Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biometals ; 36(3): 639-655, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36626098

RESUMEN

Liver cancer and leukemia are the fourth and first causes, respectively, of cancer death in children and adults worldwide. Moreover, cancer treatments, although beneficial, remain expensive, invasive, toxic, and affect the patient's quality of life. Therefore, new anticancer agents are needed to improve existing agents. Because bovine lactoferrin (bLF) and its derived peptides have antitumor properties, we investigated the anticancer effect of bLF and LF peptides (LFcin17-30, LFampin265-284 and LFchimera) on liver cancer HepG2 cells and leukemia Jurkat cells. HepG2 and Jurkat cells were incubated with bLF and LF peptides. Cell proliferation was quantified by an MTT assay, and cell morphology and damage were visualized by light microscopy or by phalloidin-TRITC/DAPI staining. The discrimination between apoptosis/necrosis was performed by staining with Annexin V-Alexa Fluor 488 and propidium iodide, and the expression of genes related to apoptosis was analyzed in Jurkat cells. Finally, the synergistic interaction of bLF and LF peptides with cisplatin or etoposide was assessed by an MTT assay and the combination index. The present study demonstrated that bLF and LF peptides inhibited the viability of HepG2 and Jurkat cells, inducing damage to the cell monolayer of HepG2 cells and morphological changes in both cell lines. bLF, LFcin17-30, and LFampin265-284 triggered apoptosis in both cell lines, whereas LFchimera induced necrosis. These results suggested that bLF and LF peptides activate apoptosis by increasing the expression of genes of the intrinsic pathway. Additionally, bLF and LF peptides synergistically interacted with cisplatin and etoposide. In conclusion, bLF and LF peptides display anticancer activity against liver cancer and leukemia cells, representing an alternative or improvement in cancer treatment.


Asunto(s)
Lactoferrina , Neoplasias Hepáticas , Niño , Humanos , Lactoferrina/farmacología , Lactoferrina/química , Células Jurkat , Células Hep G2 , Cisplatino , Etopósido , Calidad de Vida , Péptidos/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Necrosis
2.
Biochem Cell Biol ; 99(1): 149-158, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33307991

RESUMEN

Cervical, uterine, and ovarian cancers are the most common malignancies of the female genital tract worldwide. Despite advances in prevention, early diagnosis, effective screening, and treatment programs, mortality remains high. Consequently, it is important to search for new treatments. The activity of bovine lactoferrin (bLF) and LF peptides against several types of cancer has been studied; however, only a few studies report the effect of bLF and LF peptides against cervical and endometrial cancers. In this study, we explored the effect of bLF as well as LF chimera and its constituent peptides LFcin17-30 and LFampin265-284 on the viability of cervical (HeLa, SiHa) and endometrial (KLE, HEC-1A) cancer cell lines. Cell proliferation was quantified with an MTT assay, cell morphological changes and damage were determined by Giemsa and phalloidin-TRITC and DAPI staining, and apoptotic and necrotic cells were identified by Alexa Fluor® 488 Annexin V and propidium iodide staining. Additionally, the effect of combinations of bLF and LF peptides with cisplatin was assessed. bLF and LF peptides inhibited the proliferation of uterine cancer cells and caused cellular morphological changes and damage to cell monolayers. bLF induced apoptosis, LFcin17-30 and LFampin265-284 induced apoptosis and necrosis, and LF chimera induced necrosis. Additionally, bLF and LF chimera showed an additive interaction with cisplatin against uterine cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Lactoferrina/metabolismo , Fragmentos de Péptidos/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Bovinos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Lactoferrina/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
3.
Biofouling ; 36(9): 1117-1128, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33297738

RESUMEN

Chronic rhinosinusitis (CRS) is a chronic infection of the nasal cavity and paranasal sinuses associated with the presence of a microbial biofilm. Extracellular DNA (eDNA) is an important component of the biofilm matrix. Antimicrobial peptides (AMPs) are natural peptides with the ability to kill microorganisms. D-LL-31 is a synthetic variant of the AMP cathelicidin with increased resistance to proteolytic breakdown. In this study it is shown for 3 clinical CRS isolates that treatment of 24 h biofilms with DNase I enhanced the antimicrobial activity of D-LL-31. Conversely, co-incubation of D-LL-31 at the IC50 value with exogenous DNA resulted in reduced antimicrobial activity. DNase I alone did not show antimicrobial activity against the isolates tested but caused dispersal of an established biofilm. Hence, the presence of eDNA in the biofilm matrix reduced AMP-mediated killing. These results suggest that combination therapy with proteolysis resistant AMP D-LL-31 and DNase could be considered for effective treatment of CRS.


Asunto(s)
Biopelículas , Antibacterianos , Bacterias/genética , Desoxirribonucleasa I , Desoxirribonucleasas
4.
Biofouling ; 35(5): 573-584, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31282211

RESUMEN

Melioidosis is a severe disease caused by Burkholderia pseudomallei. The biofilm of B. pseudomallei acquires resistance to several antibiotics and may be related to relapse in melioidosis patients. Here, the killing activity of antimicrobial peptides (LL-37, LL-31) and the D-enantiomers (D-LL-37, D-LL-31) in combination with ceftazidime (CAZ) against B. pseudomallei 1026b, H777 and a biofilm mutant M10, derived from H777 grown under biofilm-stimulating conditions was observed. Using static conditions, D-LL-31 exhibited the strongest killing activity against the three isolates in a dose-dependent manner. IC50 values for D-LL-31 ranged from 1 to 6 µM, for isolates M10, H777, and 1026b, respectively. Moreover, D-LL-31 combined with CAZ synergistically decreased the IC50 values of the peptide and antibiotic and caused also disruption of biofilms of B. pseudomallei 1026b under flow conditions. Thus a combination of D-LL-31 and CAZ may enhance the efficacy of the currently used antibiotic treatments against B. pseudomallei.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Burkholderia pseudomallei/efectos de los fármacos , Catelicidinas/farmacología , Ceftazidima/farmacología , Péptidos/farmacología , Burkholderia pseudomallei/fisiología , Pruebas de Sensibilidad Microbiana
5.
Biol Chem ; 399(12): 1409-1419, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30138105

RESUMEN

Histatins are histidine-rich peptides present in the saliva of humans and higher primates and have been implicated in the protection of the oral cavity. Histatin 1 is one of the most abundant histatins and recent reports show that it has a stimulating effect on cellular adherence, thereby suggesting a role in maintaining the quality of the epithelial barrier and stimulating mesenchymal-to-epithelial transition. Here we summarize these findings and discuss them in the context of previous reports. The recent findings also provide new insights in the physiological functions of histatin 1, which are discussed here. Furthermore, we put forward a possible role of histatin 1 in various pathologies and its potential function in clinical applications.


Asunto(s)
Transición Epitelial-Mesenquimal , Histatinas/metabolismo , Secuencia de Aminoácidos , Adhesión Celular , Histatinas/química , Histatinas/genética , Humanos
6.
FASEB J ; 31(9): 3922-3933, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28522595

RESUMEN

Histatins are multifunctional histidine-rich peptides secreted by the salivary glands and exclusively present in the saliva of higher primates, where they play a fundamental role in the protection of the oral cavity. Our previously published results demonstrated that histatin-1 (Hst1) promotes cell-substrate adhesion in various cell types and hinted that it could also be involved in cell-cell adhesion, a process of fundamental importance to epithelial and endothelial barriers. Here we explore the effects of Hst1 on cellular barrier function. We show that Hst1 improved endothelial barrier integrity, decreased its permeability for large molecules, and prevented translocation of bacteria across epithelial cell layers. These effects are mediated by the adherens junction protein E-cadherin (E-cad) and by the tight junction protein zonula occludens 1, as Hst1 increases the levels of zonula occludens 1 and of active E-cad. Hst1 may also promote epithelial differentiation as Hst1 induced transcription of the epithelial cell differentiation marker apolipoprotein A-IV (a downstream E-cad target). In addition, Hst1 counteracted the effects of epithelial-mesenchymal transition inducers on the outgrowth of oral cancer cell spheroids, suggesting that Hst1 affects processes that are implicated in cancer progression.-Van Dijk, I. A., Ferrando, M. L., van der Wijk, A.-E., Hoebe, R. A., Nazmi, K., de Jonge, W. J., Krawczyk, P. M., Bolscher, J. G. M., Veerman, E. C. I., Stap, J. Human salivary peptide histatin-1 stimulates epithelial and endothelial cell adhesion and barrier function.


Asunto(s)
Células Endoteliales/fisiología , Células Epiteliales/fisiología , Regulación de la Expresión Génica/fisiología , Histatinas/metabolismo , Línea Celular , Histatinas/genética , Humanos
7.
Biometals ; 31(6): 941-950, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30136243

RESUMEN

Yersinia pestis is the causative agent of plague. As adequate antibiotic treatment falls short and currently no effective vaccine is available, alternative therapeutic strategies are needed. In order to contribute to solving this problem we investigated the therapeutic potential of the peptide construct LFchimera against the safer-to-handle Y. pestis simulants Yersinia enterocolitica and Yersinia pseudotuberculosis in vitro. LFchimera is a heterodimeric peptide construct mimicking two antimicrobial domains of bovine lactoferrin, i.e. lactoferrampin and lactoferricin. LFchimera has been shown to be a potent antimicrobial peptide against a variety of bacteria in vitro and in vivo. Also Y. enterocolitica and Y. pseudotuberculosis have been shown to be susceptible for LFchimera in vitro. As Yersiniae spp. adhere to and invade host cells upon infection, we here investigated the effects of LFchimera on these processes. It was found that LFchimera has the capacity to inhibit host-cell invasion by Yersiniae spp. in vitro. This effect appeared to be host-cell mediated, not bacteria-mediated. Furthermore it was found that exposure of human HeLa epithelial cells to both LFchimera and the bacterial strains evoked a pro-inflammatory cytokine release from the cells in vitro.


Asunto(s)
Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Yersinia/efectos de los fármacos , Antibacterianos/química , Péptidos Catiónicos Antimicrobianos/química , Adhesión Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Pruebas de Sensibilidad Microbiana , Relación Estructura-Actividad
8.
Biochem Cell Biol ; 95(1): 91-98, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28165293

RESUMEN

Lactoferrin (LF) is a mammalian host defense glycoprotein with diverse biological activities. Peptides derived from the cationic region of LF possess cytotoxic activity against cancer cells in vitro and in vivo. Bovine lactoferricin (LFcinB), a peptide derived from bovine LF (bLF), exhibits broad-spectrum anticancer activity, while a similar peptide derived from human LF (hLF) is not as active. In this work, several peptides derived from the N-terminal regions of bLF and hLF were studied for their anticancer activities against leukemia and breast-cancer cells, as well as normal peripheral blood mononuclear cells. The cyclized LFcinB-CLICK peptide, which possesses a stable triazole linkage, showed improved anticancer activity, while short peptides hLF11 and bLF10 were not cytotoxic to cancer cells. Interestingly, hLF11 can act as a cell-penetrating peptide; when combined with the antimicrobial core sequence of LFcinB (RRWQWR) through either a Pro or Gly-Gly linker, toxicity to Jurkat cells increased. Together, our work extends the library of LF-derived peptides tested for anticancer activity, and identified new chimeric peptides with high cytotoxicity towards cancerous cells. Additionally, these results support the notion that short cell-penetrating peptides and antimicrobial peptides can be combined to create new adducts with increased potency.


Asunto(s)
Antiinfecciosos/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Lactoferrina/farmacología , Fragmentos de Péptidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Bovinos , Femenino , Hemólisis/efectos de los fármacos , Humanos , Células Jurkat , Células Tumorales Cultivadas
9.
Biochem Cell Biol ; 95(1): 76-81, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28165291

RESUMEN

Lactoferrin (LF) is a protein with antimicrobial activity, which is conferred in part by 2 regions contained in its N-terminal lobe. These regions have been used to develop the following synthetic peptides: lactoferricin17-30, lactoferrampin265-284, and LF chimera (a fusion of lactoferricin17-30 and lactoferrampin265-284). We have reported that these LF peptides have antibacterial activity against several pathogenic bacteria; however, the exact mechanism of action has not been established. Here, we report the effects of LF peptides on the viability of enteroaggregative Escherichia coli (EAEC) and the ability of these peptides to penetrate into the bacteria cytoplasm. The viability of EAEC treated with LF peptides was determined via enumeration of colony-forming units, and the binding and internalization of the LF peptides was followed via immunogold labeling and electron microscopy. Treatment of EAEC with 20 and 40 µmol/L LF peptides reduced bacterial growth compared with untreated bacteria. Initially the peptides associated with the plasma membrane, but after 5 to 30 min of incubation, the peptides were found in the cytoplasm. Remarkably, bacteria treated with LF chimera developed cytosolic electron-dense structures that contained the antimicrobial peptide. Our results suggest that the antibacterial mechanism of LF peptides on EAEC involves their interaction with and penetration into the bacteria.


Asunto(s)
Antibacterianos/farmacología , Péptidos de Penetración Celular/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Escherichia coli/efectos de los fármacos , Lactoferrina/farmacología , Escherichia coli/aislamiento & purificación , Infecciones por Escherichia coli/microbiología , Humanos
10.
Biochem Cell Biol ; 95(1): 82-90, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28165283

RESUMEN

Giardia intestinalis is the most common infectious protozoan parasite in children. Despite the effectiveness of some drugs, the disease remains a major worldwide problem. Consequently, the search for new treatments is important for disease eradication. Biological molecules with antimicrobial properties represent a promising alternative to combat pathogens. Bovine lactoferrin (bLF) is a key component of the innate host defense system, and its peptides have exhibited strong antimicrobial activity. Based on these properties, we evaluated the parasiticidal activity of these peptides on G. intestinalis. Trophozoites were incubated with different peptide concentrations for different periods of time, and the growth or viability was determined by carboxyfluorescein-succinimidyl-diacetate-ester (CFDA) and propidium iodide (PI) staining. Endocytosis of peptides was investigated by confocal microscopy, damage was analyzed by transmission and scanning electron microscopy, and the type of programmed cell death was analyzed by flow cytometry. Our results showed that the LF peptides had giardicidal activity. The LF peptides interacted with G. intestinalis and exposure to LF peptides correlated with an increase in the granularity and vacuolization of the cytoplasm. Additionally, the formation of pores, extensive membrane disruption, and programmed cell death was observed in trophozoites treated with LF peptides. Our results demonstrate that LF peptides exhibit potent in vitro antigiardial activity.


Asunto(s)
Antiinfecciosos/farmacología , Giardia lamblia/efectos de los fármacos , Giardiasis/tratamiento farmacológico , Lactoferrina/farmacología , Fragmentos de Péptidos/farmacología , Trofozoítos/efectos de los fármacos , Animales , Bovinos , Supervivencia Celular/efectos de los fármacos , Heces/parasitología , Giardia lamblia/crecimiento & desarrollo , Giardia lamblia/aislamiento & purificación , Giardiasis/parasitología , Humanos
11.
World J Microbiol Biotechnol ; 33(9): 167, 2017 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-28823045

RESUMEN

Lactoferrin chimera (LFchimera), a heterodimeric peptide containing lactoferrampin (LFampin265-284) and a part of lactoferricin (LFcin17-30), possesses a broad spectrum of antimicrobial activity. However, there is no report on the inhibitory effects of LFchimera against multispecies oral biofilms. This study aimed to determine the effects of LFchimera in comparison to chlorhexidine digluconate (CHX) and minocycline hydrochloride (MH), on in vitro multispecies biofilms derived from subgingival plaque of periodontitis patients harboring Aggregatibacter actinomycetemcomitans. First the effects of LFchimera against planktonic and an 1-day old biofilm of the periodontopathic bacteria, A. actinomycetemcomitans ATCC 43718 were established. Then, the effects on biofilm formation and bacterial viability in the multispecies biofilm were determined by crystal violet staining and LIVE/DEAD BacLight Bacterial Viability kit, respectively. The results revealed that a significant reduction (P < 0.05) in biofilm formation occurred after 15 min exposure to 20 µM of LFchimera or CHX compared to control. In contrast, MH at concentration up to 100 µM did not inhibit biofilm formation. The ratio of live/dead bacteria in biofilm was also significantly lower after 15 min exposure to 20 µM of LFchimera compared to control and 20-50 µM of CHX and MH. Altogether, the results obtained indicate that LFchimera is able to inhibit in vitro subgingival biofilm formation and reduce viability of multispecies bacteria in biofilm better than CHX and MH.


Asunto(s)
Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Biopelículas/efectos de los fármacos , Periodontitis/microbiología , Aggregatibacter actinomycetemcomitans/efectos de los fármacos , Aggregatibacter actinomycetemcomitans/fisiología , Humanos , Técnicas In Vitro , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Plancton/efectos de los fármacos
12.
World J Microbiol Biotechnol ; 33(1): 3, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27832504

RESUMEN

Lactoferrin (LF) is an important immune protein in neutrophils and secretory fluids of mammals. Bovine LF (bLF) harbours two antimicrobial stretches, lactoferricin and lactoferampin, situated in close proximity in the N1 domain. To mimic these antimicrobial domain parts a chimeric peptide (LFchimera) has been constructed comprising parts of both stretches (LFcin17-30 and LFampin265-284). To investigate the potency of this construct to combat a set of Gram positive and Gram negative bacteria which are regarded as simulants for biological warfare agents, the effect on bacterial killing, membrane permeability and membrane polarity were determined in comparison to the constituent peptides and the native bLF. Furthermore we aimed to increase the antimicrobial potency of the bLF derived peptides by cationic amino acid substitutions. Overall, the bactericidal activity of the peptides could be related to membrane disturbing effects, i.e. membrane permeabilization and depolarization. Those effects were most prominent for the LFchimera. Arginine residues were found to be crucial for displaying antimicrobial activity, as lysine to arginine substitutions resulted in an increased antimicrobial activity, affecting mostly LFampin265-284 whereas arginine to lysine substitutions resulted in a decreased bactericidal activity, predominantly in case of LFcin17-30.


Asunto(s)
Antibacterianos/farmacología , Lactoferrina/síntesis química , Lactoferrina/farmacología , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/farmacología , Sustitución de Aminoácidos , Animales , Armas Biológicas , Bovinos , Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Lactoferrina/química , Pruebas de Sensibilidad Microbiana
13.
FASEB J ; 29(8): 3124-32, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25903106

RESUMEN

Histatins (Hsts) are histidine-rich peptides exclusively present in the saliva of higher primates. In this study, we explored the effects of Hsts on cell-substrate and cell-cell adhesion. Histatin (Hst)-1 caused a significant (>2-fold) increase (EC50 = 1 µM) in the ability of human adherent cells to attach and spread, even in conditions that impaired cell spreading. Other tested Hsts did not stimulate cell spreading, indicating a specific effect of Hst1. The effect of Hst1 on cell-cell adhesion was investigated by using transepithelial resistance (TER) measurements in the human cell line Caco-2, a widely used model for the epithelial layer. We found that 10 µM Hst1 caused a 20% increase in TER compared to the negative control, indicating a function for Hst1 in intercellular cell adhesion and epithelial integrity. A role for Hst1 in both cell-substrate and cell-cell adhesion is highly conceivable, because these 2 modes of adhesion are closely related via shared components and connected signaling pathways.


Asunto(s)
Adhesión Celular/fisiología , Histatinas/metabolismo , Histidina/metabolismo , Péptidos/metabolismo , Saliva/metabolismo , Proteínas y Péptidos Salivales/metabolismo , Células CACO-2 , Línea Celular , Línea Celular Tumoral , Células Epiteliales/metabolismo , Humanos
14.
Wound Repair Regen ; 24(1): 100-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26542883

RESUMEN

Oral wounds heal faster and with better scar quality than skin wounds. Deep skin wounds where adipose tissue is exposed, have a greater risk of forming hypertrophic scars. Differences in wound healing and final scar quality might be related to differences in mesenchymal stromal cells (MSC) and their ability to respond to intrinsic (autocrine) and extrinsic signals, such as human salivary histatin, epidermal growth factor, and transforming growth factor beta1. Dermis-, adipose-, and gingiva-derived MSC were compared for their regenerative potential with regards to proliferation, migration, and matrix contraction. Proliferation was assessed by cell counting and migration using a scratch wound assay. Matrix contraction and alpha smooth muscle actin was assessed in MSC populated collagen gels, and also in skin and gingival full thickness tissue engineered equivalents (reconstructed epithelium on MSC populated matrix). Compared to skin-derived MSC, gingiva MSC showed greater proliferation and migration capacity, and less matrix contraction in full thickness tissue equivalents, which may partly explain the superior oral wound healing. Epidermal keratinocytes were required for enhanced adipose MSC matrix contraction and alpha smooth muscle actin expression, and may therefore contribute to adverse scarring in deep cutaneous wounds. Histatin enhanced migration without influencing proliferation or matrix contraction in all three MSC, indicating that salivary peptides may have a beneficial effect on wound closure in general. Transforming growth factor beta1 enhanced contraction and alpha smooth muscle actin expression in all three MSC types when incorporated into collagen gels. Understanding the mechanisms responsible for the superior oral wound healing will aid us to develop advanced strategies for optimal skin regeneration, wound healing and scar formation.


Asunto(s)
Tejido Adiposo/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Dermis/fisiología , Encía/fisiología , Queratinocitos/fisiología , Células Madre Mesenquimatosas/fisiología , Cicatrización de Heridas/fisiología , Actinas , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dermis/citología , Dermis/efectos de los fármacos , Matriz Extracelular , Factor 6 de Crecimiento de Fibroblastos/farmacología , Encía/citología , Encía/efectos de los fármacos , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Regeneración/fisiología
15.
World J Microbiol Biotechnol ; 32(2): 33, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26754671

RESUMEN

Lactoferrin chimera (LFchimera), a hybrid peptide containing the two antimicrobial stretches of the innate immunity factor bovine lactoferrin, viz. LFampin265-284 and LFcin17-30, has strikingly high antimicrobial activity against the category B pathogen Burkholderia pseudomallei. The action mechanisms of LFchimera against B. pseudomallei is not fully understood. The aim of this study was to further investigate the effect of treated B. pseudomallei with LFchimera using (immune) electron microscopy. The effects of LFchimera on biofilm formation and against preformed biofilm of B. pseudomallei were also determined. After exposure to LFchimera, transmission electron microscopy revealed swelling of the periplasmic space of B. pseudomallei and a highly inhomogeneous electron density in the intracellular DNA region. Localization of LFchimera in B. pseudomallei using immunoelectron microscopy showed gold particles in intracellular structures without accumulation on the membranes. LFchimera also possessed stronger bactericidal activity than ceftazidime against B. pseudomallei grown in biofilm. Moreover, limited exposure of B. pseudomallei to LFchimera at subcidal concentration could reduce biofilm formation. Altogether, the results indicate that LFchimera possesses antibacterial and antibiofilm activities and can modulate B. pseudomallei colonization. Therefore, the efficacy of LFchimera merits further development of this agent for the therapy of melioidosis.


Asunto(s)
Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Biopelículas/efectos de los fármacos , Burkholderia pseudomallei/efectos de los fármacos , Burkholderia pseudomallei/ultraestructura , Secuencia de Aminoácidos , Animales , Burkholderia pseudomallei/fisiología , Bovinos , Ceftazidima/farmacología , Membrana Celular/efectos de los fármacos , Melioidosis/terapia , Pruebas de Sensibilidad Microbiana , Microscopía Electrónica
16.
Biochemistry ; 54(17): 2785-98, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25884905

RESUMEN

Lung cysteine cathepsins B, K, L, and S contribute to physiological and pathological processes including degradation of antimicrobial peptides/proteins (AMPs) such as surfactant protein SP-A, lactoferrin, secretory leukocyte peptidase inhibitor, and beta-defensins-2 and -3. Substantial amounts of uncleaved LL-37, a 37-mer cationic AMP, were observed in the sputum of patients with cystic fibrosis (CF). Nevertheless LL-37 was degraded after prolonged incubation in CF sputum, and the hydrolysis was blocked by E-64, a selective inhibitor of cysteine proteases. Cathepsins K and S, expressed in human alveolar macrophages, thoroughly hydrolyzed LL-37 in vitro, whereas it competitively inhibited cathepsin L (Ki = 150 nM). Cleavage of LL-37 by cathepsins S and K impaired its antimicrobial activity against Pseudomonas aeruginosa and Staphylococcus aureus, in a time- and concentration-dependent manner. The exchange of residues 67 and 205 in the S2 pockets of cathepsins L (Leu67Tyr/Ala205Leu) and K (Tyr67Leu/Leu205Ala) switched the specificity of these mutants toward LL-37. Molecular modeling suggested that LL-37 interacted with the active site of cathepsin L in both forward (i.e., substrate-like) and reverse orientations with similar binding energies. Our data support the hypothesis that cysteine cathepsins modulate the innate immunity response by degrading distinct and representative members of the AMP family.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Catepsina K/metabolismo , Catepsina L/antagonistas & inhibidores , Catepsinas/metabolismo , Inhibidores de Cisteína Proteinasa/metabolismo , Secuencia de Aminoácidos , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Líquido del Lavado Bronquioalveolar , Dicroismo Circular , Inhibidores de Cisteína Proteinasa/farmacología , Fibrosis Quística/microbiología , Humanos , Macrófagos Alveolares/metabolismo , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Pseudomonas aeruginosa/efectos de los fármacos , Especificidad por Sustrato , Catelicidinas
17.
Biol Chem ; 396(4): 283-93, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25581753

RESUMEN

Backbone cyclization has a profound impact on the biological activity and thermal and proteolytic stability of proteins and peptides. Chemical methods for cyclization are not always feasible, especially for large peptides or proteins. Recombinant Staphylococcus aureus sortase A shows potential as a new tool for the cyclization of both proteins and peptides. In this review, the scope and background of the sortase-mediated cyclization are discussed. High efficiency, versatility, and easy access make sortase A a promising cyclization tool, both for recombinant and chemo-enzymatic production methods.


Asunto(s)
Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Péptidos Cíclicos/metabolismo , Péptidos/metabolismo , Proteínas/metabolismo , Staphylococcus aureus/enzimología , Secuencia de Aminoácidos , Aminoaciltransferasas/química , Proteínas Bacterianas/química , Secuencia de Carbohidratos , Ciclización , Cisteína Endopeptidasas/química , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Péptidos Cíclicos/química , Peptidoglicano/química , Peptidoglicano/metabolismo , Conformación Proteica , Ingeniería de Proteínas , Proteínas/química , Proteínas Recombinantes/metabolismo , Staphylococcus aureus/metabolismo
18.
Clin Oral Investig ; 19(8): 1931-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25689982

RESUMEN

OBJECTIVES: The aim of this study was to examine levels of salivary mucins in children with deciduous and mixed dentition and to determine correlations between salivary mucins and dental caries status in two dentition stages. MATERIALS AND METHODS: Saliva samples were collected from preschool children with deciduous dentition aged between 4 and 6 years (n = 60) and school children with mixed dentition aged between 9 and 11 years (n = 60). In each age group, the subjects were divided into two categories: high and low caries risk (n = 30 each). Salivary mucins (MUC5B and MUC7) were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: There were no significant differences in MUC5B and MUC7 levels between high and low caries-risk groups in preschool children. Significantly increased MUC5B (p = 0.01) and decreased MUC7 (p = 0.04) levels in a low caries-risk group were demonstrated in school children. No significant correlations were observed between salivary mucins and dental caries in preschool children, whereas a significantly negative correlation (r = -0.29, p = 0.03) between MUC5B and the number of decayed teeth was observed in school children. CONCLUSION: Patterns of salivary mucin expression in relation to dental caries were different between preschool and school children. The present findings suggest that changes in oral environment from deciduous to mixed dentition may affect the secretion of salivary mucins in response to dental caries. CLINICAL RELEVANCE: The present study provides additional information that changes in oral environment from deciduous to mixed dentition stage possibly affect the secretion of salivary mucins in response to dental caries.


Asunto(s)
Caries Dental/metabolismo , Dentición Mixta , Mucina 5B/metabolismo , Mucinas/metabolismo , Saliva/metabolismo , Proteínas y Péptidos Salivales/metabolismo , Niño , Preescolar , Femenino , Humanos , Masculino
19.
Biochim Biophys Acta ; 1828(5): 1329-39, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23384417

RESUMEN

The structure and membrane interactions of three antimicrobial peptides from the lactoferrin family were investigated through different techniques. Circular dichroism shows that the peptides adopt a secondary structure in the presence of DMPC/DMPG, and DSC reveals that they all interact with these membranes, albeit differently, whereas only LFchimera has an effect in pure zwitterionic membranes of DMPC. DSC further shows that membrane action is weakest for LFcin17-30, increases for LFampin265-284 and is largest for LFchimera. These differences are clearly reflected in a different structure upon interaction, as revealed by SAX. This technique shows that LFcin17-30 only induces membrane segregation (two lamellar phases are apparent upon cooling from fluid phase), whereas LFampin265-284 induces micellization of the membrane with structure compatible to a micellar cubic phase of space group Pm3n, and LFchimera leads to membrane destruction through the formation of two cubic phases, Pn3m and Im3m. These structural results show a remarkable parallel with the ones obtained previously by freeze fracture microscopy of the effect of these peptides against Candida albicans.


Asunto(s)
Antifúngicos/química , Lactoferrina/química , Lípidos de la Membrana/química , Péptidos/química , Secuencia de Aminoácidos , Antifúngicos/metabolismo , Antifúngicos/farmacología , Rastreo Diferencial de Calorimetría , Candida albicans/efectos de los fármacos , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Dicroismo Circular , Dimiristoilfosfatidilcolina/química , Dimiristoilfosfatidilcolina/metabolismo , Lactoferrina/metabolismo , Lactoferrina/farmacología , Lípidos de la Membrana/metabolismo , Datos de Secuencia Molecular , Péptidos/metabolismo , Péptidos/farmacología , Unión Proteica , Estructura Secundaria de Proteína , Termodinámica , Difracción de Rayos X
20.
Antimicrob Agents Chemother ; 58(6): 3461-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24709266

RESUMEN

Mycobacterium avium causes respiratory disease in susceptible individuals, as well as disseminated infections in immunocompromised hosts, being an important cause of morbidity and mortality among these populations. Current therapies consist of a combination of antibiotics taken for at least 6 months, with no more than 60% overall clinical success. Furthermore, mycobacterial antibiotic resistance is increasing worldwide, urging the need to develop novel classes of antimicrobial drugs. One potential and interesting alternative strategy is the use of antimicrobial peptides (AMP). These are present in almost all living organisms as part of their immune system, acting as a first barrier against invading pathogens. In this context, we investigated the effect of several lactoferrin-derived AMP against M. avium. Short peptide sequences from both human and bovine lactoferricins, namely, hLFcin1-11 and LFcin17-30, as well as variants obtained by specific amino acid substitutions, were evaluated. All tested peptides significantly inhibited the axenic growth of M. avium, the bovine peptides being more active than the human. Arginine residues were found to be crucial for the display of antimycobacterial activity, whereas the all-d-amino-acid analogue of the bovine sequence displayed the highest mycobactericidal activity. These findings reveal the promising potential of lactoferricins against mycobacteria, thus opening the way for further research on their development and use as a new weapon against mycobacterial infections.


Asunto(s)
Antibacterianos/farmacología , Antibióticos Antituberculosos/farmacología , Lactoferrina/farmacología , Infecciones por Mycobacterium/tratamiento farmacológico , Mycobacterium avium/efectos de los fármacos , Péptidos/farmacología , Sustitución de Aminoácidos , Aminoácidos/metabolismo , Animales , Arginina , Bovinos , Humanos , Infecciones por Mycobacterium/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA