7 results on '"Bolscher, Jan G. M."'
Search Results
2. Antitumor activity of bovine lactoferrin and its derived peptides against HepG2 liver cancer cells and Jurkat leukemia cells.
- Author
-
Arredondo-Beltrán IG, Ramírez-Sánchez DA, Zazueta-García JR, Canizalez-Roman A, Angulo-Zamudio UA, Velazquez-Roman JA, Bolscher JGM, Nazmi K, and León-Sicairos N
- Subjects
- Child, Humans, Jurkat Cells, Hep G2 Cells, Cisplatin, Etoposide, Quality of Life, Peptides pharmacology, Necrosis, Lactoferrin pharmacology, Lactoferrin chemistry, Liver Neoplasms drug therapy
- Abstract
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., (© 2023. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2023
- Full Text
- View/download PDF
3. LFchimera protects HeLa cells from invasion by Yersinia spp. in vitro.
- Author
-
Sijbrandij T, Ligtenberg AJ, Nazmi K, van den Keijbus PAM, Veerman ECI, Bolscher JGM, and Bikker FJ
- Subjects
- Anti-Bacterial Agents chemistry, Antimicrobial Cationic Peptides chemistry, Cell Adhesion drug effects, Dose-Response Relationship, Drug, HeLa Cells, Humans, Microbial Sensitivity Tests, Structure-Activity Relationship, Anti-Bacterial Agents pharmacology, Antimicrobial Cationic Peptides pharmacology, Yersinia drug effects
- Abstract
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.
- Published
- 2018
- Full Text
- View/download PDF
4. Membrane-active mechanism of LFchimera against Burkholderia pseudomallei and Burkholderia thailandensis.
- Author
-
Kanthawong S, Puknun A, Bolscher JG, Nazmi K, van Marle J, de Soet JJ, Veerman EC, Wongratanacheewin S, and Taweechaisupapong S
- Subjects
- Amino Acid Sequence, Animals, Antimicrobial Cationic Peptides chemistry, Antimicrobial Cationic Peptides genetics, Antimicrobial Cationic Peptides pharmacology, Burkholderia metabolism, Burkholderia ultrastructure, Burkholderia pseudomallei metabolism, Burkholderia pseudomallei ultrastructure, Cattle, Cell Membrane drug effects, Freeze Fracturing, Hemolysis drug effects, Humans, Lactoferrin genetics, Molecular Sequence Data, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments pharmacology, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins pharmacology, Species Specificity, Burkholderia drug effects, Burkholderia pseudomallei drug effects, Lactoferrin chemistry, Lactoferrin pharmacology
- Abstract
LFchimera, a construct combining two antimicrobial domains of bovine lactoferrin, lactoferrampin265-284 and lactoferricin17-30, possesses strong bactericidal activity. As yet, no experimental evidence was presented to evaluate the mechanisms of LFchimera against Burkholderia isolates. In this study we analyzed the killing activity of LFchimera on the category B pathogen Burkholderia pseudomallei in comparison to the lesser virulent Burkholderia thailandensis often used as a model for the highly virulent B. pseudomallei. Killing kinetics showed that B. thailandensis E264 was more susceptible for LFchimera than B. pseudomallei 1026b. Interestingly the bactericidal activity of LFchimera appeared highly pH dependent; B. thailandensis killing was completely abolished at and below pH 6.4. FITC-labeled LFchimera caused a rapid accumulation within 15 min in the cytoplasm of both bacterial species. Moreover, freeze-fracture electron microscopy demonstrated extreme effects on the membrane morphology of both bacterial species within 1 h of incubation, accompanied by altered membrane permeability monitored as leakage of nucleotides. These data indicate that the mechanism of action of LFchimera is similar for both species and encompasses disruption of the plasma membrane and subsequently leakage of intracellular nucleotides leading to cell dead.
- Published
- 2014
- Full Text
- View/download PDF
5. Bactericidal effect of bovine lactoferrin and synthetic peptide lactoferrin chimera in Streptococcus pneumoniae and the decrease in luxS gene expression by lactoferrin.
- Author
-
León-Sicairos N, Angulo-Zamudio UA, Vidal JE, López-Torres CA, Bolscher JG, Nazmi K, Reyes-Cortes R, Reyes-López M, de la Garza M, and Canizalez-Román A
- Subjects
- Animals, Antimicrobial Cationic Peptides pharmacology, Bacterial Proteins genetics, Carbon-Sulfur Lyases genetics, Cattle, Child, Gene Expression drug effects, Genes, Bacterial drug effects, Humans, Peptide Fragments pharmacology, Pneumococcal Infections drug therapy, Recombinant Fusion Proteins pharmacology, Streptococcus pneumoniae pathogenicity, Anti-Bacterial Agents pharmacology, Lactoferrin pharmacology, Streptococcus pneumoniae drug effects, Streptococcus pneumoniae genetics
- Abstract
Streptococcus pneumoniae (pneumococcus) is responsible for nearly one million child deaths annually. Pneumococcus causes infections such as pneumonia, otitis media, meningitis, and sepsis. The human immune system includes antibacterial peptides and proteins such as lactoferrin (LF), but its activity against pneumococcus is not fully understood. The aim of this work was to evaluate the bactericidal effect of bovine lactoferrin (bLF) and the synthetic LF-peptides lactoferricin (LFcin17-30), lactoferrampin (LFampin265-284), and LFchimera against S. pneumoniae planktonic cells. The mechanism of damage was also investigated, as well as the impact of these peptides on the transcription levels of genes known to encode important virulence factors. S. pneumoniae planktonic cells were treated with bLF, LFcin17-30, LFampin265-284 and LFchimera at different time points. The viability of treated planktonic cells was assessed by dilution and plating (in CFU/ml). The interaction between LF and LF-peptides coupled to fluorescein was visualized using a confocal microscope and flow cytometry, whereas the damage at structural levels was observed by electron microscopy. Damage to bacterial membranes was further evaluated by membrane permeabilization by use of propidium iodide and flow cytometry, and finally, the expression of pneumococcal genes was evaluated by qRT-PCR. bLF and LFchimera were the best bactericidal agents. bLF and peptides interacted with bacteria causing changes in the shape and size of the cell and membrane permeabilization. Moreover, the luxS gene was down-regulated in bacteria treated with LF. In conclusion, LF and LFchimera have a bactericidal effect, and LF down-regulates genes involved in the pathogenicity of pneumococcus, thus demonstrating potential as new agents for the treatment of pneumococcal infections.
- Published
- 2014
- Full Text
- View/download PDF
6. Bovine and human lactoferricin peptides: chimeras and new cyclic analogs.
- Author
-
Arias M, McDonald LJ, Haney EF, Nazmi K, Bolscher JG, and Vogel HJ
- Subjects
- Amino Acid Sequence, Animals, Antimicrobial Cationic Peptides genetics, Antimicrobial Cationic Peptides pharmacology, Cattle, Cell Membrane Permeability drug effects, Click Chemistry, Escherichia coli drug effects, Escherichia coli pathogenicity, Escherichia coli physiology, Humans, Lactoferrin genetics, Lactoferrin pharmacology, Molecular Sequence Data, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments pharmacology, Peptides, Cyclic chemistry, Peptides, Cyclic genetics, Peptides, Cyclic pharmacology, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins pharmacology, Species Specificity, Unilamellar Liposomes chemistry, Antimicrobial Cationic Peptides chemistry, Lactoferrin chemistry
- Abstract
Lactoferrin (LF) is an important antimicrobial and immune regulatory protein present in neutrophils and most exocrine secretions of mammals. The antimicrobial activity of LF has been related to the presence of an antimicrobial peptide sequence, called lactoferricin (LFcin), located in the N-terminal region of the protein. The antimicrobial activity of bovine LFcin is considerably stronger than the human version. In this work, chimera peptides combining segments of bovine and human LFcin were generated in order to study their antimicrobial activity and mechanism of action. In addition, the relevance of the conserved disulfide bridge and the resulting cyclic structure of both LFcins were analyzed by using "click chemistry" and sortase A-catalyzed cyclization of the peptides. The N-terminal region of bovine LFcin (residues 17-25 of bovine LF) proved to be very important for the antimicrobial activity of the chimera peptides against E. coli, when combined with the C-terminal region of human LFcin. Similarly the cyclic bovine LFcin analogs generated by "click chemistry" and sortase A preserved the antimicrobial activity of the original peptide, showing the significance of these two techniques in the design of cyclic antimicrobial peptides. The mechanism of action of bovine LFcin and its active derived peptides was strongly correlated with membrane leakage in E. coli and up to some extent with the ability to induce vesicle aggregation. This mechanism was also preserved under conditions of high ionic strength (150 mM NaCl) illustrating the importance of these peptides in a more physiologically relevant system.
- Published
- 2014
- Full Text
- View/download PDF
7. Bactericidal effect of bovine lactoferrin, LFcin, LFampin and LFchimera on antibiotic-resistant Staphylococcus aureus and Escherichia coli.
- Author
-
Flores-Villaseñor H, Canizalez-Román A, Reyes-Lopez M, Nazmi K, de la Garza M, Zazueta-Beltrán J, León-Sicairos N, and Bolscher JG
- Subjects
- Animals, Anti-Bacterial Agents chemistry, Cattle, Microbial Sensitivity Tests, Peptides chemistry, Structure-Activity Relationship, Anti-Bacterial Agents pharmacology, Drug Resistance, Bacterial, Escherichia coli drug effects, Lactoferrin chemistry, Lactoferrin pharmacology, Peptides pharmacology, Staphylococcus aureus drug effects
- Abstract
Increased prevalence of antibiotic-resistant bacteria has become a major threat to the health sector worldwide due to their virulence, limited therapeutic options and distribution in both hospital and community settings. Discovery and development of new agents to combat antibiotic-resistant bacteria is thus needed. This study therefore aimed to evaluate the ability of bovine lactoferrin (LF), peptides from two antimicrobial domains lactoferricin B (LFcin17-30) and lactoferrampin (LFampin265-284) and a chimeric construct (LFchimera) containing both peptides, as potential bactericidal agents against clinical isolates of antibiotic-resistant Staphylococcus aureus and Escherichia coli. Results in kinetics of growth show that LF chimera and peptides inhibited the growth of both bacterial species. By confocal microscopy and flow cytometry it was observed that LF and FITC-labeled peptides are able to interact with these bacteria and cause membrane permeabilization, as monitored by propidium iodide staining, these effects were decreased by preincubation with lipopolysaccharide in E. coli. By electron microscopy, a clear cellular damage was observed in bacteria after treatments with LFchimera and peptides, suggesting that interaction and membrane disruption are probably involved as a mechanism of action. In conclusion, results show that LFchimera, LF and peptides have potential as bactericidal agents in the antibiotic-resistant strains of S. aureus and E. coli and also the work strongly suggest that LFcin17-30 and LFampin265-284 acts synergistically with antibiotics against multidrug resistant EPEC and MRSA in vitro.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.