Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 6947, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37935654

RESUMEN

Disease-causing mutations in genes encoding transcription factors (TFs) can affect TF interactions with their cognate DNA-binding motifs. Whether and how TF mutations impact upon the binding to TF composite elements (CE) and the interaction with other TFs is unclear. Here, we report a distinct mechanism of TF alteration in human lymphomas with perturbed B cell identity, in particular classic Hodgkin lymphoma. It is caused by a recurrent somatic missense mutation c.295 T > C (p.Cys99Arg; p.C99R) targeting the center of the DNA-binding domain of Interferon Regulatory Factor 4 (IRF4), a key TF in immune cells. IRF4-C99R fundamentally alters IRF4 DNA-binding, with loss-of-binding to canonical IRF motifs and neomorphic gain-of-binding to canonical and non-canonical IRF CEs. IRF4-C99R thoroughly modifies IRF4 function by blocking IRF4-dependent plasma cell induction, and up-regulates disease-specific genes in a non-canonical Activator Protein-1 (AP-1)-IRF-CE (AICE)-dependent manner. Our data explain how a single mutation causes a complex switch of TF specificity and gene regulation and open the perspective to specifically block the neomorphic DNA-binding activities of a mutant TF.


Asunto(s)
Factores Reguladores del Interferón , Linfoma , Humanos , Linfocitos B/metabolismo , ADN , Regulación de la Expresión Génica , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Linfoma/genética
2.
Commun Biol ; 6(1): 4, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36596888

RESUMEN

Hypertrophic cardiomyopathy is an inherited disorder due to mutations in contractile proteins that results in a stiff, hypercontractile myocardium. To understand the role of cardiac stiffness in disease progression, here we create an in vitro model of hypertrophic cardiomyopathy utilizing hydrogel technology. Culturing wild-type cardiac myocytes on hydrogels with a Young's Moduli (stiffness) mimicking hypertrophic cardiomyopathy myocardium is sufficient to induce a hypermetabolic mitochondrial state versus myocytes plated on hydrogels simulating healthy myocardium. Significantly, these data mirror that of myocytes isolated from a murine model of human hypertrophic cardiomyopathy (cTnI-G203S). Conversely, cTnI-G203S myocyte mitochondrial function is completely restored when plated on hydrogels mimicking healthy myocardium. We identify a mechanosensing feedback mechanism between the extracellular matrix and cytoskeletal network that regulates mitochondrial function under healthy conditions, but participates in the progression of hypertrophic cardiomyopathy pathophysiology resulting from sarcomeric gene mutations. Importantly, we pinpoint key 'linker' sites in this schema that may represent potential therapeutic targets.


Asunto(s)
Cardiomiopatía Hipertrófica , Ratones , Humanos , Animales , Retroalimentación , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Citoesqueleto/metabolismo , Miocitos Cardíacos/metabolismo , Troponina I/genética , Troponina I/metabolismo , Matriz Extracelular/metabolismo , Hidrogeles
3.
Sci Immunol ; 8(79): eade7953, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36662884

RESUMEN

Interferon regulatory factor 4 (IRF4) is a transcription factor (TF) and key regulator of immune cell development and function. We report a recurrent heterozygous mutation in IRF4, p.T95R, causing an autosomal dominant combined immunodeficiency (CID) in seven patients from six unrelated families. The patients exhibited profound susceptibility to opportunistic infections, notably Pneumocystis jirovecii, and presented with agammaglobulinemia. Patients' B cells showed impaired maturation, decreased immunoglobulin isotype switching, and defective plasma cell differentiation, whereas their T cells contained reduced TH17 and TFH populations and exhibited decreased cytokine production. A knock-in mouse model of heterozygous T95R showed a severe defect in antibody production both at the steady state and after immunization with different types of antigens, consistent with the CID observed in these patients. The IRF4T95R variant maps to the TF's DNA binding domain, alters its canonical DNA binding specificities, and results in a simultaneous multimorphic combination of loss, gain, and new functions for IRF4. IRF4T95R behaved as a gain-of-function hypermorph by binding to DNA with higher affinity than IRF4WT. Despite this increased affinity for DNA, the transcriptional activity on IRF4 canonical genes was reduced, showcasing a hypomorphic activity of IRF4T95R. Simultaneously, IRF4T95R functions as a neomorph by binding to noncanonical DNA sites to alter the gene expression profile, including the transcription of genes exclusively induced by IRF4T95R but not by IRF4WT. This previously undescribed multimorphic IRF4 pathophysiology disrupts normal lymphocyte biology, causing human disease.


Asunto(s)
Regulación de la Expresión Génica , Factores Reguladores del Interferón , Ratones , Animales , Humanos , Linfocitos B , ADN/metabolismo , Mutación
4.
Int J Mol Sci ; 23(14)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35887322

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of respiratory infections in infants and the elderly. Although the RSV matrix (M) protein has key roles in the nucleus early in infection, and in the cytoplasm later, the molecular basis of switching between the nuclear and cytoplasmic compartments is not known. Here, we show that protein kinase CK2 can regulate M nucleocytoplasmic distribution, whereby inhibition of CK2 using the specific inhibitor 4,5,6,7-tetrabromobenzo-triazole (TBB) increases M nuclear accumulation in infected cells as well as when ectopically expressed in transfected cells. We use truncation/mutagenic analysis for the first time to show that serine (S) 95 and threonine (T) 205 are key CK2 sites that regulate M nuclear localization. Dual alanine (A)-substitution to prevent phosphorylation abolished TBB- enhancement of nuclear accumulation, while aspartic acid (D) substitution to mimic phosphorylation at S95 increased nuclear accumulation. D95 also induced cytoplasmic aggregate formation, implying that a negative charge at S95 may modulate M oligomerization. A95/205 substitution in recombinant RSV resulted in reduced virus production compared with wild type, with D95/205 substitution resulting in an even greater level of attenuation. Our data support a model where unphosphorylated M is imported into the nucleus, followed by phosphorylation of T205 and S95 later in infection to facilitate nuclear export and cytoplasmic retention of M, respectively, as well as oligomerization/virus budding. In the absence of widely available, efficacious treatments to protect against RSV, the results raise the possibility of antiviral strategies targeted at CK2.


Asunto(s)
Virus Sincitial Respiratorio Humano , Transporte Activo de Núcleo Celular , Anciano , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Fosforilación
5.
Protein Sci ; 31(5): e4311, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35481653

RESUMEN

Excitation-contraction coupling (ECC) is the physiological process in which an electrical signal originating from the central nervous system is converted into muscle contraction. In skeletal muscle tissue, the key step in the molecular mechanism of ECC initiated by the muscle action potential is the cooperation between two Ca2+ channels, dihydropyridine receptor (DHPR; voltage-dependent L-type calcium channel) and ryanodine receptor 1 (RyR1). These two channels were originally postulated to communicate with each other via direct mechanical interactions; however, the molecular details of this cooperation have remained ambiguous. Recently, it has been proposed that one or more supporting proteins are in fact required for communication of DHPR with RyR1 during the ECC process. One such protein that is increasingly believed to play a role in this interaction is the SH3 and cysteine-rich domain-containing protein 3 (STAC3), which has been proposed to bind a cytosolic portion of the DHPR α1S subunit known as the II-III loop. In this work, we present direct evidence for an interaction between a small peptide sequence of the II-III loop and several residues within the SH3 domains of STAC3 as well as the neuronal isoform STAC2. Differences in this interaction between STAC3 and STAC2 suggest that STAC3 possesses distinct biophysical features that are potentially important for its physiological interactions with the II-III loop. Therefore, this work demonstrates an isoform-specific interaction between STAC3 and the II-III loop of DHPR and provides novel insights into a putative molecular mechanism behind this association in the skeletal muscle ECC process.


Asunto(s)
Canales de Calcio Tipo L , Canal Liberador de Calcio Receptor de Rianodina , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Acoplamiento Excitación-Contracción/fisiología , Músculo Esquelético/fisiología , Isoformas de Proteínas/metabolismo
6.
Protein Sci ; 31(4): 787-796, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34913532

RESUMEN

Interferon regulatory factor 4 (IRF4) is an essential regulator in the development of many immune cells, including B- and T-cells and has been implicated directly in numerous hematological malignancies, including adult T-cell leukemia/lymphoma (ATLL). Recently, an activating mutation in the DNA-binding domain of IRF4 (IRF4K59R ) was found as a recurrent somatic mutation in ATLL patients. However, it remains unknown how this mutation gives rise to the observed oncogenic effect. To understand the mode of IRF4K59R -mediated gain of function in ATLL pathogenesis, we have determined the structural and affinity basis of IRF4K59R /DNA homodimer complex using X-ray crystallography and surface plasmon resonance. Our study shows that arginine substitution (R59) results in the reorientation of the side chain, enabling the guanidium group to interact with the phosphate backbone of the DNA helix. This markedly contrasts with the IRF4WT wherein the K59 interacts exclusively with DNA bases. Further, the arginine mutation causes enhanced DNA bending, enabling the IRF4K59R to interact more robustly with known DNA targets, as evidenced by increased binding affinity of the protein-DNA complex. Together, we demonstrate how key structural features underpin the basis for this activating mutation, thereby providing a molecular rationale for IRF4K59R -mediated ATLL development.


Asunto(s)
Factores Reguladores del Interferón , Leucemia-Linfoma de Células T del Adulto , Adulto , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/metabolismo , Leucemia-Linfoma de Células T del Adulto/patología , Mutación
7.
Cancers (Basel) ; 13(24)2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34944852

RESUMEN

Although many breast cancer therapies show initial success in the treatment of the primary tumour, they often fail to eliminate a sub-population of cells known as cancer stem cells (CSCs). These cells are recognised for their self-renewal properties and for their capacity for differentiation often leading to chemo/radio-resistance. The antiviral drug Efavirenz has been shown to be effective in eliminating triple-negative breast cancer cells, and here we examine its effect on breast CSCs. The effects of Efavirenz on CSCs for several breast cancer cell lines were investigated by examining cellular changes upon drug treatment, including CSC numbers, morphology, RNA/microRNA expression and levels of epithelial/mesenchymal CSC subtypes. Efavirenz treatment resulted in a decrease in the size and number of tumorspheres and a reduction in epithelial-type CSC levels, but an increase in mesenchymal-type CSCs. Efavirenz caused upregulation of several CSC-related genes as well as miR-21, a CSC marker and miR-182, a CSC suppressor gene. We conclude that Efavirenz alters the phenotype and expression of key genes in breast CSCs, which has important potential therapeutic implications.

8.
Cell Calcium ; 96: 102403, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33813182

RESUMEN

The phosphoprotein AHNAK is a large, ubiquitously expressed scaffolding protein involved in mediating a host of protein-protein interactions. This enables AHNAK to participate in various multi-protein complexes thereby orchestrating a range of diverse biological processes, including tumour suppression, immune regulation and cell architecture maintenance. A less studied but nonetheless equally important function occurs in calcium homeostasis. It does so by largely interacting with the L-type voltage-gated calcium channel (LVGCC) present in the plasma membrane of excitable cells such as muscles and neurons. Several studies have characterized the underlying basis of AHNAK's functional role in calcium channel modulation, which has led to a greater understanding of this cellular process and its associated pathologies. In this article we review and examine recent advances relating to the physiological aspects of AHNAK in calcium regulation. Specifically, we will provide a broad overview of AHNAK including its structural makeup and its interaction with several isoforms of LVGCC, and how these molecular interactions regulate calcium modulation across various tissues and their implication in muscle and neuronal function.


Asunto(s)
Calcio/metabolismo , Homeostasis/fisiología , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Animales , Sitios de Unión/fisiología , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/metabolismo , Humanos
9.
Nucleic Acids Res ; 49(4): 2255-2265, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33533913

RESUMEN

Interferon regulatory factor 4 (IRF4) is a key transcription factor (TF) in the regulation of immune cells, including B and T cells. It acts by binding DNA as both a homodimer and, in conjunction with other TFs, as a heterodimer. The choice of homo and heterodimeric/ DNA interactions is a critical aspect in the control of the transcriptional program and cell fate outcome. To characterize the nature of this interaction in the homodimeric complex, we have determined the crystal structure of the IRF4/ISRE homodimeric complex. We show that the complex formation is aided by a substantial DNA deformation with co-operative binding achieved exclusively through protein-DNA contact. This markedly contrasts with the heterodimeric form where DNA bound IRF4 is shown to physically interact with PU.1 TF to engage EICE1. We also show that the hotspot residues (Arg98, Cys99 and Asn102) contact both consensus and non-consensus sequences with the L1 loop exhibiting marked flexibility. Additionally, we identified that IRF4L116R, a mutant associated with chronic lymphocytic leukemia, binds more robustly to DNA thereby providing a rationale for the observed gain of function. Together, we demonstrate key structural differences between IRF4 homo and heterodimeric complexes, thereby providing molecular insights into IRF4-mediated transcriptional regulation.


Asunto(s)
ADN/química , Factores Reguladores del Interferón/química , ADN/metabolismo , Dimerización , Mutación con Ganancia de Función , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Leucemia Linfocítica Crónica de Células B/genética , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , Proteínas Proto-Oncogénicas/química , Transactivadores/química
10.
Carcinogenesis ; 42(6): 853-863, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-33564842

RESUMEN

Inflammatory bowel disease (IBD) is characterized by multiple alterations in cytokine expression and is a risk factor for colon cancer. The Omega class glutathione transferase GSTO1-1 regulates the release of the pro-inflammatory cytokines interleukin 1ß (IL-1ß) and interleukin 18 (IL-18) by deglutathionylating NEK7 in the NLRP3 inflammasome. When treated with azoxymethane and dextran sodium sulphate (AOM/DSS) as a model of IBD, Gsto1-/- mice were highly sensitive to colitis and showed a significant increase in the size and number of colon tumours compared with wild-type (WT) mice. Gsto1-/- mice treated with AOM/DSS had significantly lower serum IL-1ß and IL-18 levels as well as significantly decreased interferon (IFN)-γ, decreased pSTAT1 and increased pSTAT3 levels in the distal colon compared with similarly treated WT mice. Histologically, AOM/DSS treated Gsto1-/- mice showed increased active chronic inflammation with macrophage infiltration, epithelial dysplasia and invasive adenocarcinoma compared with AOM/DSS treated WT mice. Thus, this study shows that GSTO1-1 regulates IL-1ß and IL-18 activation and protects against colorectal cancer formation in the AOM/DSS model of IBD. The data suggest that while GSTO1-1 is a new target for the regulation of the NLRP3 inflammasome-associated cytokines IL-1ß and IL-18 by small molecule inhibitors, there is a possibility that anti-inflammatory drugs targeting these cytokines may potentiate colon cancer in some situations.


Asunto(s)
Azoximetano/toxicidad , Proteínas Portadoras/fisiología , Colitis/complicaciones , Neoplasias Colorrectales/prevención & control , Glutatión Transferasa/fisiología , Inflamación/prevención & control , Interleucina-18/sangre , Interleucina-1beta/sangre , Animales , Carcinógenos/toxicidad , Colitis/inducido químicamente , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Sulfato de Dextran/toxicidad , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Biophys Rev ; 13(6): 1219-1227, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35059038

RESUMEN

Interferon regulatory factor 4 (IRF4) is a lymphoid transcription factor and a key regulator in the development of various immune cells, including T and B cells. It is well-known that IRF4 controls numerous decision-making processes relating to B cell development, including differentiation, maturation, and signalling. Consequently, genetic alterations that affect the functional aspects of IRF4 can result in clonal transformation and have been identified in various lymphoid malignancies. Over the last decades, a series of studies have demonstrated the critical cellular and structural basis underpinning IRF4-mediated B cell development and associated malignancies. In this review, we will briefly summarise the recent advances in understanding IRF4-mediated B cell development and related malignancies, with a particular focus on the molecular aspects that govern these processes.

12.
Life Sci ; 260: 118234, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-32791148

RESUMEN

AIMS: Our aim was to characterise the actions of novel BIT compounds with structures based on peptides and toxins that bind to significant regulatory sites on ryanodine receptor (RyR) Ca2+ release channels. RyRs, located in sarcoplasmic reticulum (SR) Ca2+ store membranes of striated muscle, are essential for muscle contraction. Although severe sometimes-deadly myopathies occur when the channels become hyperactive following genetic or acquired changes, specific inhibitors of RyRs are rare. MAIN METHODS: The effect of BIT compounds was determined by spectrophotometric analysis of Ca2+ release from isolated SR vesicles, analysis of single RyR channel activity in artificial lipid bilayers and contraction of intact and skinned skeletal muscle fibres. KEY FINDINGS: The inhibitory compounds reduced: (a) Ca2+ release from SR vesicles with IC50s of 1.1-2.5 µM, competing with activation by parent peptides and toxins; (b) single RyR ion channel activity with IC50s of 0.5-1.5 µM; (c) skinned fibre contraction. In contrast, activating BIT compounds increased Ca2+ release with an IC50 of 5.0 µM and channel activity with AC50s of 2 to 12 nM and enhanced skinned fibre contraction. Sub-conductance activity dominated channel activity with both inhibitors and activators. Effects of all compounds on skeletal and cardiac RyRs were similar and reversible. Competition experiments suggest that the BIT compounds bind to the regulatory helical domains of the RyRs that impact on channel gating mechanisms through long-range allosteric interactions. SIGNIFICANCE: The BIT compounds are strong modulators of RyR activity and provide structural templates for novel research tools and drugs to combat muscle disease.


Asunto(s)
Péptidos/farmacología , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos , Retículo Sarcoplasmático/química , Animales , Biomimética , Calcio/metabolismo , Contracción Muscular/efectos de los fármacos , Músculo Esquelético/ultraestructura , Miocardio/ultraestructura , Conejos , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Venenos de Escorpión , Ovinos
13.
J Med Chem ; 63(6): 2894-2914, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32105470

RESUMEN

Glutathione transferase omega-1 (GSTO1-1) is an enzyme whose function supports the activation of interleukin (IL)-1ß and IL-18 that are implicated in a variety of inflammatory disease states for which small-molecule inhibitors are sought. The potent reactivity of the active-site cysteine has resulted in reported inhibitors that act by covalent labeling. In this study, structure-activity relationship (SAR) elaboration of the reported GSTO1-1 inhibitor C1-27 was undertaken. Compounds were evaluated for inhibitory activity toward purified recombinant GSTO1-1 and for indicators of target engagement in cell-based assays. As covalent inhibitors, the kinact/KI values of selected compounds were determined, as well as in vivo pharmacokinetics analysis. Cocrystal structures of key novel compounds in complex with GSTO1-1 were also solved. This study represents the first application of a biochemical assay for GSTO1-1 to determine kinact/KI values for tested inhibitors and the most extensive set of cell-based data for a GSTO1-1 inhibitor SAR series reported to date. Our research culminated in the discovery of 25, which we propose as the preferred biochemical tool to interrogate cellular responses to GSTO1-1 inhibition.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glutatión Transferasa/antagonistas & inhibidores , Sulfonamidas/química , Sulfonamidas/farmacología , Animales , Desarrollo de Medicamentos , Glutatión Transferasa/química , Glutatión Transferasa/metabolismo , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Bencenosulfonamidas
14.
Cell Rep ; 29(1): 151-161.e5, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31577945

RESUMEN

The NLRP3 inflammasome is a cytosolic complex sensing phagocytosed material and various damage-associated molecular patterns, triggering production of the pro-inflammatory cytokines interleukin-1 beta (IL)-1ß and IL-18 and promoting pyroptosis. Here, we characterize glutathione transferase omega 1-1 (GSTO1-1), a constitutive deglutathionylating enzyme, as a regulator of the NLRP3 inflammasome. Using a small molecule inhibitor of GSTO1-1 termed C1-27, endogenous GSTO1-1 knockdown, and GSTO1-1-/- mice, we report that GSTO1-1 is involved in NLRP3 inflammasome activation. Mechanistically, GSTO1-1 deglutathionylates cysteine 253 in NIMA related kinase 7 (NEK7) to promote NLRP3 activation. We therefore identify GSTO1-1 as an NLRP3 inflammasome regulator, which has potential as a drug target to limit NLRP3-mediated inflammation.


Asunto(s)
Glutatión Transferasa/metabolismo , Inflamasomas/metabolismo , Quinasas Relacionadas con NIMA/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Citocinas/metabolismo , Células HEK293 , Humanos , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL
15.
F1000Res ; 72018.
Artículo en Inglés | MEDLINE | ID: mdl-30542613

RESUMEN

The ryanodine receptor calcium release channel is central to cytoplasmic Ca 2+ signalling in skeletal muscle, the heart, and many other tissues, including the central nervous system, lymphocytes, stomach, kidney, adrenal glands, ovaries, testes, thymus, and lungs. The ion channel protein is massive (more than 2.2 MDa) and has a structure that has defied detailed determination until recent developments in cryo-electron microscopy revealed much of its structure at near-atomic resolution. The availability of this high-resolution structure has provided the most significant advances in understanding the function of the ion channel in the past 30 years. We can now visualise the molecular environment of individual amino acid residues that form binding sites for essential modulators of ion channel function and determine its role in Ca 2+ signalling. Importantly, the structure has revealed the structural environment of the many deletions and point mutations that disrupt Ca 2+ signalling in skeletal and cardiac myopathies and neuropathies. The implications are of vital importance to our understanding of the molecular basis of the ion channel's function and for the design of therapies to counteract the effects of ryanodine receptor-associated disorders.


Asunto(s)
Señalización del Calcio/genética , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Animales , Sitios de Unión/genética , Microscopía por Crioelectrón , Humanos , Conformación Proteica , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/genética
16.
ChemMedChem ; 13(18): 1957-1971, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30058107

RESUMEN

Ryanodine receptor (RyR) Ca2+ -release channels are essential for contraction in skeletal and cardiac muscle and are prime targets for modification of contraction in disorders that affect either the skeletal or heart musculature. We designed and synthesized a number of compounds with structures based on a naturally occurring peptide (A peptides) that modifies the activity of RyRs. In total, 34 compounds belonging to eight different classes were prepared. The compounds were screened for their ability to enhance Ca2+ release from isolated cardiac sarcoplasmic reticulum (SR) vesicles, with 25 displaying enhanced Ca2+ release. Competition studies with the parent peptides indicated that the synthetic compounds act at a competing site. The activity of the most effective of the compounds, BIT 180, was further explored using Ca2+ release from skeletal SR vesicles and contraction in intact skeletal muscle fibers. The compounds did not alter tension in intact fibers, indicating that (as expected) they are not membrane permeable, but importantly, that they are not toxic to the intact cells. Proof in principal that the compounds would be effective in intact muscle fibers if rendered membrane permeable was obtained with a structurally related membrane-permeable scorpion toxin (imperatoxin A), which was found to enhance contraction.


Asunto(s)
Péptidos/farmacología , Peptidomiméticos/farmacología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Estructura Molecular , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Péptidos/síntesis química , Péptidos/química , Peptidomiméticos/síntesis química , Peptidomiméticos/química , Conejos , Ovinos , Relación Estructura-Actividad
17.
J Med Chem ; 61(17): 7448-7470, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-29652143

RESUMEN

Early stage drug discovery reporting on relatively new or difficult targets is often associated with insufficient hit triage. Literature reviews of such targets seldom delve into the detail required to critically analyze the associated screening hits reported. Here we take the enzyme glutathione transferase omega-1 (GSTO1-1) as an example of a relatively difficult target and review the associated literature involving small-molecule inhibitors. As part of this process we deliberately pay closer-than-usual attention to assay interference and hit quality aspects. We believe this Perspective will be a useful guide for future development of GSTO1-1 inhibitors, as well serving as a template for future review formats of new or difficult targets.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glutatión Transferasa/antagonistas & inhibidores , Glutatión Transferasa/química , Diseño de Fármacos , Descubrimiento de Drogas , Polarización de Fluorescencia/métodos , Glutatión Transferasa/metabolismo , Humanos
18.
Biomol NMR Assign ; 12(2): 253-257, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29594929

RESUMEN

Ahnak is a ~ 700 kDa polypeptide that was originally identified as a tumour-related nuclear phosphoprotein, but later recognized to play a variety of diverse physiological roles related to cell architecture and migration. A critical function of Ahnak is modulation of Ca2+ signaling in cardiomyocytes by interacting with the ß subunit of the L-type Ca2+ channel (CaV1.2). Previous studies have identified the C-terminal region of Ahnak, designated as P3 and P4 domains, as a key mediator of its functional activity. We report here the nearly complete 1H, 13C and 15N backbone NMR chemical shift assignments of the 11 kDa C-terminal P4 domain of Ahnak. This study lays the foundations for future investigations of functional dynamics, structure determination and interaction site mapping of the CaV1.2-Ahnak complex.


Asunto(s)
Proteínas de la Membrana/química , Resonancia Magnética Nuclear Biomolecular , Sitios de Unión , Canales de Calcio Tipo L/metabolismo , Proteínas de la Membrana/metabolismo , Dominios Proteicos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo
19.
Am J Physiol Cell Physiol ; 314(3): C323-C333, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212769

RESUMEN

Malignant hyperthermia (MH) susceptibility has been recently linked to a novel variant of ß1a subunit of the dihydropyridine receptor (DHPR), a channel essential for Ca2+ regulation in skeletal muscle. Here we evaluate the effect of the mutant variant V156A on the structure/function of DHPR ß1a subunit and assess its role on Ca2+ metabolism of cultured myotubes. Using differential scanning fluorimetry, we show that mutation V156A causes a significant reduction in thermal stability of the Src homology 3/guanylate kinase core domain of ß1a subunit. Expression of the variant subunit in ß1-null mouse myotubes resulted in increased sensitivity to caffeine stimulation. Whole cell patch-clamp analysis of ß1a-V156A-expressing myotubes revealed a -2 mV shift in voltage dependence of channel activation, but no changes in Ca2+ conductance, current kinetics, or sarcoplasmic reticulum Ca2+ load were observed. Measurement of resting free Ca2+ and Na+ concentrations shows that both cations were significantly elevated in ß1a-V156A-expressing myotubes and that these changes were linked to increased rates of plasmalemmal Ca2+ entry through Na+/Ca2+ exchanger and/or transient receptor potential canonical channels. Overall, our data show that mutant variant V156A results in instability of protein subdomains of ß1a subunit leading to a phenotype of Ca2+ dysregulation that partly resembles that of other MH-linked mutations of DHPR α1S subunit. These data prove that homozygous expression of variant ß1a-V156A has the potential to be a pathological variant, although it may require other gene defects to cause a full MH phenotype.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Hipertermia Maligna/metabolismo , Mioblastos/metabolismo , Animales , Cafeína/farmacología , Calcio/metabolismo , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/genética , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Homocigoto , Humanos , Cinética , Hipertermia Maligna/genética , Hipertermia Maligna/fisiopatología , Ratones Noqueados , Mutación , Mioblastos/efectos de los fármacos , Dominios Proteicos , Estabilidad Proteica , Relación Estructura-Actividad
20.
Sci Rep ; 7(1): 17832, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29259211

RESUMEN

Glutathione transferase Omega 1 (GSTO1-1) is an atypical GST reported to play a pro-inflammatory role in response to LPS. Here we show that genetic knockout of Gsto1 alters the response of mice to three distinct inflammatory disease models. GSTO1-1 deficiency ameliorates the inflammatory response stimulated by LPS and attenuates the inflammatory impact of a high fat diet on glucose tolerance and insulin resistance. In contrast, GSTO1-1 deficient mice show a more severe inflammatory response and increased escape of bacteria from the colon into the lymphatic system in a dextran sodium sulfate mediated model of inflammatory bowel disease. These responses are similar to those of TLR4 and MyD88 deficient mice in these models and confirm that GSTO1-1 is critical for a TLR4-like pro-inflammatory response in vivo. In wild-type mice, we show that a small molecule inhibitor that covalently binds in the active site of GSTO1-1 can be used to ameliorate the inflammatory response to LPS. Our findings demonstrate the potential therapeutic utility of GSTO1-1 inhibitors in the modulation of inflammation and suggest their possible application in the treatment of a range of inflammatory conditions.


Asunto(s)
Proteínas Portadoras/metabolismo , Colitis/metabolismo , Glutatión Transferasa/metabolismo , Inflamación/metabolismo , Obesidad/metabolismo , Animales , Proteínas Portadoras/genética , Colitis/tratamiento farmacológico , Colitis/genética , Glutatión Transferasa/genética , Inflamación/tratamiento farmacológico , Inflamación/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/genética , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...