruption of either the IP or MD. We were able to capture qualitative changes in several properties that matched what was seen in the experimental data. Lastly, simulations were run to examine the effect of disruption of the IP or MD on twitch dynamics. Our results suggest that both domains are required to keep diastolic cross-bridge activity to a minimum and accelerate myofilament relaxation. Overall, our analyses support a paradigm in which two domains of TnI bind with moderate affinity to actin, working in tandem to complete Ca2+-dependent regulation of the thin filament.


Asunto(s)
Modelos Biológicos , Contracción Miocárdica , Miofibrillas/metabolismo , Dominios y Motivos de Interacción de Proteínas , Troponina I/metabolismo , Algoritmos , Animales , Humanos , Cadenas de Markov , Método de Montecarlo , Unión Proteica , Troponina I/química
12.
J Mol Cell Cardiol ; 150: 44-53, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33080242

RESUMEN

Troponin is the Ca2+ molecular switch that regulates striated muscle contraction. In the heart, troponin Ca2+ sensitivity is also modulated by the PKA-dependent phosphorylation of a unique 31-residue N-terminal extension region of the Troponin I subunit (NH2-TnI). However, the detailed mechanism for the propagation of the phosphorylation signal through Tn, which results in the enhancement of the myocardial relaxation rate, is difficult to examine within whole Tn. Several models exist for how phosphorylation modulates the troponin response in cardiac cells but these are mostly built from peptide-NMR studies and molecular dynamics simulations. Here we used a paramagnetic spin labeling approach to position and track the movement of the NH2-TnI region within whole Tn. Through paramagnetic relaxation enhancement (PRE)-NMR experiments, we show that the NH2-TnI region interacts with a broad surface area on the N-domain of the Troponin C subunit. This region includes the Ca2+ regulatory Site II and the TnI switch-binding site. Phosphorylation of the NH2-TnI both weakens and shifts this region to an adjacent site on TnC. Interspin EPR distances between NH2-TnI and TnC further reveal a phosphorylation induced re-orientation of the TnC N-domain under saturating Ca2+ conditions. We propose an allosteric model where phosphorylation triggered cooperative changes in both the interaction of the NH2-TnI region with TnC, and the re-orientation of the TnC interdomain orientation, together promote the release of the TnI switch-peptide. Enhancement of the myocardial relaxation rate then occurs. Knowledge of this unique role of phosphorylation in whole Tn is important for understanding pathological processes affecting the heart.


Asunto(s)
Contracción Miocárdica/fisiología , Miocardio/metabolismo , Troponina I/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Isótopos de Nitrógeno , Fosforilación , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Ratas , Marcadores de Spin , Troponina I/química
13.
J Biol Chem ; 295(14): 4398-4410, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32086378

RESUMEN

Heart muscle contractility and performance are controlled by posttranslational modifications of sarcomeric proteins. Although myosin regulatory light chain (RLC) phosphorylation has been studied extensively in vitro and in vivo, the precise role of cardiac myosin light chain kinase (cMLCK), the primary kinase acting upon RLC, in the regulation of cardiomyocyte contractility remains poorly understood. In this study, using recombinantly expressed and purified proteins, various analytical methods, in vitro and in situ kinase assays, and mechanical measurements in isolated ventricular trabeculae, we demonstrate that human cMLCK is not a dedicated kinase for RLC but can phosphorylate other sarcomeric proteins with well-characterized regulatory functions. We show that cMLCK specifically monophosphorylates Ser23 of human cardiac troponin I (cTnI) in isolation and in the trimeric troponin complex in vitro and in situ in the native environment of the muscle myofilament lattice. Moreover, we observed that human cMLCK phosphorylates rodent cTnI to a much smaller extent in vitro and in situ, suggesting species-specific adaptation of cMLCK. Although cMLCK treatment of ventricular trabeculae exchanged with rat or human troponin increased their cross-bridge kinetics, the increase in sensitivity of myofilaments to calcium was significantly blunted by human TnI, suggesting that human cTnI phosphorylation by cMLCK modifies the functional consequences of RLC phosphorylation. We propose that cMLCK-mediated phosphorylation of TnI is functionally significant and represents a critical signaling pathway that coordinates the regulatory states of thick and thin filaments in both physiological and potentially pathophysiological conditions of the heart.


Asunto(s)
Contracción Miocárdica/fisiología , Miocardio/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Troponina I/metabolismo , Animales , Calcio/metabolismo , Humanos , Masculino , Miofibrillas/metabolismo , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/química , Quinasa de Cadena Ligera de Miosina/genética , Péptidos/análisis , Péptidos/química , Fosforilación , Ratas , Ratas Wistar , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Transducción de Señal , Troponina I/química , Troponina I/genética
14.
Biochem Biophys Res Commun ; 551: 27-32, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33714756

RESUMEN

Tropomyosin and troponin regulate muscle contraction by participating in a macromolecular scale steric-mechanism to control myosin-crossbridge - actin interactions and consequently contraction. At low-Ca2+, the C-terminal 30% of troponin subunit-I (TnI) is proposed to trap tropomyosin in a position on thin filaments that sterically interferes with myosin-binding, thus causing muscle relaxation. In contrast, at high-Ca2+, inhibition is released after the C-terminal domains dissociate from F-actin-tropomyosin as its component switch-peptide domain binds to the N-lobe of troponin-C (TnC). Recent, paradigm-shifting, cryo-EM reconstructions by the Namba group have revealed density attributed to TnI along cardiac muscle thin filaments at both low- and high-Ca2+ concentration. Modeling the reconstructions showed expected high-Ca2+ hydrophobic interactions of the TnI switch-peptide and TnC. However, under low-Ca2+ conditions, sparse interactions of TnI and tropomyosin, and in particular juxtaposition of non-polar switch-peptide residues and charged tropomyosin amino acids in the published model seem difficult to reconcile with an expected steric-blocking conformation. This anomaly is likely due to inaccurate fitting of tropomyosin into the cryo-EM volume. In the current study, the low-Ca2+ cryo-EM volume was fitted with a more accurate tropomyosin model and representation of cardiac TnI. Our results show that at low-Ca2+ a cluster of hydrophobic residues at the TnI switch-peptide and adjacent H4 helix (Ala149, Ala151, Met 154, Leu159, Gly160, Ala161, Ala163, Leu167, Leu169, Ala171, Leu173) draw-in tropomyosin surface residues (Ile143, Ile146, Ala151, Ile154), presumably attracting the entire tropomyosin cable to its myosin-blocking position on actin. The modeling confirms that neighboring TnI "inhibitory domain" residues (Arg145, Arg148) bind to thin filaments at actin residue Asp25, as previously suggested. ClusPro docking of TnI residues 137-184 to actin-tropomyosin, including the TnI inhibitory-domain, switch-peptide and Helix H4, verified the modeled configuration. Our residue-to-residue contact-mapping of the TnI-tropomyosin association lends itself to experimental validation and functional localization of disease-bearing mutations.


Asunto(s)
Músculo Esquelético/metabolismo , Tropomiosina/metabolismo , Troponina I/química , Troponina I/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actinas/química , Actinas/metabolismo , Animales , Calcio/metabolismo , Microscopía por Crioelectrón , Humanos , Simulación del Acoplamiento Molecular , Relajación Muscular , Músculo Esquelético/química , Unión Proteica , Dominios Proteicos , Reproducibilidad de los Resultados , Tropomiosina/química , Troponina I/genética
15.
Anal Bioanal Chem ; 413(19): 4847-4854, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34115147

RESUMEN

An interference-free photoelectrochemical (PEC) immunoassay was developed for cardiac troponin I (cTnI) detection. Covalent linkage of cTnI antibody to carboxymethylated (CM-) dextran pre-immobilized onto a gold nanoparticles (AuNPs)-modified TiO2 nanotube array (NTA) affords five consecutive analyte captures with surface regenerations in between. Changes in the photocurrents at this photoanode before and after cTnI captures can be well fitted with the Langmuir isotherm from 0.220 pM to 2.20 nM cTnI. Owing to the inherently high sensitivity of the PEC detection, the detection limit (2.20 pg/mL) is lower than the range attainable with the enzyme-linked immunosorbent assay (ELISA) (6.00-40.0 pg/mL). Furthermore, CM-dextran prevents species in complex biological matrices from nonspecifically adsorbing onto the sensor surface, a feature not attainable with uncoated semiconductor electrodes or those coated with non-hydrogel-based chemical modifiers. The excellent anti-fouling property of dextran hydrogel allowed us to validate the accuracy of our regenerable sensors through a comparison of PEC immunoassays of patient sera to those of ELISA.


Asunto(s)
Dextranos/química , Técnicas Electroquímicas , Oro/química , Inmunoensayo/métodos , Procesos Fotoquímicos , Titanio/química , Biomarcadores , Electrodos , Humanos , Nanotubos/química , Troponina I/química
16.
Mikrochim Acta ; 189(1): 22, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34882274

RESUMEN

A sensitive label-based SERS strategy composed of magnetic bimetallic nanoparticles Fe3O4@Ag@Au, specific aptamer, and Bradford method was developed for the quantitative determination of cardiac troponin I (cTnI) in human serum. The prepared substrate with high magnetic character, signal enhancement, and uniformity exhibited significant Raman response. After the substrate was bound to the aptamer, the target protein cTnI was specifically captured, and it showed the Raman signal when the signal reporter Coomassie Brilliant Blue G-250 (CBBG) was supplied. The Raman signal intensity at 1621 cm-1 showed a wide linear relationship with the log value of the cTnI concentration in the range 0.01 to 100 ng·mL-1, and the estimated limit of detection (LOD) was 5.50 pg·mL-1. The recovery and relative standard deviation (RSD) of the spike experiment in human serum samples were 92-115% and 7.4-12.7%, respectively.


Asunto(s)
Aptámeros de Nucleótidos/química , Espectrometría Raman/métodos , Troponina I/sangre , Técnicas Biosensibles/métodos , ADN/química , Oro/química , Humanos , Límite de Detección , Fenómenos Magnéticos , Nanopartículas de Magnetita/química , Colorantes de Rosanilina/química , Plata/química , Troponina I/química
17.
Biochemistry ; 59(43): 4189-4201, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33074652

RESUMEN

Calcium binding to troponin C (TnC) activates striated muscle contraction by removing TnI (troponin I) from its inhibitory site on actin. Troponin T (TnT) links TnI with tropomyosin, causing tropomyosin to move from an inhibitory position on actin to an activating position. Positive charges within the C-terminal region of human cardiac TnT limit Ca2+ activation. We now show that the positively charged region of TnT has an even larger impact on skeletal muscle regulation. We prepared one variant of human skeletal TnT that had the C-terminal 16 residues truncated (Δ16) and another with an added C-terminal Cys residue and Ala substituted for the last 6 basic residues (251C-HAHA). Both mutants reduced (based on S1 binding kinetics) or eliminated (based on acrylodan-tropomyosin fluorescence) the first inactive state of actin at <10 nM free Ca2+. 251C-HAHA-TnT and Δ16-TnT mutants greatly increased ATPase activation at 0.2 mM Ca2+, even without high-affinity cross-bridge binding. They also shifted the force-pCa curve of muscle fibers to lower Ca2+ by 0.8-1.2 pCa units (the larger shift for 251C-HAHA-TnT). Shifts in force-pCa were maintained in the presence of para-aminoblebbistatin. The effects of modification of the C-terminal region of TnT on the kinetics of S1 binding to actin were somewhat different from those observed earlier with the cardiac analogue. In general, the C-terminal region of human skeletal TnT is critical to regulation, just as it is in the cardiac system, and is a potential target for modulating activity.


Asunto(s)
Calcio/farmacología , Troponina T/metabolismo , Humanos , Cinética , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Tropomiosina/química , Tropomiosina/metabolismo , Troponina C/química , Troponina C/metabolismo , Troponina I/química , Troponina I/metabolismo , Troponina T/química
18.
Anal Chem ; 92(21): 14640-14647, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33090771

RESUMEN

A simple, dual-modular aptasensor for accurate determination of cardiac troponin I (cTnI), a sensitive biomarker of acute myocardial infarction, is reported. It has the parallel output of electrochemiluminescence (ECL) and electrochemical impedance spectroscopy (EIS) based on target-gated transportation of signal probes (luminol/H2O2 or Fe(CN)63-/4-). The sensing capacity is originated from the amino-functionalized mouth margin of the nanochannels in a vertically oriented mesoporous silica film, which was in situ-grown on indium tin oxide-coated glass. With the linkage of glutaraldehyde to couple the aptamer as a trapper, it brings in the high specific target-gated response toward cTnI as decreased ECL or increased EIS. The concentration of cTnI is measurable by the ECL response within a wide linear range from 0.05 pg mL-1 to 10 ng mL-1, as well as the EIS response for a linear range between 0.05 pg mL-1 and 1 ng mL-1. Significantly, the self-verification of these two data from ECL and EIS validated each other with a satisfactory linear correlation (R2 = 0.999), thereby realizing the more reliable and accurate quantification to avoid false results. The designed strategy is an effective method for detection of cTnI, which is of great potential to apply in clinical detection.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Técnicas Biosensibles/métodos , Dióxido de Silicio/química , Troponina I/análisis , Aptámeros de Nucleótidos/química , Espectroscopía Dieléctrica , Electroquímica , Electrodos , Humanos , Límite de Detección , Luminiscencia , Porosidad , Troponina I/química , Troponina I/metabolismo
19.
Proc Natl Acad Sci U S A ; 114(27): 7089-7094, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28630342

RESUMEN

The role of autoimmunity in cardiovascular (CV) diseases has been increasingly recognized. Autoimmunity is most commonly examined by the levels of circulating autoantibodies in clinical practices. Measurement of autoantibodies remains, however, challenging because of the deficiency of reproducible, sensitive, and standardized assays. The lack of multiplexed assays also limits the potential to identify a CV-specific autoantibody profile. To overcome these challenges, we developed a nanotechnology-based plasmonic gold chip for autoantibody profiling. This approach allowed simultaneous detection of 10 CV autoantibodies targeting the structural myocardial proteins, the neurohormonal regulatory proteins, the vascular proteins, and the proteins associated with apoptosis and coagulation. Autoantibodies were measured in four groups of participants across the continuum of hypertensive heart diseases. We observed higher levels of all 10 CV autoantibodies in hypertensive subjects (n = 77) compared with healthy participants (n = 30), and the autoantibodies investigated were related to each other, forming a highly linked network. In addition, we established that autoantibodies to troponin I, annexin-A5, and beta 1-adrenegic receptor best discriminated hypertensive subjects with adverse left ventricular (LV) remodeling or dysfunction (n = 49) from hypertensive subjects with normal LV structure and function (n = 28). By further linking these three significant CV autoantibodies to the innate and growth factors, we revealed a positive but weak association between autoantibodies to troponin I and proinflammatory cytokine IL-18. Overall, we demonstrated that this platform can be used to evaluate autoantibody profiles in hypertensive subjects at risk for heart failure.


Asunto(s)
Autoanticuerpos/química , Oro/química , Cardiopatías/diagnóstico , Hipertensión/diagnóstico , Nanopartículas del Metal/química , Anciano , Anexina A5/química , Autoinmunidad , Estudios de Casos y Controles , Ecocardiografía , Femenino , Cardiopatías/inmunología , Insuficiencia Cardíaca/inmunología , Humanos , Hipertensión/inmunología , Masculino , Persona de Mediana Edad , Nanotecnología , Receptores Adrenérgicos beta 1/química , Riesgo , Troponina I/química
20.
J Mol Cell Cardiol ; 136: 42-52, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31505197

RESUMEN

The C-terminal end segment of troponin subunit I (TnI) is a structure highly conserved among the three muscle type-specific isoforms and across vertebrate species. Partial deletion or point mutation in this segment impairs cardiac muscle relaxation. In the present study, we characterized the C-terminal 27 amino acid peptide of human cardiac TnI (HcTnI-C27) for its role in modulating muscle contractility. Biologically or chemically synthesized HcTnI-C27 peptide retains an epitope structure in physiological solutions similarly to that in intact TnI as recognized by an anti-TnI C-terminus monoclonal antibody (mAb TnI-1). Protein binding studies found that HcTnI-C27 retains the binding affinity for tropomyosin as previously shown with intact cardiac TnI. A restrictive cardiomyopathy mutation R192H in this segment abolishes the bindings to mAb TnI-1 and tropomyosin, demonstrating a pathogenic loss of function. Contractility studies using skinned muscle preparations demonstrated that addition of HcTnI-C27 peptide reduces the Ca2+-sensitivity of myofibrils without decreasing maximum force production. The results indicate that the C-terminal end segment of TnI is a regulatory element of troponin, which retains the native configuration in the form of free peptide to confer an effect on myofilament Ca2+-desensitization. Without negative inotropic impact, this short peptide may be developed into a novel reagent to selectively facilitate cardiac muscle relaxation at the activated state as a potential treatment for heart failure.


Asunto(s)
Calcio/metabolismo , Miofibrillas/metabolismo , Troponina I/química , Troponina I/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Epítopos/química , Evolución Molecular , Ventrículos Cardíacos/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Relajación Muscular/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Ratas , Tropomiosina/metabolismo , Troponina I/genética , Troponina I/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA