Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharmacol ; 105(1): 39-53, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37977824

RESUMEN

Hematopoietic cell transplantation (HCT) is often considered a last resort leukemia treatment, fraught with limited success due to microbial infections, a leading cause of mortality in leukemia patients. To address this critical issue, we explored a novel approach by synthesizing antileukemic agents containing antibacterial substances. This innovative strategy involves conjugating fluoroquinolone antibiotics, such as ciprofloxacin (CIP) or levofloxacin (LVX), with the cell-penetrating peptide transportan 10 (TP10). Here, we demonstrate that the resultant compounds display promising biologic activities in preclinical studies. These novel conjugates not only exhibit potent antimicrobial effects but are also selective against leukemia cells. The cytotoxic mechanism involves rapid disruption of cell membrane asymmetry leading to membrane damage. Importantly, these conjugates penetrated mammalian cells, accumulating within the nuclear membrane without significant effect on cellular architecture or mitochondrial function. Molecular simulations elucidated the aggregation tendencies of TP10 conjugates within lipid bilayers, resulting in membrane disruption and permeabilization. Moreover, mass spectrometry analysis confirmed efficient reduction of disulfide bonds within TP10 conjugates, facilitating release and activation of the fluoroquinolone derivatives. Intriguingly, these compounds inhibited human topoisomerases, setting them apart from traditional fluoroquinolones. Remarkably, TP10 conjugates generated lower intracellular levels of reactive oxygen species compared with CIP and LVX. The combination of antibacterial and antileukemic properties, coupled with selective cytostatic effects and minimal toxicity toward healthy cells, positions TP10 derivatives as promising candidates for innovative therapeutic approaches in the context of antileukemic HCT. This study highlights their potential in search of more effective leukemia treatments. SIGNIFICANCE STATEMENT: Fluoroquinolones are commonly used antibiotics, while transportan 10 (TP10) is a cell-penetrating peptide (CPP) with anticancer properties. In HCT, microbial infections are the primary cause of illness and death. Combining TP10 with fluoroquinolones enhanced their effects on different cell types. The dual pharmacological action of these conjugates offers a promising proof-of-concept solution for leukemic patients undergoing HCT. Strategically designed therapeutics, incorporating CPPs with antibacterial properties, have the potential to reduce microbial infections in the treatment of malignancies.


Asunto(s)
Antineoplásicos , Péptidos de Penetración Celular , Leucemia , Animales , Humanos , Fluoroquinolonas/farmacología , Péptidos de Penetración Celular/farmacología , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/metabolismo , Antineoplásicos/farmacología , Antibacterianos/farmacología , Leucemia/tratamiento farmacológico , Trasplante de Células , Mamíferos/metabolismo
2.
Int J Mol Sci ; 24(8)2023 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-37108504

RESUMEN

Thioredoxin (Trx) plays a critical role in maintaining redox balance in various cells and exhibits anti-oxidative, anti-apoptotic, and anti-inflammatory effects. However, whether exogenous Trx can inhibit intracellular oxidative damage has not been investigated. In previous study, we have identified a novel Trx from the jellyfish Cyanea capillata, named CcTrx1, and confirmed its antioxidant activities in vitro. Here, we obtained a recombinant protein, PTD-CcTrx1, which is a fusion of CcTrx1 and protein transduction domain (PTD) of HIV TAT protein. The transmembrane ability and antioxidant activities of PTD-CcTrx1, and its protective effects against H2O2-induced oxidative damage in HaCaT cells were also detected. Our results revealed that PTD-CcTrx1 exhibited specific transmembrane ability and antioxidant activities, and it could significantly attenuate the intracellular oxidative stress, inhibit H2O2-induced apoptosis, and protect HaCaT cells from oxidative damage. The present study provides critical evidence for application of PTD-CcTrx1 as a novel antioxidant to treat skin oxidative damage in the future.


Asunto(s)
Péptidos de Penetración Celular , Escifozoos , Animales , Productos del Gen tat/metabolismo , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/metabolismo , Péptidos de Penetración Celular/farmacología , Péptidos de Penetración Celular/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Estrés Oxidativo , Escifozoos/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/farmacología , Tiorredoxinas/química
3.
Molecules ; 27(11)2022 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-35684309

RESUMEN

The anticancer agent doxorubicin(dox) has been widely used in the treatment of a variety of hematological malignancies and solid tumors. Despite doxorubicin's efficiency in killing tumor cells, severe damage to healthy tissues, along with cardiotoxicity, limits its clinical use. To overcome these adverse side effects, improve patient safety, and enhance therapeutic efficacy, we have designed a thermally responsive biopolymer doxorubicin carrier that can be specifically targeted to tumor tissue by locally applying mild hyperthermia (41 °C). The developed drug vehicle is composed of the following: a cell penetrating peptide (SynB1) to promote tumor and cellular uptake; thermally responsive Elastin-like polypeptide (ELP); and the (6-maleimidocaproyl) hydrazone derivative of doxorubicin (DOXO-EMCH) containing a pH-sensitive hydrazone linker that releases doxorubicin in the acidic tumor environment. We used the in vivo imaging system, IVIS, to determine biodistribution of doxorubicin-delivered ELP in MDA-MB-231 xenografts in nude mice. Tumor bearing mice were treated with a single IV injection of 10 mg/kg doxorubicin equivalent dose with free doxorubicin, thermally responsive SynB1 ELP 1-DOXO, and a thermally nonresponsive control biopolymer, SynB1 ELP 2-DOXO. Following a 2 h treatment with hyperthermia, tumors showed a 2-fold higher uptake when treated with SynB1 ELP 1-DOXO compared to free doxorubicin. Accumulation of the thermally non-responsive control SynB1 ELP2 -DOXO was comparable to free doxorubicin, indicating that an increase in dox accumulation with ELP is due to aggregation in response to thermal targeting. Higher levels of SynB1 ELP1-DOXO and SynB1 ELP2 -DOXO with respect to free doxorubicin were observed in kidneys. Fluorescence intensity from hearts of animals treated with SynB1 ELP1-DOXO show a 5-fold decrease in accumulation of doxorubicin than the same dose of free doxorubicin. SynB1-ELP1-DOXO biopolymers demonstrated a 6-fold increase in tumor/heart ratio in comparison to free doxorubicin, indicating preferential accumulation of the drug in tumors. These results demonstrate that thermally targeted polymers are a promising therapy to enhance tumor targeting and uptake of anticancer drugs and to minimize free drug toxicity in healthy tissues, representing a great potential for clinical application.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Péptidos de Penetración Celular , Hipertermia Inducida , Animales , Neoplasias de la Mama/tratamiento farmacológico , Cardiotoxicidad/prevención & control , Péptidos de Penetración Celular/farmacología , Doxorrubicina , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Hidrazonas , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Desnudos , Distribución Tisular
4.
J Biotechnol ; 354: 34-44, 2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-35724765

RESUMEN

Gold nanoparticles (AuNPs) are widely used as an agent in photothermal therapy (PTT) against various cancers. However, a drug delivery system (DDS) is required for effective PTT using AuNPs as AuNPs accumulate passively in tumors. In the present study, we used polyhistidine peptide, a novel cell-penetrating peptide, which is efficiently internalized into tumor cells, as a DDS carrier for PTT using AuNPs. Polyhistidine peptide-modified AuNPs are efficiently internalized into RERF-LC-AI human lung squamous cancer cells and localized to the intracellular lysosome, which is based on the nature of the polyhistidine peptide. Furthermore, the polyhistidine peptide-modified AuNPs inhibited proliferation of RERF-LC-AI cells in a polyhistidine peptide modification-dependent manner under 660 nm laser irradiation. Quantitative real-time PCR showed increased expression levels of an apoptosis-related gene (bax) and heat stress-related gene (hsp70) in RERF-LC-AI cells treated with polyhistidine peptide-modified AuNPs and laser. Our findings highlight the efficacy of AuNPs modified with H16 peptide in PTT.


Asunto(s)
Péptidos de Penetración Celular , Histidina , Nanopartículas del Metal , Línea Celular Tumoral , Péptidos de Penetración Celular/farmacología , Oro/farmacología , Histidina/farmacología , Humanos , Nanopartículas del Metal/uso terapéutico , Fototerapia/métodos
5.
Eur J Pharmacol ; 913: 174638, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34801531

RESUMEN

Cerebral stroke, commonly caused due to hindrance in blood flow, is broadly classified into two categories-ischemic and haemorrhagic strokes. The onset of stroke triggers multiple mechanisms causing inflammation, generation of free radicals and protein damage leading to apoptosis of neuronal cells. The current therapies available for cerebral strokes involve use of complex surgical treatments and tissue plasminogen activator which increases the risk of internal bleeding, brain edema and cerebral damage, thereby restricting their use in clinical setting. The alarming need to develop safe, effective, target specific systems which, promote neuronal growth and reduce cerebral inflammation can be accomplished with use of biotechnological approaches. The article gives an insight to biotechnology-based advancements for tissue plasminogen activators, cell penetrating peptides, growth factors, ribonucleic acid systems and monoclonal antibodies for cerebral stroke. We also emphasis on challenges and future perspective of biotechnology-based therapeutics for better management of stroke.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Biotecnología/tendencias , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Péptidos de Penetración Celular/farmacología , Péptidos de Penetración Celular/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Accidente Cerebrovascular/patología
6.
J Pept Sci ; 26(10): e3277, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32729203

RESUMEN

Herein, we make an effort to enhance the antimicrobial activity of levofloxacin (LVX) antibiotic via conjugation to a cell-penetrating peptide (CPP) including Cys-Gly-Ala-Phe-Pro-His-Arg. For this purpose, cysteine is used as a linker between the LVX and CPP chain, and two heterogeneous nanoscale catalytic systems are employed as the substantial alternatives for traditional peptide coupling reagents like N,N,N',N'-tetramethyl-O-(benzotriazol-1-yl)uronium tetrafluoroborate (TBTU). Briefly, it has been found out that the antimicrobial potency of the synthesized CPP-LVX conjugate (on the gram-positive and gram-negative bacteria) is noticeably enhanced (~20% more). It has been revealed via zone of inhibition (ZOI) and optical density (OD) evaluations. As a convenient method for making this type of the effective conjugations, ultrasound waves (with a specific frequency and power density) activate the catalytic sites of the heterogeneous nanoparticles. Through this synergistic effect, peptide/amide bond is formed during a short time (10 min), and high reaction yield (>90%) is obtained under mild conditions. Moreover, as a simple purification process, the catalyst nanoparticles are collected and separated through their high magnetic property.


Asunto(s)
Antibacterianos/farmacología , Péptidos de Penetración Celular/farmacología , Levofloxacino/farmacología , Nanopartículas/química , Ondas Ultrasónicas , Antibacterianos/síntesis química , Antibacterianos/química , Catálisis , Supervivencia Celular/efectos de los fármacos , Péptidos de Penetración Celular/química , Relación Dosis-Respuesta a Droga , Escherichia coli/efectos de los fármacos , Levofloxacino/química , Fenómenos Magnéticos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Staphylococcus aureus/efectos de los fármacos
7.
J Nanobiotechnology ; 18(1): 48, 2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32183823

RESUMEN

BACKGROUND: Specific targeting ability and good cell penetration are two critical requirements of tumor-targeted delivery systems. In the present work, we developed a novel matrix metalloprotein-triggered, cell-penetrating, peptide-modified, star-shaped nanoparticle (NP) based on a functionalized copolymer (MePEG-Peptide-Tri-CL), with the peptide composed of GPLGIAG (matrix metalloprotein-triggered peptide for targeted delivery) and r9 (cell-penetrating peptide for penetration improvement) to enhance its biological specificity and therapeutic effect. RESULTS: Based on the in vitro release study, a sustained release profile was achieved for curcumin (Cur) release from the Cur-P-NPs at pH 7.4. Furthermore, the release rate of Cur was accelerated in the enzymatic reaction. MTT assay results indicated that the biocompatibility of polymer NPs (P-NPs) was inversely related to the NP concentration, while the efficiency toward tumor cell inhibition was positively related to the Cur-P-NP concentration. In addition, Cur-P-NPs showed higher fluorescence intensity than Cur-NPs in tumor cells, indicating improved penetration of tumor cells. An in vivo biodistribution study further demonstrated that Cur-P-NPs exhibited stronger targeting to A549 xenografts than to normal tissue. Furthermore, the strongest tumor growth inhibition (76.95%) was observed in Cur-P-NP-treated A549 tumor xenograft nude mice, with slight pulmonary toxicity. CONCLUSION: All results demonstrated that Cur-P-NP is a promising drug delivery system that possesses specific enzyme responsiveness for use in anti-tumor therapy.


Asunto(s)
Péptidos de Penetración Celular/farmacología , Sistemas de Liberación de Medicamentos/métodos , Metaloproteínas/farmacología , Nanopartículas/administración & dosificación , Animales , Línea Celular Tumoral , Péptidos de Penetración Celular/química , Curcumina/farmacología , Portadores de Fármacos , Liberación de Fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Tamaño de la Partícula , Polímeros/química , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Pharm ; 17(2): 683-694, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31913047

RESUMEN

Plant-based saponins are amphipathic glycosides composed of a hydrophobic aglycone backbone covalently bound to one or more hydrophilic sugar moieties. Recently, the endosomal escape activity of triterpenoid saponins has been investigated as a potentially powerful tool for improved cytosolic penetration of protein drugs internalized by endocytic uptake, thereby greatly enhancing their pharmacological effects. However, only a few saponins have been studied, and the paucity in understanding the structure-activity relationship of saponins imposes significant limitations on their applications. To address this knowledge gap, 12 triterpenoid saponins with diverse structural side chains were screened for their utility as endosomolytic agents. These compounds were used in combination with a toxin (MAP30-HBP) comprising a type I ribosome-inactivating protein fused to a cell-penetrating peptide. Suitability of saponins as endosomolytic agents was assessed on the basis of cytotoxicity, endosomal escape promotion, and synergistic effects on toxins. Five saponins showed strong endosomal escape activity, enhancing MAP30-HBP cytotoxicity by more than 106 to 109 folds. These saponins also enhanced the apoptotic effect of MAP30-HBP in a pH-dependent manner. Additionally, growth inhibition of MAP30-HBP-treated SMMC-7721 cells was greater than that of similarly treated HeLa cells, suggesting that saponin-mediated endosomolytic effect is likely to be cell-specific. Furthermore, the structural features and hydrophobicity of the sugar side chains were analyzed to draw correlations with endosomal escape activity and derive predictive rules, thus providing new insights into structure-activity relationships of saponins. This study revealed new saponins that can potentially be exploited as efficient cytosolic delivery reagents for improved therapeutic drug effects.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Endosomas/efectos de los fármacos , Saponinas/química , Saponinas/farmacología , Triterpenos/química , Triterpenos/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/farmacología , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Sinergismo Farmacológico , Glicosilación , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Extractos Vegetales/química , Extractos Vegetales/farmacología , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Relación Estructura-Actividad
9.
J Biol Chem ; 294(51): 19604-19615, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31727740

RESUMEN

Disulfide-rich plant peptides with molecular masses of 2-6 kDa represent an expanding class of peptidyl-type natural products with diverse functions. They are structurally compact, hyperstable, and underexplored as cell-penetrating agents that inhibit intracellular functions. Here, we report the discovery of an anionic, 34-residue peptide, the disulfide-rich roseltide rT7 from Hibiscus sabdariffa (of the Malvaceae family) that penetrates cells and inhibits their proteasomal activities. Combined proteomics and NMR spectroscopy revealed that roseltide rT7 is a cystine-knotted, six-cysteine hevein-like cysteine-rich peptide. A pair-wise comparison indicated that roseltide rT7 is >100-fold more stable against protease degradation than its S-alkylated analog. Confocal microscopy studies and cell-based assays disclosed that after roseltide rT7 penetrates cells, it causes accumulation of ubiquitinated proteins, inhibits human 20S proteasomes, reduces tumor necrosis factor-induced IκBα degradation, and decreases expression levels of intercellular adhesion molecule-1. Structure-activity studies revealed that roseltide rT7 uses a canonical substrate-binding mechanism for proteasomal inhibition enabled by an IIML motif embedded in its proline-rich and exceptionally long intercysteine loop 4. Taken together, our results provide mechanistic insights into a novel disulfide-rich, anionic, and cell-penetrating peptide, representing a potential lead for further development as a proteasomal inhibitor in anti-cancer or anti-inflammatory therapies.


Asunto(s)
Péptidos de Penetración Celular/farmacología , Hibiscus/química , Extractos Vegetales/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Células A549 , Antiinflamatorios/farmacología , Péptidos Catiónicos Antimicrobianos , Antineoplásicos Fitogénicos/farmacología , Cisteína/química , Disulfuros , Endocitosis , Citometría de Flujo , Humanos , Espectroscopía de Resonancia Magnética , Microscopía Confocal , Conformación Molecular , Lectinas de Plantas , Proteínas de Plantas/química , Proteómica , Relación Estructura-Actividad , Ubiquitina/química
10.
J Pept Sci ; 25(10): e3205, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31612571

RESUMEN

In this study, six N-1, N-2, or N-11 derivatives of TD-34 (a cationic cyclic cell-penetrating peptide [CPP], ACSSKKSKHCG) were designed and synthesized including both linear peptides and cyclic peptides, such as DL-1 (KWSSKKSKHCG), DLCC-1 (cyclopeptide, KWSSKKSKHCG), DL-2 (KWSSKKSKHCG-NH2 ), DLCC-2 (cyclopeptide, KWSSKKSKHCG-NH2 ), DL-3 (RWSSKKSKHCG), and DLCC-3 (cyclopeptide, RWSSKKSKHCG). The cyclic peptides were synthesized by disulfide bound linkages formed by N-2 and N-10 cysteine. In vitro penetration experiment was conducted to investigate the transdermal enhancement ability of these derivatives, using triptolide (TP) as model drug. The results display that at the presence of DLCC-2, the accumulative penetration amount of TP increased 1.71-fold (P < .05) within 12 hours, displaying better transdermal enhancing ability than TD-34. Meanwhile, DL-3 and DLCC-3 slightly decreased the transdermal delivery of TP, and the presence of DL-1 and DLCC-1 shows no obvious effect. In order to clarify the factors on the transdermal ability of peptides, the solubility of TP in phosphate buffer saline (PBS) at the presence of different peptides and the mechanism of transdermal delivery of CPPs was investigated. The result shows that most of these peptides have no significant effect on the solubility of TP except DLCC-3 (the solubility of TP slightly increased). And in order to investigate transdermal absorption route of DLCC-2, polyarginine linked to rhodamine b (Rh b) derivative is used. The result proved that the transdermal route of polyarginine is via hair follicle, which may change the transdermal route of its cargo molecule (TP). Our group previously proved that polyarginine and TD-34 have similar transdermal enhancing mechanism (changing the transdermal route of their cargo molecule); it is reasonably speculated that the transdermal route of DLCC-2 is the same as polyarginine and then changes the transdermal absorption route of TP. Furthermore, such results have laid a solid foundation for further investigation of CPPs and paved a way for both designing and synthesizing of new drug delivery system for therapy molecules.


Asunto(s)
Péptidos de Penetración Celular , Absorción Cutánea/efectos de los fármacos , Piel/metabolismo , Animales , Péptidos de Penetración Celular/síntesis química , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/farmacocinética , Péptidos de Penetración Celular/farmacología , Evaluación Preclínica de Medicamentos , Masculino , Ratones
11.
Cell ; 177(5): 1262-1279.e25, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31056284

RESUMEN

Ferroptosis, a non-apoptotic form of programmed cell death, is triggered by oxidative stress in cancer, heat stress in plants, and hemorrhagic stroke. A homeostatic transcriptional response to ferroptotic stimuli is unknown. We show that neurons respond to ferroptotic stimuli by induction of selenoproteins, including antioxidant glutathione peroxidase 4 (GPX4). Pharmacological selenium (Se) augments GPX4 and other genes in this transcriptional program, the selenome, via coordinated activation of the transcription factors TFAP2c and Sp1 to protect neurons. Remarkably, a single dose of Se delivered into the brain drives antioxidant GPX4 expression, protects neurons, and improves behavior in a hemorrhagic stroke model. Altogether, we show that pharmacological Se supplementation effectively inhibits GPX4-dependent ferroptotic death as well as cell death induced by excitotoxicity or ER stress, which are GPX4 independent. Systemic administration of a brain-penetrant selenopeptide activates homeostatic transcription to inhibit cell death and improves function when delivered after hemorrhagic or ischemic stroke.


Asunto(s)
Isquemia Encefálica , Péptidos de Penetración Celular/farmacología , Ferroptosis/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemorragias Intracraneales , Neuronas , Fosfolípido Hidroperóxido Glutatión Peroxidasa/biosíntesis , Selenio/farmacología , Accidente Cerebrovascular , Transcripción Genética/efectos de los fármacos , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Hemorragias Intracraneales/tratamiento farmacológico , Hemorragias Intracraneales/metabolismo , Hemorragias Intracraneales/patología , Masculino , Ratones , Neuronas/metabolismo , Neuronas/patología , Factor de Transcripción Sp1/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Factor de Transcripción AP-2/metabolismo
12.
J Invest Dermatol ; 139(2): 412-421, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30287285

RESUMEN

PAR2 has been proposed to contribute to lesion formation and intense itch in atopic dermatitis. Here, we tested the ability of a cell-penetrating pepducin, PZ-235, to mitigate the potentially deleterious effects of PAR2 in models of atopic dermatitis. PZ-235 significantly inhibited PAR2-mediated expression of inflammatory factors NF-κB, TSLP, TNF-α, and differentiation marker K10 by 94%-98% (P < 0.001) in human keratinocytes and suppressed IL-4 and IL-13 by 68%-83% (P < 0.05) in mast cells. In delayed pepducin treatment models of oxazolone- and DNFB-induced dermatitis, PZ-235 significantly attenuated skin thickening by 43%-100% (P < 0.01) and leukocyte crusting by 57% (P < 0.05), and it inhibited ex vivo chemotaxis of leukocytes toward PAR2 agonists. Daily PZ-235 treatment of filaggrin-deficient mice exposed to dust mite allergens for 8 weeks significantly suppressed total leukocyte and T-cell infiltration by 50%-68%; epidermal thickness by 60%-77%; and skin thickening, scaling, excoriation, and total lesion severity score by 46%-56%. PZ-235 significantly reduced itching caused by wasp venom peptide degranulation of mast cells in mice by 51% (P < 0.05), which was comparable to the protective effects conferred by PAR2 deficiency. Taken together, these results suggest that a PAR2 pepducin may confer broad therapeutic benefits as a disease-modifying treatment for atopic dermatitis and itch.


Asunto(s)
Péptidos de Penetración Celular/farmacología , Dermatitis Atópica/tratamiento farmacológico , Prurito/tratamiento farmacológico , Receptor PAR-2/antagonistas & inhibidores , Animales , Péptidos de Penetración Celular/uso terapéutico , Dermatitis Atópica/etiología , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Proteínas Filagrina , Humanos , Queratinocitos , Masculino , Ratones , Prurito/etiología , Prurito/patología , Receptor PAR-2/inmunología , Receptor PAR-2/metabolismo
13.
J Am Chem Soc ; 140(47): 16140-16151, 2018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30388366

RESUMEN

New strategies are urgently needed to target MRSA, a major global health problem and the leading cause of mortality from antibiotic-resistant infections in many countries. Here, we report a general approach to this problem exemplified by the design and synthesis of a vancomycin-d-octaarginine conjugate (V-r8) and investigation of its efficacy in addressing antibiotic-insensitive bacterial populations. V-r8 eradicated MRSA biofilm and persister cells in vitro, outperforming vancomycin by orders of magnitude. It also eliminated 97% of biofilm-associated MRSA in a murine wound infection model and displayed no acute dermal toxicity. This new dual-function conjugate displays enhanced cellular accumulation and membrane perturbation as compared to vancomycin. Based on its rapid and potent activity against biofilm and persister cells, V-r8 is a promising agent against clinical MRSA infections.


Asunto(s)
Antibacterianos/uso terapéutico , Biopelículas/efectos de los fármacos , Péptidos de Penetración Celular/uso terapéutico , Staphylococcus aureus Resistente a Meticilina/fisiología , Vancomicina/análogos & derivados , Vancomicina/uso terapéutico , Animales , Antibacterianos/síntesis química , Antibacterianos/farmacología , Antibacterianos/toxicidad , Línea Celular , Péptidos de Penetración Celular/síntesis química , Péptidos de Penetración Celular/farmacología , Péptidos de Penetración Celular/toxicidad , Diseño de Fármacos , Humanos , Masculino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Oligopéptidos/síntesis química , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Oligopéptidos/toxicidad , Vancomicina/farmacología , Vancomicina/toxicidad , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/fisiología
14.
Sci Rep ; 8(1): 12538, 2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30135446

RESUMEN

Cell penetrating peptides (CPPs) offer great potential to deliver therapeutic molecules to previously inaccessible intracellular targets. However, many CPPs are inefficient and often leave their attached cargo stranded in the cell's endosome. We report a versatile platform for the isolation of peptides delivering a wide range of cargos into the cytoplasm of cells. We used this screening platform to identify multiple "Phylomer" CPPs, derived from bacterial and viral genomes. These peptides are amenable to conventional sequence optimization and engineering approaches for cell targeting and half-life extension. We demonstrate potent, functional delivery of protein, peptide, and nucleic acid analog cargos into cells using Phylomer CPPs. We validate in vivo activity in the cytoplasm, through successful transport of an oligonucleotide therapeutic fused to a Phylomer CPP in a disease model for Duchenne's muscular dystrophy. This report thus establishes a discovery platform for identifying novel, functional CPPs to expand the delivery landscape of druggable intracellular targets for biological therapeutics.


Asunto(s)
Péptidos de Penetración Celular/farmacología , Sistemas de Liberación de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/métodos , Animales , Bacteriófago T7 , Biotinilación , Células CHO , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Péptidos de Penetración Celular/genética , Péptidos de Penetración Celular/toxicidad , Dicroismo Circular , Cricetulus , Modelos Animales de Enfermedad , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Microscopía Fluorescente , Distrofia Muscular de Duchenne/tratamiento farmacológico , Biblioteca de Péptidos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
15.
In Vitro Cell Dev Biol Anim ; 54(5): 335-345, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29560557

RESUMEN

The balance of oxidation and reduction in the body requires the synergistic effect of various antioxidant enzymes. Therefore, the construction of enzyme mimics with multiple antioxidant activities is important and beneficial for further research on the synergistic effects of antioxidant enzymes and their mechanism of action. To explore the synergistic effect of superoxide dismutase (SOD) and glutathione peroxidase (GPx), a 76-mer selenium-containing peptide (Se-76P) mimic containing the active SOD and GPx centers was designed. Moreover, a cell-penetrating peptide was introduced into Se-76P by structure modeling, and then, Se-76P was expressed by a single-protein production combined with the cysteine auxotrophic double-expression system of Escherichia coli. The results suggest that Se-76P exhibits SOD and GPx activities, following the GPx activity of 109 U/mg protein and the SOD activity of 1218 U/mg protein. The labeled Se-76P with FITC fluorescence was verified to enter the L02 cells successfully; it improved the antioxidant activity in cells and promoted the consumption of glucose and synthesis of glycogen. The injection of Se-76P subcutaneously decreased the levels of blood glucose and malondialdehyde of lipid peroxidation produced in mice, indicating that Se-76P had antioxidative properties and a certain regulatory role of glucose metabolism. The data analysis provides further clarification that Se-76P can regulate insulin signal transduction to play an insulin-like role, which not only has a greater significance for further elucidating the catalytic mechanism of the enzyme and their synergistic effects on each other but also has enormous medicinal potential.


Asunto(s)
Péptidos de Penetración Celular/química , Insulina/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Péptidos/química , Antioxidantes/química , Antioxidantes/metabolismo , Metabolismo de los Hidratos de Carbono/genética , Dominio Catalítico/efectos de los fármacos , Péptidos de Penetración Celular/farmacología , Glucosa/metabolismo , Glutatión Peroxidasa/química , Humanos , Insulina/química , Oxidación-Reducción/efectos de los fármacos , Péptidos/genética , Péptidos/farmacología , Selenio/química , Superóxido Dismutasa/química
16.
Adv Healthc Mater ; 7(11): e1800121, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29582583

RESUMEN

Autophagy is a homeostatic lysosome-dependent metabolic process to eliminate damaged or dysfunctional cellular organelles, which is closely associated with tumor progression. Indocyanine green (ICG) can convert NIR light energy to localized heat for cancer cell and tissue ablation. However, the effect of autophagy modulation on ICG-mediated photothermal therapy remains unknown. In this study, it is found that primaquine (PQ) suppresses autophagy flux at a late stage through the impeding fusion of the autophagosome with the lysosome to form an autophagolysosome, leading to cell apoptosis or necrosis. This autophagosome-lysosome fusion inhibitory effect and the autophagosome accumulation are more evident in the photothermal therapy combined with autophagy inhibition. Motivated by this notable effect, a cascade-targeting nanocapsule (HCP) is constructed using an organic solvent-free strategy to coencapsulate PQ and ICG. By targeting the cluster designation 44 molecule and sequentially enhancing the cell-penetrating peptide-mediated endocytosis, the codelivery of PQ/ICG by HCP achieves selective recognition and reinforces the internalization by MCF-7 cells to exert a synergistic therapeutic effect on MCF-7 cells both in vitro and in vivo. The HCP system for the photothermal and autophagy inhibition combination therapy represents a novel strategy for the treatment of breast cancer.


Asunto(s)
Autofagia/efectos de los fármacos , Neoplasias de la Mama/terapia , Péptidos de Penetración Celular , Hipertermia Inducida , Nanocápsulas , Fototerapia , Primaquina , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/farmacocinética , Péptidos de Penetración Celular/farmacología , Endocitosis/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanocápsulas/química , Nanocápsulas/uso terapéutico , Primaquina/química , Primaquina/farmacocinética , Primaquina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Res Vet Sci ; 114: 502-510, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28987957

RESUMEN

The absence of an effective therapy against human solid tumors has fostered the development of promising antineoplastic therapeutic candidates, as the CIGB-552 peptide. This synthetic peptide has shown to be effective in reducing tumor size and increasing the lifespan in tumor-bearing mice. Therefore, this work was aimed to explore the safety profile and preliminary assessment of antitumor activity of the CIGB-552 peptide therapeutic candidate in a small population of dogs (n=9) having malignant spontaneously-arising solid tumors. The peptide was administered by subcutaneous (s.c.) route, at three dosage levels (0.075, 0.15 and 0.3mg/kg). The results showed no dose-limiting toxicities in any dogs. The antitumor activity observed in dogs receiving CIGB-552 was associated with the reduction in the tumor volume. Given the antitumor effects of CIGB-552 as mediated by COMMD1 protein, which function is highly conserved among eukaryotic organisms, and the similarities of canine and human types of cancer with respect to tumor biology, it is likely that CIGB-552 could demonstrate comparable anti-cancer activity in human patients. Synthetic peptide, COMMD1, Tumor, Dog, CIGB-552.


Asunto(s)
Antineoplásicos/farmacología , Péptidos de Penetración Celular/farmacología , Enfermedades de los Perros/tratamiento farmacológico , Neoplasias/veterinaria , Animales , Antineoplásicos/administración & dosificación , Péptidos de Penetración Celular/administración & dosificación , Perros , Evaluación Preclínica de Medicamentos/veterinaria , Femenino , Masculino , Neoplasias/tratamiento farmacológico
18.
Sci Rep ; 7(1): 12352, 2017 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-28955044

RESUMEN

Cell penetrating peptide derived from human eosinophil cationic protein (CPPecp) is a 10-amino-acid peptide containing a core heparan sulfate (HS)-binding motif of human eosinophil cationic protein (ECP). It binds and penetrates bronchial epithelial cells without cytotoxic effects. Here we investigated airway-protective effects of CPPecp in BEAS-2B cell line and mite-induced airway allergic inflammation in BALB/c mice. In BEAS-2B cell, CPPecp decreases ECP-induced eotaxin mRNA expression. CPPecp also decreases eotaxin secretion and p-STAT6 activation induced by ECP, as well as by IL-4. In vivo studies showed CPPecp decreased mite-induced airway inflammation in terms of eosinophil and neutrophil count in broncho-alveolar lavage fluid, peri-bronchiolar and alveolar pathology scores, cytokine production in lung protein extract including interleukin (IL)-5, IL-13, IL-17A/F, eotaxin; and pause enhancement from methacholine stimulation. CPPecp treated groups also showed lower serum mite-specific IgE level. In this study, we have demonstrated the in vitro and in vivo anti-asthma effects of CPPecp.


Asunto(s)
Antiasmáticos/farmacología , Asma/tratamiento farmacológico , Péptidos de Penetración Celular/farmacología , Proteína Catiónica del Eosinófilo/química , Mucosa Respiratoria/efectos de los fármacos , Alérgenos/inmunología , Animales , Antiasmáticos/uso terapéutico , Antígenos Dermatofagoides/inmunología , Asma/inmunología , Asma/patología , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/inmunología , Línea Celular , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/uso terapéutico , Citocinas/inmunología , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Eosinófilos/inmunología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Resultado del Tratamiento
19.
J Drug Target ; 25(6): 523-531, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28140690

RESUMEN

Notch pathway was found to be activated in most glioblastomas (GBMs), underlining the importance of Notch in formation and recurrence of GBM. In this study, a Notch inhibitory peptide, dominant negative MAML (dnMAML), was conjugated to elastin-like polypeptide (ELP) for tumor targeted delivery. ELP is a thermally responsive polypeptide that can be actively and passively targeted to the tumor site by localized application of hyperthermia. This complex was further modified with the addition of a cell penetrating peptide, SynB1, for improved cellular uptake and blood-brain barrier penetration. The SynB1-ELP1-dnMAML was examined for its cellular uptake, cytotoxicity, apoptosis, cell cycle inhibition and the inhibition of target genes' expression. SynB1-ELP1-dnMAML inhibited the growth of D54 and U251 cells by inducing apoptosis and cell cycle arrest, especially in the presence of hyperthermia. Hyperthermia increased overall uptake of the polypeptide by the cells and enhanced the resulting pharmacological effects of dnMAML, showing the inhibition of targets of Notch pathway such as Hes-1 and Hey-L. These results confirm that dnMAML is an effective Notch inhibitor and combination with ELP may allow thermal targeting of the SynB1-ELP1-dnMAML complex in cancer cells while avoiding the dangers of systemic Notch inhibition.


Asunto(s)
Péptidos de Penetración Celular/administración & dosificación , Proteínas de Unión al ADN/administración & dosificación , Glioblastoma/tratamiento farmacológico , Receptores Notch/antagonistas & inhibidores , Factores de Transcripción/administración & dosificación , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Péptidos de Penetración Celular/farmacología , Proteínas de Unión al ADN/farmacocinética , Proteínas de Unión al ADN/farmacología , Sistemas de Liberación de Medicamentos , Elastina/administración & dosificación , Glioblastoma/patología , Humanos , Hipertermia Inducida/métodos , Péptidos/administración & dosificación , Factores de Transcripción/farmacocinética , Factores de Transcripción/farmacología
20.
J Biomed Nanotechnol ; 12(5): 878-93, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27305812

RESUMEN

A novel nanomaterial with precisely-defined size and shape, biocompatible composition, and excellent stability, which can integrate multi modal targeted imaging and therapy into a single system for visualized therapeutics, has recently attracted significant research interest. Here, we developed a multifunctional nanoplatform based on silica-coated 4-mercaptobenzoic acid-modified gold nanorods (Au NRs) decorated with gold nanoclusters rich in the photosensitizer Ce6 (Au-Ce6 NCs). The nanoparticles also comprised folic acid and cell penetrating peptide molecules anchored on the surface, obtaining the Au@SiO2@Au-cell penetrating peptide nanocomposite. The Au-Ce6 NCs enhanced the photophysical stability, provided numerous bonding sites and offered a large surface-area and interior space to achieve a high drug loading efficiency (up to 55%). The anchored folic acid and cell penetrating peptide synergistically enhanced the efficiency of uptake of nanocomposites by HeLa cells (up to 70.7%) and improved therapeutic efficacy. The nanocomposite also has good water-solubility, excellent biocompatibility, and long-term stability against illumination and exposure to pH 3-12, thus facilitating their bioapplications in cancer theranostics. Here, the nanocomposite was established for high-resolution and noninvasive tri-modal surface-enhanced Raman spectrum/dark-field/fluorescence imaging-guided high-efficiency synergistic photodynamic/photothermal therapy of cancer. Our studies demonstrate that the multifunctional nanocomposite has the potential as a novel and sensitive contrast agent for complementary and synergistic theranostics in the clinic.


Asunto(s)
Antineoplásicos/farmacología , Péptidos de Penetración Celular/farmacología , Ácido Fólico/farmacología , Fototerapia , Nanomedicina Teranóstica , Muerte Celular/efectos de los fármacos , Sinergismo Farmacológico , Oro/química , Células HeLa , Calor , Humanos , Imagenología Tridimensional , Espacio Intracelular/metabolismo , Nanocompuestos/química , Nanocompuestos/ultraestructura , Imagen Óptica , Tamaño de la Partícula , Dióxido de Silicio/química , Espectrometría de Fluorescencia , Espectrofotometría Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA