Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Endocrinol Diabetes Nutr (Engl Ed) ; 70(10): 619-627, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38065627

RESUMEN

BACKGROUND AND AIMS: Neuregulin 1 (NRG-1) is one of the members of the epidermal growth factors proteins. The present study provides novel insights into the relationship between serum levels of NRG-1 and insulin resistance, subclinical atherosclerosis and cardiac dysfunction that occur in type 2 diabetes (T2D). METHODS: The study included 50 patients with T2D and 40 healthy age- and gender-matched controls. Serum NRG-1 was measured using ELISA. Glycemic parameters, lipid profile and insulin resistance were assessed. Trans-thoracic echocardiography and carotid intima media thickness (CIMT) were studied for all study subjects. RESULTS: T2D patients had significantly lower serum NRG-1 levels than controls. Serum NRG-1 was negatively correlated with age, fasting blood glucose, HbA1c, insulin resistance, blood urea, serum creatinine and LDL-C, and positively correlated with HDL-C, eGFR and CIMT. Regarding echocardiographic variables, serum NRG-1 was found to correlate positively with left ventricular global longitudinal strain and negatively with E/Ea ratio. NRG-1 was found to predict subclinical atherosclerosis in type 2 diabetes patients at a cut-off value<108.5pg/ml with 78% sensitivity and 80% specificity. CONCLUSIONS: A robust relationship was found between serum NRG-1 levels and hyperglycemia, insulin resistance, subclinical atherosclerosis, and cardiac dysfunction in patients with type 2 diabetes. These results shed light on a possible role of NRG-1 as a potential noninvasive biomarker for detection of cardiometabolic risk in T2D.


Asunto(s)
Aterosclerosis , Diabetes Mellitus Tipo 2 , Cardiopatías , Resistencia a la Insulina , Neurregulina-1 , Humanos , Aterosclerosis/etiología , Aterosclerosis/diagnóstico , Grosor Intima-Media Carotídeo , Diabetes Mellitus Tipo 2/complicaciones , Neurregulina-1/sangre , Neurregulina-1/química , Neurregulina-1/metabolismo , Factores de Riesgo , Función Ventricular
2.
Nature ; 600(7888): 339-343, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34759323

RESUMEN

Human epidermal growth factor receptor 2 (HER2) and HER3 form a potent pro-oncogenic heterocomplex1-3 upon binding of growth factor neuregulin-1ß (NRG1ß). The mechanism by which HER2 and HER3 interact remains unknown in the absence of any structures of the complex. Here we isolated the NRG1ß-bound near full-length HER2-HER3 dimer and, using cryo-electron microscopy, reconstructed the extracellulardomain module, revealing unexpected dynamics at the HER2-HER3 dimerization interface. We show that the dimerization arm of NRG1ß-bound HER3 is unresolved because the apo HER2 monomer does not undergo a ligand-induced conformational change needed to establish a HER3 dimerization arm-binding pocket. In a structure of the oncogenic extracellular domain mutant HER2(S310F), we observe a compensatory interaction with the HER3 dimerization arm that stabilizes the dimerization interface. Both HER2-HER3 and HER2(S310F)-HER3 retain the capacity to bind to the HER2-directed therapeutic antibody trastuzumab, but the mutant complex does not bind to pertuzumab. Our structure of the HER2(S310F)-HER3-NRG1ß-trastuzumab Fab complex reveals that the receptor dimer undergoes a conformational change to accommodate trastuzumab. Thus, similar to oncogenic mutations, therapeutic agents exploit the intrinsic dynamics of the HER2-HER3 heterodimer. The unique features of a singly liganded HER2-HER3 heterodimer underscore the allosteric sensing of ligand occupancy by the dimerization interface and explain why extracellular domains of HER2 do not homo-associate via a canonical active dimer interface.


Asunto(s)
Microscopía por Crioelectrón , Neurregulina-1/química , Multimerización de Proteína , Receptor ErbB-2/química , Receptor ErbB-3/química , Regulación Alostérica , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/ultraestructura , Sitios de Unión , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Modelos Moleculares , Mutación , Neurregulina-1/ultraestructura , Oncogenes/genética , Estabilidad Proteica , Receptor ErbB-2/ultraestructura , Receptor ErbB-3/ultraestructura , Trastuzumab/química , Trastuzumab/ultraestructura
3.
Nat Commun ; 12(1): 3335, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099661

RESUMEN

Plants utilise intracellular nucleotide-binding, leucine-rich repeat (NLR) immune receptors to detect pathogen effectors and activate local and systemic defence. NRG1 and ADR1 "helper" NLRs (RNLs) cooperate with enhanced disease susceptibility 1 (EDS1), senescence-associated gene 101 (SAG101) and phytoalexin-deficient 4 (PAD4) lipase-like proteins to mediate signalling from TIR domain NLR receptors (TNLs). The mechanism of RNL/EDS1 family protein cooperation is not understood. Here, we present genetic and molecular evidence for exclusive EDS1/SAG101/NRG1 and EDS1/PAD4/ADR1 co-functions in TNL immunity. Using immunoprecipitation and mass spectrometry, we show effector recognition-dependent interaction of NRG1 with EDS1 and SAG101, but not PAD4. An EDS1-SAG101 complex interacts with NRG1, and EDS1-PAD4 with ADR1, in an immune-activated state. NRG1 requires an intact nucleotide-binding P-loop motif, and EDS1 a functional EP domain and its partner SAG101, for induced association and immunity. Thus, two distinct modules (NRG1/EDS1/SAG101 and ADR1/EDS1/PAD4) mediate TNL receptor defence signalling.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Proteínas de Unión al ADN/metabolismo , Neurregulina-1/metabolismo , Inmunidad de la Planta/fisiología , Receptores Inmunológicos/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Arabidopsis/microbiología , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Hidrolasas de Éster Carboxílico/química , Hidrolasas de Éster Carboxílico/genética , Muerte Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Inmunidad Innata , Neurregulina-1/química , Neurregulina-1/genética , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/microbiología , Inmunidad de la Planta/genética , Plantas Modificadas Genéticamente , Dominios Proteicos , Pseudomonas syringae , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Transducción de Señal , Nicotiana/genética , Nicotiana/metabolismo
4.
Breast Cancer Res ; 23(1): 3, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33413557

RESUMEN

BACKGROUND: NRG1 gene fusions may be clinically actionable, since cancers carrying the fusion transcripts can be sensitive to tyrosine kinase inhibitors. The NRG1 gene encodes ligands for the HER2(ERBB2)-ERBB3 heterodimeric receptor tyrosine kinase, and the gene fusions are thought to lead to autocrine stimulation of the receptor. The NRG1 fusion expressed in the breast cancer cell line MDA-MB-175 serves as a model example of such fusions, showing the proposed autocrine loop and exceptional drug sensitivity. However, its structure has not been properly characterised, its oncogenic activity has not been fully explained, and there is limited data on such fusions in breast cancer. METHODS: We analysed genomic rearrangements and transcripts of NRG1 in MDA-MB-175 and a panel of 571 breast cancers. RESULTS: We found that the MDA-MB-175 fusion-originally reported as a DOC4(TENM4)-NRG1 fusion, lacking the cytoplasmic tail of NRG1-is in reality a double fusion, PPP6R3-TENM4-NRG1, producing multiple transcripts, some of which include the cytoplasmic tail. We hypothesise that many NRG1 fusions may be oncogenic not for lacking the cytoplasmic domain but because they do not encode NRG1's nuclear-localised form. The fusion in MDA-MB-175 is the result of a very complex genomic rearrangement, which we partially characterised, that creates additional expressed gene fusions, RSF1-TENM4, TPCN2-RSF1, and MRPL48-GAB2. We searched for NRG1 rearrangements in 571 breast cancers subjected to genome sequencing and transcriptome sequencing and found four cases (0.7%) with fusions, WRN-NRG1, FAM91A1-NRG1, ARHGEF39-NRG1, and ZNF704-NRG1, all splicing into NRG1 at the same exon as in MDA-MB-175. However, the WRN-NRG1 and ARHGEF39-NRG1 fusions were out of frame. We identified rearrangements of NRG1 in many more (8% of) cases that seemed more likely to inactivate than to create activating fusions, or whose outcome could not be predicted because they were complex, or both. This is not surprising because NRG1 can be pro-apoptotic and is inactivated in some breast cancers. CONCLUSIONS: Our results highlight the complexity of rearrangements of NRG1 in breast cancers and confirm that some do not activate but inactivate. Careful interpretation of NRG1 rearrangements will therefore be necessary for appropriate patient management.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Mama/genética , Neurregulina-1/genética , Proteínas de Fusión Oncogénica/genética , Empalme Alternativo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Reordenamiento Génico , Sitios Genéticos , Humanos , Neurregulina-1/química , Neurregulina-1/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Transducción de Señal , Translocación Genética
5.
Sci Rep ; 10(1): 17257, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-33057063

RESUMEN

Neuregulin protein 1 (NRG1) is a large (> 60-amino-acid) natural peptide ligand for the ErbB protein family members HER3 and HER4. We developed an agonistic antibody modality, termed antibody ligand mimetics (ALM), by incorporating complex ligand agonists such as NRG1 into an antibody scaffold. We optimized the linker and ligand length to achieve native ligand activity in HEK293 cells and cardiomyocytes derived from induced pluripotent stem cells (iPSCs) and used a monomeric Fc-ligand fusion platform to steer the ligand specificity toward HER4-dominant agonism. With the help of selectivity engineering, these enhanced ALM molecules can provide an antibody scaffold with increased receptor specificity and the potential to greatly improve the pharmacokinetics, stability, and downstream developability profiles from the natural ligand approach. This ligand mimetic design and optimization approach can be expanded to apply to other cardiovascular disease targets and emerging therapeutic areas, providing differentiated drug molecules with increased specificity and extended half-life.


Asunto(s)
Anticuerpos Monoclonales/química , Neurregulina-1/química , Receptor ErbB-4/agonistas , Anticuerpos Monoclonales/metabolismo , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/química , Células Madre Pluripotentes Inducidas/metabolismo , Cinética , Ligandos , Miocitos Cardíacos/química , Miocitos Cardíacos/metabolismo , Neurregulina-1/metabolismo , Unión Proteica , Receptor ErbB-4/metabolismo , Transducción de Señal
6.
Pharm Res ; 37(4): 75, 2020 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-32232574

RESUMEN

PURPOSE: Doxorubicin (Dox) being a hydrophobic drug needs a unique carrier for the effective encapsulation with uniformity in the aqueous dispersion, cell culture media and the biological-fluids that may efficiently target its release at the tumor site. METHODS: Circular DNA-nanotechnology was employed to synthesize DNA Nano-threads (DNA-NTs) by polymerization of triangular DNA-tiles. It involved circularizing a linear single-stranded scaffold strand to make sturdier and rigid triangles. DNA-NTs were characterized by the AFM and Native-PAGE tests. Dox binding and loading to the Neuregulin1 (NRG1) functionalized DNA based nano-threads (NF-DBNs) was estimated by the UV-shift analysis. The biocompatibility of the blank NRG-1/DNA-NTs and enhanced cytotoxicity of the NF-DBNs was assessed by the MTT assay. Cell proliferation/apoptosis was analyzed through the Flow-cytometry experiment. Cell-surface binding and the cell-internalization of the NF-DBNs was captured by the double-photon confocal microscopy (DPCM). RESULTS: The AFM images revealed uniform DNA-NTs with the diameter 30 to 80 nm and length 400 to 800 nm. PAGE native gel was used for the further confirmation of the successful assembly of the strands to synthesize DNA-NTs that gave one sharp band with the decreased electrophoretic mobility down the gel. MTT assay showed that blank DNA-NTs were biocompatible to the cells with less cytotoxicity even at elevated concentrations with most of the cells (94%) remaining alive compared to the dose-dependent enhanced cytotoxicity of NF-DBNs further evidenced by the Flow-cytometry analysis. CONCLUSION: Uniform and stiffer DNA-NTs for the potential applications in targeted drug delivery was achieved through circular DNA scaffolding.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , ADN Circular/química , Doxorrubicina/administración & dosificación , Portadores de Fármacos/síntesis química , Resistencia a Antineoplásicos/efectos de los fármacos , Nanopartículas/química , Receptor ErbB-3/metabolismo , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Portadores de Fármacos/química , Humanos , Ligandos , Microscopía de Fuerza Atómica , Microscopía Confocal , Neurregulina-1/química , Propiedades de Superficie
7.
J Alzheimers Dis ; 74(2): 535-544, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32065797

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) includes a large spectrum of neurodegenerative disorders. OBJECTIVE: To identify the relationship of ErbB4 mutation and ALS/FTD. METHODS: Here, we report an atypical case of frontal variant behavioral abnormalities at the initial stage, a stable plateau stage of 5 years, and paralysis involving both upper and lower motor neurons followed by progressive cognitive dysfunction at the advanced stage. The clinical findings suggested a diagnosis of ALS/FTD, and genetic testing revealed erb-b2 receptor tyrosine kinase 4 (ErbB4) heterozygous mutation (c.2136 T>G, p.I712M), identified in an ALS pedigree previously. We modeled mutant ErbB4 protein through the SWISS-MODEL Server, and speculated on the structural change caused by the mutation. We also identified that ErbB4 (I712M) mutation led to reduced auto-phosphorylation of ErbB4 upon neuregulin-1 (NRG1) stimulation. RESULTS: A functional analysis of ErbB4 mutation demonstrated an obviously decreased auto-phosphorylation of ErbB4 involving in the pathogenesis of ALS/FTD. CONCLUSION: We firstly found ErbB4 mutation to be identified in ALS/FTD.


Asunto(s)
Esclerosis Amiotrófica Lateral/diagnóstico por imagen , Esclerosis Amiotrófica Lateral/genética , Demencia Frontotemporal/diagnóstico por imagen , Demencia Frontotemporal/genética , Mutación/fisiología , Neurregulina-1/genética , Receptor ErbB-4/genética , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/metabolismo , Femenino , Demencia Frontotemporal/metabolismo , Humanos , Persona de Mediana Edad , Neurregulina-1/química , Neurregulina-1/metabolismo , Linaje , Estructura Secundaria de Proteína , Receptor ErbB-4/química , Receptor ErbB-4/metabolismo , Transducción de Señal/fisiología
8.
J Pharm Biomed Anal ; 182: 113133, 2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32004770

RESUMEN

DNA based nano-carriers synthesized from short circular scaffolds (circular DNA nanotechnology) attains stiffer topology for ligand functionalization (neuregulin-1/NRG-1 ligand) and biological applications (targeted drug delivery). Daunorubicin (DR) is a hydrophobic chemical that requires robust vectors to efficiently encapsulate and avoid its free dispersion in water, biological media and cell culture. Here we design DNA nanospindels (DNA-NS) to efficiently load DR and target the (highly expressed) HER2/neu receptors on the plasma membrane of drug-resistant MCF-7 (breast cancer) cells. DNA-NS were synthesized by polymerizing the DNA-triangles (utilizing 84-nt short circular scaffold strand) into larger DNA nano-ribbons characterized by the native-PAGE testing. AFM results revealed the spinning of DNA nanoribbons on its (own) axis because of the intrinsic curvature of the DNA double helix resulting in the formation of the firm and twisted DNA-NS with the diameter (50-70 nm) and length (0.5-4 µm). DA loading onto DNA-NS was confirmed by the UV shift analysis. The MTT results with the blank DNA-NS evidenced its biocompatibility (remained value of 93%) compared to the decreased viability of the MCF-7 cells after treatment with DNA-NS (DR loaded). These findings were further supported by the analysis of cell proliferation/apoptosis through flow cytometry showing 64% apoptosis after treating with the DR loaded DNA-NS. Hence, through the short circular DNA nanotechnology, we have achieved a stiffer, uniform, and biocompatible DNA-NS for applications in the targeted therapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Daunorrubicina/administración & dosificación , Nanoestructuras , Receptor ErbB-2/metabolismo , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , ADN/química , Daunorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Femenino , Citometría de Flujo , Humanos , Células MCF-7 , Neurregulina-1/química , Tamaño de la Partícula
9.
Nucleic Acids Res ; 47(21): 11020-11043, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31617560

RESUMEN

RNA interference represents a potent intervention for cancer treatment but requires a robust delivery agent for transporting gene-modulating molecules, such as small interfering RNAs (siRNAs). Although numerous molecular approaches for siRNA delivery are adequate in vitro, delivery to therapeutic targets in vivo is limited by payload integrity, cell targeting, efficient cell uptake, and membrane penetration. We constructed nonviral biomaterials to transport small nucleic acids to cell targets, including tumor cells, on the basis of the self-assembling and cell-penetrating activities of the adenovirus capsid penton base. Our recombinant penton base chimera contains polypeptide domains designed for noncovalent assembly with anionic molecules and tumor homing. Here, structural modeling, molecular dynamics simulations, and functional assays suggest that it forms pentameric units resembling viral capsomeres that assemble into larger capsid-like structures when combined with siRNA cargo. Pentamerization forms a barrel lined with charged residues mediating pH-responsive dissociation and exposing masked domains, providing insight on the endosomolytic mechanism. The therapeutic impact was examined on tumors expressing high levels of HER3/ErbB3 that are resistant to clinical inhibitors. Our findings suggest that our construct may utilize ligand mimicry to avoid host attack and target the siRNA to HER3+ tumors by forming multivalent capsid-like structures.


Asunto(s)
Portadores de Fármacos/uso terapéutico , Nanopartículas/uso terapéutico , ARN Interferente Pequeño/farmacología , Receptor ErbB-3/antagonistas & inhibidores , Proteínas Recombinantes/uso terapéutico , Animales , Proteínas de la Cápside/química , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos BALB C , Neurregulina-1/química , Interferencia de ARN
10.
Mol Neurobiol ; 56(12): 8345-8363, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31240601

RESUMEN

Unprocessed pro-neuregulin 2 (pro-NRG2) accumulates on neuronal cell bodies at junctions between the endoplasmic reticulum and plasma membrane (ER-PM junctions). NMDA receptors (NMDARs) trigger NRG2 ectodomain shedding from these sites followed by activation of ErbB4 receptor tyrosine kinases, and ErbB4 signaling cell-autonomously downregulates intrinsic excitability of GABAergic interneurons by reducing voltage-gated sodium channel currents. NMDARs also promote dispersal of Kv2.1 clusters from ER-PM junctions and cause a hyperpolarizing shift in its voltage-dependent channel activation, suggesting that NRG2/ErbB4 and Kv2.1 work together to regulate intrinsic interneuron excitability in an activity-dependent manner. Here we explored the cellular processes underlying NMDAR-dependent NRG2 shedding in cultured rat hippocampal neurons. We report that NMDARs control shedding by two separate but converging mechanisms. First, NMDA treatment disrupts binding of pro-NRG2 to ER-PM junctions by post-translationally modifying conserved Ser/Thr residues in its intracellular domain. Second, using a mutant NRG2 protein that cannot be modified at these residues and that fails to accumulate at ER-PM junctions, we demonstrate that NMDARs also directly promote NRG2 shedding by ADAM-type metalloproteinases. Using pharmacological and shRNA-mediated knockdown, and metalloproteinase overexpression, we unexpectedly find that ADAM10, but not ADAM17/TACE, is the major NRG2 sheddase acting downstream of NMDAR activation. Together, these findings reveal how NMDARs exert tight control over the NRG2/ErbB4 signaling pathway, and suggest that NRG2 and Kv2.1 are co-regulated components of a shared pathway that responds to elevated extracellular glutamate levels.


Asunto(s)
Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Factores de Crecimiento Nervioso/química , Factores de Crecimiento Nervioso/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secuencia de Aminoácidos , Animales , Regulación hacia Abajo , Femenino , Hipocampo/metabolismo , Humanos , Masculino , Neurregulina-1/química , Neurregulina-1/metabolismo , Neuronas/metabolismo , Fosforilación , Unión Proteica , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Ratas Sprague-Dawley
11.
Biomed Mater ; 13(4): 045007, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29386409

RESUMEN

The use of artificial dermis as a skin substitute is a field of active study, as acellular dermal matrices from cadavers are susceptible to infection owing to their human origin. One such alternative dermal replacement scaffold, INSUREGRAF®, is derived primarily from extracellular matrix proteins such as collagen and elastin and has been clinically used to treat severe skin wounds such as burns. This scaffold has proven to be useful to minimize wound contraction and scar formation owing to its biocompatibility, interconnected pore structure, sufficient biodegradability, and suitable mechanical properties. However, INSUREGRAF® does not provide scar-free wound healing in cases of severe skin damage such as full-thickness (FT) excision. Considering that the efficient recruitment of fibroblasts and keratinocytes into a wound site represents a critical step in the regeneration of damaged skin, we attempted to enhance the efficiency for wound healing by fabricating growth factor-functionalized INSUREGRAF®. In particular, we utilized epidermal growth factor (EGF) and an EGF family member, neuregulin-1 (NRG1), not previously studied in the context of wound healing, whose cellular role is to promote proliferation and migration in fibroblasts and keratinocytes. Both artificial dermis-growth factor combinations led to efficient recruitment of fibroblasts and keratinocytes into a wound site during the early steps of skin regeneration. Notably, EGF- or NRG1-functionalized INSUREGRAF® induced rapid proliferation of skin cells in an ERK pathway-dependent manner and exhibited efficient wound healing in a Sprague-Dawley rat FT excision and grafting model. These results provide the foundation for expanding the use of growth factor-functionalized INSUREGRAF® to clinical application in cases of severe skin injury.


Asunto(s)
Factor de Crecimiento Epidérmico/química , Neurregulina-1/química , Regeneración , Piel Artificial , Ingeniería de Tejidos/instrumentación , Andamios del Tejido/química , Cicatrización de Heridas/efectos de los fármacos , Animales , Quemaduras/terapia , Movimiento Celular , Proliferación Celular , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Piel/patología , Estrés Mecánico , Ingeniería de Tejidos/métodos
12.
Eur J Pharmacol ; 796: 76-89, 2017 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-27993643

RESUMEN

Neuregulins are important growth factors involved in cardiac development and response to stress. Certain isoforms and fragments of neuregulin have been found to be cardioprotective. The effects of a full-length neuregulin-1ß isoform, glial growth factor 2 (GGF2; USAN/INN; also called cimaglermin) were investigated in vitro. Various dosing regimens were then evaluated for their effects on left ventricular (LV) function in rats with surgically-induced myocardial infarction. In vitro, GGF2 bound with high affinity to erythroblastic leukemia viral oncogene (ErbB) 4 receptors, potently promoted Akt phosphorylation, as well as reduced cell death following doxorubicin exposure in HL1 cells. Daily GGF2 treatment beginning 7-14 days after left anterior descending coronary artery ligation produced improvements in LV ejection fraction and other measures of LV function and morphology. The improvements in LV function (e.g. 10% point increase in absolute LV ejection fraction) with GGF2 were dose-dependent. LV performance was substantially improved when GGF2 treatment was delivered infrequently, despite a serum half-life of less than 2h and could be maintained for more than 10 months with treatment once weekly or once every 2 weeks. These studies confirm previous findings that GGF2 may improve contractile performance in the failing rat heart and that infrequent exposure to GGF2 may improve LV function and impact remodeling in the failing myocardium. GGF2 is now being developed for the treatment of heart failure in humans.


Asunto(s)
Ventrículos Cardíacos/efectos de los fármacos , Infarto del Miocardio/fisiopatología , Neurregulina-1/farmacología , Disfunción Ventricular/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , Citoprotección/efectos de los fármacos , Doxorrubicina/efectos adversos , Esquema de Medicación , Insuficiencia Cardíaca/complicaciones , Humanos , Ratones , Infarto del Miocardio/complicaciones , Neurregulina-1/administración & dosificación , Neurregulina-1/química , Neurregulina-1/metabolismo , Ratas , Receptor ErbB-4/metabolismo
13.
Sci Rep ; 6: 36133, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27808268

RESUMEN

With the application of advanced molecular cytogenetic techniques, the number of patients identified as having abnormal chromosome 8p has increased progressively. Individuals with terminal 8p deletion have been extensively described in previous studies. The manifestations usually include cardiac anomalies, developmental delay/mental retardation, craniofacial abnormalities, and multiple other minor anomalies. However, some patients with proximal deletion also presented with similar phenotypic features. Here we describe a female child with an 18.5-Mb deletion at 8p11.23-p22 that include the cardiac-associated loci NKX2-6 and NRG1. Further mutation screening of these two candidate genes in 143 atrial septal defect patients, two heterozygous mutations NKX2-6 (c.1A > T) and NRG1 (c.1652G > A) were identified. The mutations were described for the first time in patients with congenital heart disease (CHD). The c.1A > T NKX2-6 generated a protein truncated by 45 amino acids with a decreased level of mRNA expression, whereas the NRG1 mutation had no significant effect on protein functions. Our findings suggest that 8p21-8p12 may be another critical region for 8p-associated CHD, and some cardiac malformations might be due to NKX2-6 haploinsufficiency. This study also links the NKX2-6 mutation to ASD for the first time, providing novel insight into the molecular underpinning of this common form of CHD.


Asunto(s)
Cromosomas Humanos Par 8/genética , Estudios de Asociación Genética , Cardiopatías Congénitas/genética , Adulto , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Deleción Cromosómica , Análisis Citogenético , Análisis Mutacional de ADN , Femenino , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Humanos , Lactante , Recién Nacido , Mutación/genética , Neurregulina-1/química , Neurregulina-1/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fracciones Subcelulares/metabolismo
14.
Sci Rep ; 6: 37464, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27876763

RESUMEN

Release of cytokines, growth factors and other life-essential molecules from precursors by a-disintegrin-and-metalloproteases (ADAMs) is regulated with high substrate-specificity. We hypothesized that this is achieved by cleavage-regulatory intracellular-domain (ICD)-modifications of the precursors. We show here that cleavage-stimuli-induced specific ICD-modifications cause structural substrate changes that enhance ectodomain sensitivity of neuregulin-1 (NRG1; epidermal-growth-factor) or CD44 (receptor-tyrosine-kinase (RTK) co-receptor) to chymotrypsin/trypsin or soluble ADAM. This inside-out signal transfer required substrate homodimerization and was prevented by cleavage-inhibitory ICD-mutations. In chimeras, regulation could be conferred to a foreign ectodomain, suggesting a common higher-order structure. We predict that substrate-specific protease-accessibility-regulation controls release of numerous ADAM substrates.


Asunto(s)
Proteínas ADAM/genética , Receptores de Hialuranos/genética , Neurregulina-1/genética , Proteínas ADAM/química , Animales , Quimera/genética , Receptores de Hialuranos/química , Ratones , Mutación , Células 3T3 NIH , Neurregulina-1/química , Péptido Hidrolasas/química , Péptido Hidrolasas/genética , Dominios Proteicos/genética , Especificidad por Sustrato/genética
15.
Cell Rep ; 14(9): 2127-2141, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26923602

RESUMEN

Development of disease-modifying therapeutics is urgently needed for treating Alzheimer disease (AD). AD is characterized by toxic ß-amyloid (Aß) peptides produced by ß- and γ-secretase-mediated cleavage of the amyloid precursor protein (APP). ß-secretase inhibitors reduce Aß levels, but mechanism-based side effects arise because they also inhibit ß-cleavage of non-amyloid substrates like Neuregulin. We report that ß-secretase has a higher affinity for Neuregulin than it does for APP. Kinetic studies demonstrate that the affinities and catalytic efficiencies of ß-secretase are higher toward non-amyloid substrates than toward APP. We show that non-amyloid substrates are processed by ß-secretase in an endocytosis-independent manner. Exploiting this compartmentalization of substrates, we specifically target the endosomal ß-secretase by an endosomally targeted ß-secretase inhibitor, which blocked cleavage of APP but not non-amyloid substrates in many cell systems, including induced pluripotent stem cell (iPSC)-derived neurons. ß-secretase inhibitors can be designed to specifically inhibit the Alzheimer process, enhancing their potential as AD therapeutics without undesired side effects.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/metabolismo , Oligopéptidos/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/química , Animales , Ácido Aspártico Endopeptidasas/química , Ácido Aspártico Endopeptidasas/metabolismo , Células Cultivadas , Endocitosis , Endosomas/metabolismo , Aparato de Golgi/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Cinética , Ratones , Simulación de Dinámica Molecular , Neurregulina-1/química , Neurregulina-1/metabolismo , Oligopéptidos/química , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteolisis , Especificidad por Sustrato
16.
J Control Release ; 220(Pt A): 388-396, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26546270

RESUMEN

The growth factor neuregulin (NRG) is one of the most promising candidates in protein therapy as potential treatment for myocardial infarction (MI). In the last few years, biomaterial based delivery systems, such as polymeric microparticles (MPs) made of poly(lactic co glycolic acid) and polyethylene glycol (PLGA and PEG-PLGA MPs), have improved the efficacy of protein therapy in preclinical studies. However, no cardiac treatment based on MPs has yet been commercialized since this is a relatively new field and total characterization of polymeric MPs remains mandatory before they reach the clinical arena. Therefore, the objective of this study was to characterize the in vivo release, bioactivity and biodegradation of PLGA and PEG-PLGA MPs loaded with biotinylated NRG in a rat model of MI. The effect of PEGylation in the clearance of the particles from the cardiac tissue was also evaluated. Interestingly, MPs were detected in the cardiac tissue for up to 12 weeks after administration. In vivo release analysis showed that bNRG was released in a controlled manner throughout the twelve week study. Moreover, the biological cardiomyocyte receptor (ErbB4) for NRG was detected in its activated form only in those animals treated with bNRG loaded MPs. On the other hand, the PEGylation strategy was effective in diminishing phagocytosis of these MPs compared to noncoated MPs in the long term (12 weeks after injection). Taking all this together, we report new evidence in favor of the use of polymeric PLGA and PEG-PLGA MPs as delivery systems for treating MI, which could be soon included in clinical trials.


Asunto(s)
Fármacos Cardiovasculares/farmacocinética , Portadores de Fármacos , Ácido Láctico/química , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Neurregulina-1/farmacocinética , Poliésteres/química , Polietilenglicoles/química , Ácido Poliglicólico/química , Animales , Disponibilidad Biológica , Biotinilación , Fármacos Cardiovasculares/administración & dosificación , Fármacos Cardiovasculares/química , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Composición de Medicamentos , Estabilidad de Medicamentos , Femenino , Humanos , Macrófagos/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Miocardio/patología , Neurregulina-1/administración & dosificación , Neurregulina-1/química , Tamaño de la Partícula , Fagocitosis , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas Sprague-Dawley , Receptor ErbB-4/efectos de los fármacos , Receptor ErbB-4/metabolismo , Proteínas Recombinantes/farmacocinética
17.
Mol Cell Biol ; 35(19): 3381-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26217011

RESUMEN

Ectodomain cleavage by A-disintegrin and -metalloproteases (ADAMs) releases many important biologically active substrates and is therefore tightly controlled. Part of the regulation occurs on the level of the enzymes and affects their cell surface abundance and catalytic activity. ADAM-dependent proteolysis occurs outside the plasma membrane but is mostly controlled by intracellular signals. However, the intracellular domains (ICDs) of ADAM10 and -17 can be removed without consequences for induced cleavage, and so far it is unclear how intracellular signals address cleavage. We therefore explored whether substrates themselves could be chosen for proteolysis via ICD modification. We report here that CD44 (ADAM10 substrate), a receptor tyrosine kinase (RTK) coreceptor required for cellular migration, and pro-NRG1 (ADAM17 substrate), which releases the epidermal growth factor (EGF) ligand neuregulin required for axonal outgrowth and myelination, are indeed posttranslationally modified at their ICDs. Tetradecanoyl phorbol acetate (TPA)-induced CD44 cleavage requires dephosphorylation of ICD serine 291, while induced neuregulin release depends on the phosphorylation of several NRG1-ICD serines, in part mediated by protein kinase Cδ (PKCδ). Downregulation of PKCδ inhibits neuregulin release and reduces ex vivo neurite outgrowth and myelination of trigeminal ganglion explants. Our results suggest that specific selection among numerous substrates of a given ADAM is determined by ICD modification of the substrate.


Asunto(s)
Receptores de Hialuranos/metabolismo , Neurregulina-1/metabolismo , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Técnicas de Cocultivo , Células HEK293 , Humanos , Receptores de Hialuranos/química , Datos de Secuencia Molecular , Neurregulina-1/química , Neuritas/fisiología , Fosforilación , Proteína Quinasa C-delta/metabolismo , Proteolisis , Células de Schwann/metabolismo , Transducción de Señal , Ganglio del Trigémino/citología
18.
Sci Rep ; 5: 9834, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25940691

RESUMEN

Human pluripotent stem cells (hPSC) are used to study the early stages of human development in vitro and, increasingly due to somatic cell reprogramming, cellular and molecular mechanisms of disease. Cell culture medium is a critical factor for hPSC to maintain pluripotency and self-renewal. Numerous defined culture media have been empirically developed but never systematically optimized for culturing hPSC. We applied design of experiments (DOE), a powerful statistical tool, to improve the medium formulation for hPSC. Using pluripotency and cell growth as read-outs, we determined the optimal concentration of both basic fibroblast growth factor (bFGF) and neuregulin-1 beta 1 (NRG1ß1). The resulting formulation, named iDEAL, improved the maintenance and passage of hPSC in both normal and stressful conditions, and affected trimethylated histone 3 lysine 27 (H3K27me3) epigenetic status after genetic reprogramming. It also enhances efficient hPSC plating as single cells. Altogether, iDEAL potentially allows scalable and controllable hPSC culture routine in translational research. Our DOE strategy could also be applied to hPSC differentiation protocols, which often require numerous and complex cell culture media.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neurregulina-1/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Proyectos de Investigación , Algoritmos , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular/fisiología , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/metabolismo , Factor 2 de Crecimiento de Fibroblastos/química , Humanos , Modelos Estadísticos , Neurregulina-1/química
19.
J Biol Chem ; 290(28): 17041-54, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25925953

RESUMEN

Ectodomain shedding of transmembrane precursor proteins generates numerous life-essential molecules, such as epidermal growth factor receptor ligands. This cleavage not only releases the regulatory growth factor, but it is also the required first step for the subsequent processing by γ-secretase and the release of gene regulatory intracellular fragments. Signaling within the cell modifies the cytoplasmic tails of substrates, a step important in starting the specific and regulated cleavage of a large number of studied substrates. Ectodomain cleavage occurs, however, on the outside of the plasma membrane and is carried out by membrane-bound metalloproteases. How the intracellular domain modification communicates with the ectodomain of the substrate to allow for cleavage to occur is unknown. Here, we show that homodimerization of a cluster-of-differentiation-44 or of pro-neuregulin-1 monomers represents an essential pre-condition for their regulated ectodomain cleavage. Both substrates are associated with their respective metalloproteases under both basal or cleavage-stimulated conditions. These interactions only turn productive by specific intracellular signal-induced intracellular domain modifications of the substrates, which in turn regulate metalloprotease access to the substrates' ectodomain and cleavage. We propose that substrate intracellular domain modification induces a relative rotation or other positional change of the dimerization partners that allow metalloprotease cleavage in the extracellular space. Our findings fill an important gap in understanding substrate-specific inside-out signal transfer along cleaved transmembrane proteins and suggest that substrate dimerization (homo- or possibly heterodimerization) might represent a general principle in ectodomain shedding.


Asunto(s)
Receptores de Hialuranos/metabolismo , Neurregulina-1/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Proteína ADAM17 , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Bovinos , Línea Celular Tumoral , Membrana Celular/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Receptores de Hialuranos/química , Receptores de Hialuranos/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Células 3T3 NIH , Neurregulina-1/química , Neurregulina-1/genética , Multimerización de Proteína , Estructura Terciaria de Proteína , Proteolisis , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato
20.
Amino Acids ; 47(8): 1567-77, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25944317

RESUMEN

Axonally expressed neuregulin 1 (NRG1) type III is a transmembrane protein involved in various neurodevelopmental processes, including myelination and Schwann cell migration. NRG1 type III has one transmembrane domain and a C-terminal extracellular segment, which contains an epidermal growth factor homology domain. Little is known, however, about the intracellular N terminus of NRG1 type III, and the structure-function relationships of this cytoplasmic domain have remained uncharacterized. In the current study, we carried out the first structural and functional studies on the NRG1 type III cytoplasmic domain. Based on sequence analyses, the domain is predicted to be largely disordered, while a strictly conserved region close to the transmembrane segment may contain helical structure and bind metal ions. As shown by synchrotron radiation circular dichroism spectroscopy, the recombinant NRG1 type III cytoplasmic domain was disordered in solution, but it was able to fold partially into a helical structure, especially when both metals and membrane-mimicking compounds were present. NRG1 cytoplasmic tail binding to metals was further confirmed by calorimetry. These results suggest that the juxtamembrane segment of the NRG1 type III cytoplasmic domain may fold onto the membrane surface upon metal binding. Using synchrotron small-angle X-ray scattering, we further proved that the NRG1 cytoplasmic domain is intrinsically disordered, highly elongated, and behaves like a random polymer. Our work provides the first biochemical and biophysical data on the previously unexplored cytoplasmic domain of NRG1 type III, which will help elucidate the detailed structure-function relationships of this domain.


Asunto(s)
Neurregulina-1/química , Secuencia de Aminoácidos , Animales , Cationes/química , Dicroismo Circular , Citoplasma/metabolismo , ADN Complementario/genética , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metales/química , Metales/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Neurregulina-1/genética , Neurregulina-1/metabolismo , Pliegue de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Dispersión del Ángulo Pequeño , Alineación de Secuencia , Análisis de Secuencia de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...