Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Phytomedicine ; 118: 154942, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37421767

RESUMEN

BACKGROUND: The continuous evolution of SARS-CoV-2 has underscored the development of broad-spectrum prophylaxis. Antivirals targeting the membrane fusion process represent promising paradigms. Kaempferol (Kae), an ubiquitous plant flavonol, has been shown efficacy against various enveloped viruses. However, its potential in anti-SARS-CoV-2 invasion remains obscure. PURPOSE: To evaluate capabilities and mechanisms of Kae in preventing SARS-CoV-2 invasion. METHODS: To avoid interference of viral replication, virus-like particles (VLPs) constructed with luciferase reporter were applied. To investigate the antiviral potency of Kae, human induced pluripotent stem cells (hiPSC)-derived alveolar epithelial cells type II (AECII) and human ACE2 (hACE2) transgenic mice were utilized as in vitro and in vivo models, respectively. Using dual split protein (DSP) assays, inhibitory activities of Kae in viral fusion were determined in Alpha, Delta and Omicron variants of SARS-CoV-2, as well as in SARS-CoV and MERS-CoV. To further reveal molecular determinants of Kae in restricting viral fusion, synthetic peptides corresponding to the conserved heptad repeat (HR) 1 and 2, involved in viral fusion, and the mutant form of HR2 were explored by circular dichroism and native polyacrylamide gel electrophoresis. RESULTS: Kae inhibited SARS-CoV-2 invasion both in vitro and in vivo, which was mainly attributed to its suppressive effects on viral fusion, but not endocytosis, two pathways that mediate viral invasion. In accordance with the proposed model of anti-fusion prophylaxis, Kae functioned as a pan-inhibitor of viral fusion, including three emerged highly pathogenic coronaviruses, and the currently circulating Omicron BQ.1.1 and XBB.1 variants of SARS-CoV-2. Consistent with the typical target of viral fusion inhibitors, Kae interacted with HR regions of SARS-CoV-2 S2 subunits. Distinct from previous inhibitory fusion peptides which prevent the formation of six-helix bundle (6-HB) by competitively interacting with HRs, Kae deformed HR1 and directly reacted with lysine residues within HR2 region, the latter of which was considered critical for the preservation of stabilized S2 during SARS-CoV-2 invasion. CONCLUSIONS: Kae prevents SARS-CoV-2 infection by blocking membrane fusion and possesses a broad-spectrum anti-fusion ability. These findings provide valuable insights into potential benefits of Kae-containing botanical products as a complementary prophylaxis, especially during the waves of breakthrough infections and re-infections.


Asunto(s)
COVID-19 , Células Madre Pluripotentes Inducidas , Ratones , Animales , Humanos , SARS-CoV-2 , Secuencia de Aminoácidos , Quempferoles/farmacología , Glicoproteína de la Espiga del Coronavirus , Células Madre Pluripotentes Inducidas/metabolismo , Péptidos/química , Antivirales/farmacología , Antivirales/uso terapéutico
2.
Int J Nanomedicine ; 17: 3877-3892, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36097444

RESUMEN

Purpose: Peptide-based high-density lipoprotein (pHDL) structurally and functionally resembles the natural HDL as anti-atherosclerosis (AS) therapies. Since pHDL contains a large hydrophobic core, this study aims to evaluate the potentials of pHDL as a hydrophobic drug carrier and the efficiency of drug-loaded pHDL in the control of AS. Methods: The pHDL encapsulation of hydrophobic components from natural plants, including curcumin (Cur) and tanshinone IIA (TanIIA), was achieved using one-step microfluidics. Then, morphological features and loading efficiencies of pHDL-Cur and pHDL-TanIIA were determined by TEM and high-performance liquid chromatography (HPLC), respectively. Taking the fluorescence advantage of Cur, localizations of loaded Cur in pHDL were investigated by fluorescence quenchers, and recruitments of Cur to AS plaques were assessed with ex vivo imaging. Based on anti-inflammatory properties of TanIIA, pHDL-TanIIA was accordingly developed to evaluate the anti-AS effects through examinations of plasma lipid parameters and pathological alterations of plaque-associated regions. Results: Both lipophilic Cur and TanIIA can be efficiently loaded into pHDL carriers. The resultant pHDL-Cur and pHDL-TanIIA inherit the homogeneous nano-disk structure of pHDL. By using pHDL-Cur, the encapsulated hydrophobics are tracked in the core of pHDL, and incorporations of Cur with pHDL vehicles greatly improve the bioavailability and association of Cur with AS plaques. Moreover, when loaded with TanIIA, which has established its role in anti-AS as an anti-inflammatory candidate, synergistic effects in reducing AS lesions and improving pathological alterations of main organs related to AS were achieved. Conclusion: The pHDL system could potentially be applied for both imaging and therapy in animal models of AS. Benefits of pHDL-based drug delivery will potentially extend the application scenarios of bioactive chemicals from natural plants which are underutilized due to features like low bioavailability and facilitate the clinical translation of synthetic HDL therapies in HDL-associated disorders, including but not limited to AS.


Asunto(s)
Aterosclerosis , Curcumina , Animales , Aterosclerosis/tratamiento farmacológico , Disponibilidad Biológica , Portadores de Fármacos/química , Péptidos
3.
Nanotechnology ; 32(1): 015101, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33043894

RESUMEN

Electrospun composite nanofibrous scaffolds have been regarded as a potential carrier for local drug delivery to prevent tumor recurrence. Herein, a model drug (paclitaxel) was creatively loaded into lignin nanoparticles (PLNPs) and then encapsulated into the polymer of poly (vinyl alcohol)/polyvinyl pyrrolidone which has been fabricated into a composite nanofibrous membrane (PVA/PVP-PLNPs) for use as a drug carrier using the electrospinning technique. The fabricated PVA/PVP-PLNPs membranes exhibited good particle distribution, mechanical properties, thermal stability and biocompatibility. In vitro experiments showed that combining lignin nanoparticles by electrospinning not only improved the drug release profile, but also enhanced the hydrophilicity of nanofibrous membranes which was beneficial to cell adhesion and proliferation. Cellular experiments demonstrated that PVA/PVP-2%PLNPs membrane showed good cell inhibition ability, and the cell survival rate was only 21% at day 7. It indicates that the as-prepared PVA/PVP-PLNPs composite nanofibers are promising candidates for local anticancer therapy.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Portadores de Fármacos/química , Lignina/química , Paclitaxel/administración & dosificación , Alcohol Polivinílico/química , Neoplasias del Cuello Uterino/tratamiento farmacológico , Antineoplásicos Fitogénicos/farmacología , Femenino , Células HeLa , Humanos , Nanofibras/química , Paclitaxel/farmacología , Povidona/química
4.
J Control Release ; 301: 129-139, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30880079

RESUMEN

In bone tissue engineering, electrospun fibrous scaffolds can provide excellent mechanical support, extracellular matrix mimicking components, such as 3D spacial fibrous environment for cell growth and controlled release of signaling molecules for osteogenesis. Here, a facile strategy comprising the incorporation of an osteogenic inductive peptide H1, derived from the cysteine knot (CT) domain of connective tissue growth factor (CTGF), in the core of Silk Fibroin (SF) was developed for osteogenic induction, synergistically with co-delivering hydroxyapatite (HA) from the shell of poly(l-lactic acid-co-ε-caprolactone) (PLCL). The core-shell nanofibrous structure was confirmed by transmission electron microscopy (TEM). Furthermore, the sustained released H1 has effectively promoted proliferation and osteoblastic differentiation of human induced pluripotent stem cells-derived mesenchymal stem cells (hiPS-MSCs). Moreover, after 8 weeks implantation in mice, this SF-H1/PLCL-HA composite induced bone tissue formation significantly faster than SF/PLCL as indicated by µCT. The present study is the first to demonstrate that release of short hydrophilic peptides derived from CTGF combined with HA potentiated the regenerative capacity for healing critical sized calvarial defect in vivo.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/administración & dosificación , Durapatita/administración & dosificación , Osteogénesis/efectos de los fármacos , Péptidos/administración & dosificación , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/química , Liberación de Fármacos , Durapatita/química , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Péptidos/química , Cráneo/diagnóstico por imagen , Cráneo/efectos de los fármacos , Cráneo/fisiología , Microtomografía por Rayos X
5.
J Biomed Mater Res B Appl Biomater ; 106(6): 2266-2274, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29087024

RESUMEN

Compared to terminal differentiated cells, stem cells play important roles in the maintenance and regeneration, and thus have been intensively researched as the most promising cell based therapy. In order to maximize the effectiveness of stem cell based therapies, it is essential to understand the environmental (niche) signals that regulate stem cell behavior. Recent findings suggest that fibroblasts have a mesenchymal origin and that mesenchymal stem cells (MSCs) demonstrate proangiogenic function, where both fibrogenic and angiogenic activities are associated with connective tissue growth factor (CTGF), a matricellular protein that serves as an essential mediator of skeletogenesis in development and vascular remodeling. Here, for the first time, we demonstrate that upon local delivery of CTGF from a three dimensional (3D) nanocomposite scaffold, human induced pluripotent stem cells derived MSCs can be directed to differentiate toward fibroblasts in a 3D nanocomposite scaffold in female nonobese diabetic CB-17/Icr-severe combined immunodeficient mice. The stem cell-scaffold constructs present not only intriguingly strong fibroblastic commitments but also angiogenic induction in vivo. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 2266-2274, 2018.


Asunto(s)
Células Inmovilizadas , Factor de Crecimiento del Tejido Conjuntivo , Células Madre Pluripotentes Inducidas/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Nanocompuestos/química , Andamios del Tejido/química , Animales , Diferenciación Celular/efectos de los fármacos , Células Inmovilizadas/citología , Células Inmovilizadas/metabolismo , Células Inmovilizadas/trasplante , Factor de Crecimiento del Tejido Conjuntivo/química , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Xenoinjertos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID
6.
Sci Rep ; 7(1): 8509, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28819120

RESUMEN

The success of mesenchymal stem cell transplantation is highly dependent on their survival and controlled fate regulation. This study demonstrates that dual-delivery of connective tissue growth factor (CTGF) and fibroblast growth factor 2 (FGF-2) from a core-shell fiber of Silk Fibroin/poly(L-lactic acid-co-ε-caprolactone)-polyethylene oxide (SF/PLCL-PEO) enhanced fibrogenic lineage differentiation of MSCs. The core-shell structure was confirmed by transmission electron microscopy (TEM), fluorescence microscopy and attenuated total reflection (ATR) Fourier transform infrared (FTIR) spectroscopy. A sequential release of FGF-2 and CTGF was successfully achieved in this manner. FGF-2 plays an important role in stem cell proliferation and, meanwhile when accompanied with CTGF, has a slightly additive effect on fibrogenic differentiation of MSCs, whereas CTGF promotes fibrogenesis and alleviates osteogenesis, chondrogenesis and adipogenesis.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento del Tejido Conjuntivo/farmacocinética , Factor 2 de Crecimiento de Fibroblastos/farmacocinética , Fibroínas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Poliésteres/metabolismo , Animales , Células Cultivadas , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Fibroínas/química , Fibroínas/ultraestructura , Células Madre Mesenquimatosas/fisiología , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Poliésteres/química , Ratas , Espectroscopía Infrarroja por Transformada de Fourier
7.
ACS Appl Mater Interfaces ; 8(25): 15864-73, 2016 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-27265317

RESUMEN

Electrospinning has been widely applied for tissue engineering due to its versatility of fabricating extracellular matrix (ECM) mimicking fibrillar scaffolds. Yet there are still challenges such as that these two-dimensional (2D) tightly packed, hydrophobic fibers often hinder cell infiltration and cell-scaffold integration. In this study, polycaprolactone (PCL) was electrospun into a grounded coagulation bath collector, resulting in 3D coiled microfibers with in situ surface functionalization with hydrophilic, catecholic polydopamine (pDA). The 3D scaffolds showed biocompatibility and were well-integrated with human bone marrow derived human mesenchymal stem cells (hMSCs), with significantly higher cell penetration depth compared to that of the 2D PCL microfibers from traditional electrospinning. Further differentiation of human mesenchymal stem cells (hMSCs) into fibroblast phenotype in vitro indicates that, compared to the stiff, tightly packed, 2D scaffolds which aggravated myofibroblasts related activities, such as upregulated gene and protein expression of α-smooth muscle actin (α-SMA), 3D scaffolds induced milder myofibroblastic differentiation. The flexible 3D fibers further allowed contraction with the well-integrated, mechanically active myofibroblasts, monitored under live-cell imaging, whereas the stiff 2D scaffolds restricted that.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Indoles/farmacología , Células Madre Mesenquimatosas/citología , Polímeros/farmacología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Proliferación Celular , Células Cultivadas , Humanos , Indoles/química , Poliésteres/química , Polímeros/química , Andamios del Tejido/química
8.
Cell Physiol Biochem ; 35(6): 2437-50, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25967873

RESUMEN

BACKGROUND/AIMS: Pluripotent stem cells differentiating into cardiomyocyte-like cells in an appropriate cellular environment have attracted significant attention, given the potential use of such cells for regenerative medicine. However, the precise mechanisms of lineage specification of pluripotent stem cells are still largely to be explored. Identifying the role of various small synthetic peptides involved in cardiomyogenesis may provide new insights into pathways promoting cardiomyogenesis. METHODS: In the present study, using a transgenic murine embryonic stem (ES) cell lineage expressing enhanced green fluorescent protein (EGFP) under the control of α-myosin heavy chain (α-MHC) promoter (pαMHC-EGFP), we investigated the cardiomyogenic effects of 7 synthetic peptides (Betrofin3, FGLs, FGL(L), hNgf_C2, EnkaminE, Plannexin and C3) on cardiac differentiation. The expression of several cardiac-specific markers was determined by RT-PCR whereas the structural and functional properties of derived cardiomyocytes were examined by immunofluorescence and electrophysiology, respectively. RESULTS: The results revealed that Betrofin3, an agonist of brain derived neurotrophic factor (BDNF) peptide exerted the most striking pro-cardiomyogenic effect on ES cells. We found that BDNF receptor, TrkB expression was up-regulated during differentiation. Treatment of differentiating cells with Betrofin3 between days 3 and 5 enhanced the expression of cardiac-specific markers and improved cardiomyocyte differentiation and functionality as revealed by genes regulation, flow cytometry and patch clamp analysis. Thus Betrofin3 may exert its cardiomyogenic effects on ES cells via TrkB receptor. CONCLUSION: Taken together, the results suggest that Betrofin3 modulates BDNF signaling with positive cardiomyogenic effect in stage and dose-dependent manner providing an effective strategy to increase ES cell-based generation of cardiomyocytes and offer a novel therapeutic approach to cardiac pathologies where BDNF levels are impaired.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias de Ratones/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/farmacología , Péptidos/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Dendrímeros/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Miocitos Cardíacos/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Oligopéptidos/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Receptor trkB/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Phys Chem Chem Phys ; 17(14): 9446-53, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25766518

RESUMEN

Based on the catecholic chemistry of a mussel inspired coating, norepinephrine (NE), a catecholamine found both in neurotransmitters and mussel adhesive proteins, was for the first time applied as a unique bio-interface integrating multi-functions facilitating PC12 neuron-like differentiation. A uniform, ultra-smooth pNE coating was achieved on electrospun submicron PLCL fibers, proven by surface characterization. The introduced catechol groups from pNE were further used to not only anchor collagen to enhance cell adhesion but also localize nerve growth factor to promote neuron-like differentiation. The obtained pNE-collagen coating was found to be a superior substrate for PC12 differentiation, confirmed by both cellular toxicity/viability assays and immunochemical staining. The aligned PLCL fiber conformation further steered neurite formation along the fiber direction and contributed to neurite extension and increased the number of neurites per cell body. This facile pNE coating might lead to a more efficient use of growth factor, drugs and other bioactive molecules with lower loading dosage and sustained activity resulting in enhanced therapeutic effects and decreased adverse effects.


Asunto(s)
Diferenciación Celular , Colágeno/química , Neuritas/fisiología , Neuronas/citología , Norepinefrina/química , Andamios del Tejido/química , Animales , Adhesión Celular , Células PC12 , Ratas
10.
Sci Rep ; 5: 8480, 2015 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-25684543

RESUMEN

Fibroblasts are ubiquitous cells that constitute the stroma of virtually all tissues and play vital roles in homeostasis. The poor innate healing capacity of fibroblastic tissues is attributed to the scarcity of fibroblasts as collagen-producing cells. In this study, we have developed a functional ECM mimicking scaffold that is capable to supply spatial allocation of stem cells as well as anchorage and storage of growth factors (GFs) to direct stem cells differentiate towards fibroblasts. Electrospun PCL fibers were embedded in a PEG-fibrinogen (PF) hydrogel, which was infiltrated with connective tissue growth factor (CTGF) to form the 3D nanocomposite PFP-C. The human induced pluripotent stem cells derived mesenchymal stem cells (hiPS-MSCs) with an advance in growth over adult MSCs were applied to validate the fibrogenic capacity of the 3D nanocomposite scaffold. The PFP-C scaffold was found not only biocompatible with the hiPS-MSCs, but also presented intriguingly strong fibroblastic commitments, to an extent comparable to the positive control, tissue culture plastic surfaces (TCP) timely refreshed with 100% CTGF. The novel scaffold presented not only biomimetic ECM nanostructures for homing stem cells, but also sufficient cell-approachable bio-signaling cues, which may synergistically facilitate the control of stem cell fates for regenerative therapies.


Asunto(s)
Diferenciación Celular , Matriz Extracelular , Fibroblastos/citología , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/citología , Andamios del Tejido , Materiales Biocompatibles , Técnicas de Cultivo de Célula , Supervivencia Celular , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Humanos , Inmunofenotipificación , Células Madre Mesenquimatosas/metabolismo , Nanocompuestos , Fenotipo
11.
AAPS J ; 16(3): 400-12, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24557747

RESUMEN

Differentiation of pluripotent stem cells, PSCs, towards neural lineages has attracted significant attention, given the potential use of such cells for in vitro studies and for regenerative medicine. The present experiments were designed to identify bioactive peptides which direct PSC differentiation towards neural cells. Fifteen peptides were designed based on NCAM, FGFR, and growth factors sequences. The effect of peptides was screened using a mouse embryonic stem cell line expressing luciferase dual reporter construct driven by promoters for neural tubulin and for elongation factor 1. Cell number was estimated by measuring total cellular DNA. We identified five peptides which enhanced activities of both promoters without relevant changes in cell number. We selected the two most potent peptides for further analysis: the NCAM-derived mimetic FGLL and the synthetic NCAM ligand, Plannexin. Both compounds induced phenotypic neuronal differentiation, as evidenced by increased neurite outgrowth. In summary, we used a simple, but sensitive screening approach to identify the neurogenic peptides. These peptides will not only provide new clues concerning pathways of neurogenesis, but they may also be interesting biotechnology tools for in vitro generation of neurons.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Células-Madre Neurales/efectos de los fármacos , Péptidos/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Humanos , Indicadores y Reactivos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Neuritas/efectos de los fármacos , Neuritas/ultraestructura
12.
Neurochem Res ; 38(12): 2550-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24132641

RESUMEN

ErbB receptors not only function in cancer, but are also key developmental regulators in the nervous system. We previously identified an ErbB1 peptide antagonist, Inherbin3, that is capable of inhibiting tumor growth in vitro and in vivo. In this study, we found that inhibition of ErbB1 kinase activity and activation of ErbB4 by NRG-1ß induced neurite extension, suggesting that ErbB1 and ErbB4 act as negative and positive regulators, respectively, of the neuritogenic response. Inherbin3, inhibited activation not only of ErbB1 but also of ErbB4 in primary neurons, strongly induced neurite outgrowth in rat cerebellar granule neurons, indicating that this effect mainly was due to inhibition of ErbB1 activation.


Asunto(s)
Cerebelo/efectos de los fármacos , Gránulos Citoplasmáticos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Neuritas/efectos de los fármacos , Péptidos/farmacología , Animales , Secuencia de Bases , Células Cultivadas , Cerebelo/citología , Cartilla de ADN , Receptores ErbB/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Fosforilación , Reacción en Cadena de la Polimerasa , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Cell Oncol (Dordr) ; 36(3): 201-11, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23580313

RESUMEN

BACKGROUND: Receptors of the ErbB family are involved in the development of various cancers, and the inhibition of these receptors represents an attractive therapeutic concept. Upon ligand binding, ErbB receptors become activated as homo- or heterodimers, leading to the activation of downstream signaling cascades that result in the facilitation of cell proliferation and migration. A region of the extracellular part of the receptor, termed the 'dimerization arm', is important for the formation of receptor dimers and represents an attractive target for the design of ErbB inhibitors. METHODS: An ErbB1 targeting peptide, termed Herfin-1, was designed based on a model of the tertiary structure of the EGF-EGFR ternary complex. The binding kinetics of this peptide were determined employing surface plasmon resonance analyses. ErbB1-4 expression and phosphorylation in human glioblastoma cell lines U87 and U118 were determined by Western blotting using specific antibodies. Cell proliferation was determined by MTS staining. Cell migration was examined using a Chemotaxis Migration Kit. Neurite outgrowth from primary cerebellar granule neurons was evaluated by fluorescence microscopy and image processing. RESULTS: The present study shows that Herfin-1 functions as an ErbB1 antagonist. It binds to the extracellular domain of ErbB1 with a KD value of 361 nM. In U87 and U118 cells, both expressing high levels of ErbB1, Herfin-1 inhibits EGF-induced ErbB1 phosphorylation and cell migration. Additionally, Herfin-1 was found to increase neurite outgrowth in cerebellar granule neurons, likely through the inhibition of a sustained weak ErbB1 activation. CONCLUSIONS: Targeting the ErbB1 receptor dimerization interface is a promising strategy to inhibit receptor activation in ErbB1-expressing glioma cells.


Asunto(s)
Neoplasias Encefálicas/patología , Movimiento Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Glioblastoma/patología , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Diseño de Fármacos , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/química , Receptores ErbB/metabolismo , Glioblastoma/tratamiento farmacológico , Humanos , Datos de Secuencia Molecular , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neurogénesis/efectos de los fármacos , Péptidos/química , Péptidos/uso terapéutico , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Ratas Wistar
14.
Cell Oncol ; 32(4): 259-74, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20364069

RESUMEN

The epidermal growth factor family of receptor tyrosine kinases (ErbBs) plays essential roles in tumorigenesis and cancer disease progression, and therefore has become an attractive target for structure-based drug design. ErbB receptors are activated by ligand-induced homo- and heterodimerization. Structural studies have revealed that ErbB receptor dimers are stabilized by receptor-receptor interactions, primarily mediated by a region in the second extracellular domain, termed the "dimerization arm". The present study is the first biological characterization of a peptide, termed Inherbin3, which constitutes part of the dimerization arm of ErbB3. Inherbin3 binds to the extracellular domains of all four ErbB receptors, with the lowest peptide binding affinity for ErbB4. Inherbin3 functions as an antagonist of epidermal growth factor (EGF)-ErbB1 signaling. We show that Inherbin3 inhibits EGF-induced ErbB1 phosphorylation, cell growth, and migration in two human tumor cell lines, A549 and HN5, expressing moderate and high ErbB1 levels, respectively. Furthermore, we show that Inherbin3 inhibits tumor growth in vivo and induces apoptosis in a tumor xenograft model employing the human non-small cell lung cancer cell line A549. The Inherbin3 peptide may be a useful tool for investigating the mechanisms of ErbB receptor homo- and heterodimerization. Moreover, the here described biological effects of Inherbin3 suggest that peptide-based targeting of ErbB receptor dimerization is a promising anti-cancer therapeutic strategy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Mucosa Respiratoria/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones SCID , Trasplante de Neoplasias , Fragmentos de Péptidos/química , Agregación de Receptores/efectos de los fármacos , Receptor ErbB-3/química , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Carga Tumoral/efectos de los fármacos
15.
Exp Mol Med ; 41(10): 717-27, 2009 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-19561401

RESUMEN

Triptolide, a diterpenoid triepoxide from the traditional Chinese medicinal herb Tripterygium wilfordii Hook. f., is a potential treatment for autoimmune diseases as well a possible anti-tumor agent. It inhibits proliferation of colorectal cancer cells in vitro and in vivo. In this study, its ability to block progress of colitis to colon cancer, and its molecular mechanism of action are investigated. A mouse model for colitis-induced colorectal cancer was used to test the effect of triptolide on cancer progression. Treatment of mice with triptolide decreased the incidence of colon cancer formation, and increased survival rate. Moreover, triptolide decreased the incidence of tumors in nude mice inoculated with cultured colon cancer cells dose-dependently. In vitro, triptolide inhibited the proliferation, migration and colony formation of colon cancer cells. Secretion of IL6 and levels of JAK1, IL6R and phosphorylated STAT3 were all reduced by triptolide treatment. Triptolide prohibited Rac1 activity and blocked cyclin D1 and CDK4 expression, leading to G1 arrest. Triptolide interrupted the IL6R-JAK/STAT pathway that is crucial for cell proliferation, survival, and inflammation. This suggests that triptolide might be a candidate for prevention of colitis induced colon cancer because it reduces inflammation and prevents tumor formation and development.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Diterpenos/administración & dosificación , Fenantrenos/administración & dosificación , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rac1/biosíntesis , Animales , Colitis/complicaciones , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Sulfato de Dextran/toxicidad , Dimetilhidrazinas/toxicidad , Compuestos Epoxi/administración & dosificación , Humanos , Interleucina-6/biosíntesis , Quinasas Janus/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Desnudos , Trasplante de Neoplasias , Factor de Transcripción STAT3/metabolismo , Carga Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA