1
|
Morovati S, Baghkheirati AA, Sekhavati MH, Razmyar J. A Review on cLF36, a Novel Recombinant Antimicrobial Peptide-Derived Camel Lactoferrin. Probiotics Antimicrob Proteins 2024; 16:1886-1905. [PMID: 38722550 DOI: 10.1007/s12602-024-10285-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2024] [Indexed: 10/02/2024]
Abstract
Lactoferrin is an antimicrobial peptide (AMP) playing a pivotal role in numerous biological processes. The primary antimicrobial efficacy of lactoferrin is associated with its N-terminal end, which contains various peptides, such as lactoferricin and lactoferrampin. In this context, our research team has developed a refined chimeric 42-mer peptide known as cLF36 over the past few years. This peptide encompasses the complete amino acid sequence of camel lactoferrampin and partial amino acid sequence of lactoferricin. The peptide's activity against human, avian, and plant bacterial pathogens has been assessed using different biological platforms, including prokaryotic (P170 and pET) and eukaryotic (HEK293) expression systems. The peptide positively influenced the growth performance and intestinal morphology of chickens challenged with pathogen bacteria. Computational methods and in vitro studies showed the peptide's antiviral effects against hepatitis C virus, influenza virus, and rotavirus. The chimeric peptide exhibited higher activity against certain tumor cell lines compared to normal cells, which may be attributed to the peptide's interaction with negatively charged glycosaminoglycans on the surface of tumor cells. Importantly, this peptide exhibited no toxicity against host cells and demonstrated remarkable thermal and protease stability in serum. In conclusion, while our investigations suggest that the chimeric peptide, cLF36, may offer potential as a candidate or complementary option to some available antibiotics, antiviral agents, and chemical pesticides, significant uncertainties remain regarding its cost-effectiveness, as well as its pharmacodynamic and pharmacokinetic characteristics, which require further elucidation.
Collapse
Affiliation(s)
- Solmaz Morovati
- Department of Pathobiology, Division of Biotechnology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | | | - Mohammad Hadi Sekhavati
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Jamshid Razmyar
- Department of Avian Diseases, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
2
|
Tosca MA, Varricchio A, Schiavetti I, Naso M, Damiani V, Ciprandi G. Managing children with frequent respiratory infections and associated wheezing: a preliminary randomized study with a new multicomponent nasal spray. Allergol Immunopathol (Madr) 2024; 52:22-30. [PMID: 38721952 DOI: 10.15586/aei.v52i3.1040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/05/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Preschoolers frequently have respiratory infections (RIs), which may cause wheezing in some subjects. Type 2 polarization may favor increased susceptibility to RIs and associated wheezing. Non-pharmacological remedies are garnering increasing interest as possible add-on therapies. The present preliminary study investigated the efficacy and safety of a new multi-component nasal spray in preschoolers with frequent RIs and associated wheezing. METHODS Some preschoolers with these characteristics randomly took this product, containing lactoferrin, dipotassium glycyrrhizinate, carboxymethyl-beta-glucan, and vitamins C and D3 (Saflovir), two sprays per nostril twice daily for 3 months. Other children were randomly treated only with standard therapy. Outcomes included the number of RIs and wheezing episodes, use of medications, and severity of clinical manifestations. RESULTS Preschoolers treated add-on with this multicomponent product experienced fewer RIs and used fewer beta-2 agonists than untreated children (P = 0.01 and 0.029, respectively). CONCLUSIONS This preliminary study demonstrated that a multicomponent product, administered add-on as a nasal spray, could reduce the incidence of RIs and use of symptomatic drugs for relieving wheezing in children.
Collapse
Affiliation(s)
| | | | | | - Matteo Naso
- Allergy Center, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | | |
Collapse
|
3
|
Yen CC, Wu PY, Ou-Yang H, Chen HL, Chong KY, Chang RL, Chen CM. Production of Bioactive Porcine Lactoferrin through a Novel Glucose-Inducible Expression System in Pichia pastoris: Unveiling Antimicrobial and Anticancer Functionalities. Int J Mol Sci 2024; 25:1818. [PMID: 38339093 PMCID: PMC10855427 DOI: 10.3390/ijms25031818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Lactoferrin (LF) stands as one of the extensively investigated iron-binding glycoproteins within milk, exhibiting diverse biological functionalities. The global demand for LF has experienced consistent growth. Biotechnological strategies aimed at enhancing LF productivity through microbial expression systems offer substantial cost-effective advantages and exhibit fewer constraints compared to traditional animal bioreactor technologies. This study devised a novel recombinant plasmid, wherein the AOX1 promoter was replaced with a glucose-inducible G1 promoter (PG1) to govern the expression of recombinant porcine LF (rpLF) in Pichia pastoris GS115. High-copy-number PG1-rpLF yeast clones were meticulously selected, and subsequent induction with 0.05 g/L glucose demonstrated robust secretion of rpLF. Scaling up production transpired in a 5 L fermenter, yielding an estimated rpLF productivity of approximately 2.8 g/L by the conclusion of glycerol-fed fermentation. A three-step purification process involving tangential-flow ultrafiltration yielded approximately 6.55 g of rpLF crude (approximately 85% purity). Notably, exceptional purity of rpLF was achieved through sequential heparin and size-exclusion column purification. Comparatively, the present glucose-inducible system outperformed our previous methanol-induced system, which yielded a level of 87 mg/L of extracellular rpLF secretion. Furthermore, yeast-produced rpLF demonstrated affinity for ferric ions (Fe3+) and exhibited growth inhibition against various pathogenic microbes (E. coli, S. aureus, and C. albicans) and human cancer cells (A549, MDA-MB-231, and Hep3B), similar to commercial bovine LF (bLF). Intriguingly, the hydrolysate of rpLF (rpLFH) manifested heightened antimicrobial and anticancer effects compared to its intact form. In conclusion, this study presents an efficient glucose-inducible yeast expression system for large-scale production and purification of active rpLF protein with the potential for veterinary or medical applications.
Collapse
Affiliation(s)
- Chih-Ching Yen
- Department of Internal Medicine, China Medical University Hospital, College of Health Care, China Medical University, Taichung 404, Taiwan;
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (P.-Y.W.); (H.O.-Y.)
| | - Pei-Ying Wu
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (P.-Y.W.); (H.O.-Y.)
| | - Huan Ou-Yang
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (P.-Y.W.); (H.O.-Y.)
| | - Hsiao-Ling Chen
- Department of Biomedical Science, Da-Yeh University, Changhua 515, Taiwan;
| | - Kowit-Yu Chong
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Hyperbaric Oxygen Medical Research Laboratory, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ro-Lin Chang
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (P.-Y.W.); (H.O.-Y.)
| | - Chuan-Mu Chen
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (P.-Y.W.); (H.O.-Y.)
- The iEGG and Animal Biotechnology Center, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
4
|
Sadiq IZ, Muhammad A, Mada SB, Ibrahim B, Umar UA. Biotherapeutic effect of cell-penetrating peptides against microbial agents: a review. Tissue Barriers 2021; 10:1995285. [PMID: 34694961 DOI: 10.1080/21688370.2021.1995285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Selective permeability of biological membranes represents a significant barrier to the delivery of therapeutic substances into both microorganisms and mammalian cells, restricting the access of drugs into intracellular pathogens. Cell-penetrating peptides usually 5-30 amino acids with the characteristic ability to penetrate biological membranes have emerged as promising antimicrobial agents for treating infections as well as an effective delivery modality for biological conjugates such as nucleic acids, drugs, vaccines, nanoparticles, and therapeutic antibodies. However, several factors such as antimicrobial resistance and poor drug delivery of the existing medications justify the urgent need for developing a new class of antimicrobials. Herein, we review cell-penetrating peptides (CPPs) used to treat microbial infections. Although these peptides are biologically active for infections, effective transduction into membranes and cargo transport, serum stability, and half-life must be improved for optimum functions and development of next-generation antimicrobial agents.
Collapse
Affiliation(s)
- Idris Zubairu Sadiq
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Sanusi Bello Mada
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Bashiru Ibrahim
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Umar Aliyu Umar
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| |
Collapse
|
5
|
Nishanth MAD, Bhoomika S, Gourkhede D, Dadimi B, Vergis J, Malik SVS, Barbuddhe SB, Rawool DB. Antibacterial efficacy of in-house designed cell-penetrating peptide against multi-drug resistant strains of Salmonella Enteritidis and Salmonella Typhimurium. Environ Microbiol 2021; 24:2747-2758. [PMID: 34528343 DOI: 10.1111/1462-2920.15778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022]
Abstract
The in vitro antibacterial efficacy of an in-house designed cell-penetrating peptide (CPP) variant of Cecropin A (1-7)-Melittin (CAMA) (CAMA-CPP) against the characterized multi-drug resistant (MDR) field strains of Salmonella Enteritidis and Salmonella Typhimurium were evaluated and compared with two identified CPPs namely, P7 and APP, keeping CAMA as control. Initially, the minimum inhibitory concentration (MIC) (μg ml-1 ) of in-house designed CAMA-CPP, APP and CAMA was determined to be 3.91, whereas that of P7 was 7.81; however, the minimum bactericidal concentration (MBC) of all the peptides were twice the MIC. CAMA-CPP and CAMA were found to be stable under different conditions (high-end temperatures, proteinase-K, cationic salts, pH and serum) when compared to the other CPPs. Moreover, CAMA-CPP exhibited negligible cytotoxicity in HEp-2 and RAW 264.7 cell lines as well as haemolysis in the sheep and human erythrocytes with no adverse effects against the commensal gut lactobacilli. In vitro time-kill assay revealed that the MBC levels of CAMA-CPP and APP could eliminate the intracellular MDR-Salmonella infections from mammalian cell lines; however, CAMA and P7 peptides were ineffective. CAMA-CPP appears to be a promising antimicrobial candidate and opens up further avenues for its in vivo clinical translation.
Collapse
Affiliation(s)
- Maria Anto Dani Nishanth
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243 122, India
| | - Sirsant Bhoomika
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243 122, India.,Department of Veterinary Public Health, Bihar Veterinary College, Bihar Animal Sciences University, Patna, Bihar, 800 014, India
| | - Diksha Gourkhede
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243 122, India
| | - Bhargavi Dadimi
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243 122, India
| | - Jess Vergis
- Department of Veterinary Public Health, College of Veterinary and Animal Sciences, Pookode, Kerala Veterinary and Animal Sciences University, Wayanad, Kerala, 673 576, India
| | - Satya Veer Singh Malik
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243 122, India
| | | | - Deepak Bhiwa Rawool
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243 122, India.,ICAR-National Research Centre on Meat, Hyderabad, Telangana, 500 092, India
| |
Collapse
|
6
|
Ramírez-Sánchez DA, Arredondo-Beltrán IG, Canizalez-Roman A, Flores-Villaseñor H, Nazmi K, Bolscher JGM, León-Sicairos N. Bovine lactoferrin and lactoferrin peptides affect endometrial and cervical cancer cell lines. Biochem Cell Biol 2020; 99:149-158. [PMID: 33307991 DOI: 10.1139/bcb-2020-0074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Diana A Ramírez-Sánchez
- CIASaP, Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán Sinaloa, México.,Programa Regional del Noroeste para el Doctorado en Biotecnología, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Sinaloa, Culiacán Sinaloa, México
| | - Izamar G Arredondo-Beltrán
- CIASaP, Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán Sinaloa, México.,Maestría en Ciencias en Biomedicina Molecular, Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán Sinaloa, México
| | - Adrián Canizalez-Roman
- CIASaP, Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán Sinaloa, México.,Hospital de la Mujer, Servicios de Salud de Sinaloa, Culiacán Sinaloa, México
| | | | - Kamran Nazmi
- Department of Oral Biochemistry ACTA, University of Amsterdam and VU University, Amsterdam, the Netherlands
| | - Jan G M Bolscher
- Department of Oral Biochemistry ACTA, University of Amsterdam and VU University, Amsterdam, the Netherlands
| | - Nidia León-Sicairos
- CIASaP, Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán Sinaloa, México.,Departamento de Investigación del Hospital Pediátrico, Servicios de Salud de Sinaloa, Culiacán Sinaloa, México
| |
Collapse
|
7
|
Pirkhezranian Z, Tahmoorespur M, Monhemi H, Sekhavati MH. Computational Peptide Engineering Approach for Selection the Best Engendered Camel Lactoferrin-Derive Peptide with Potency to Interact with DNA. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-10012-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
8
|
Vergis J, Malik SS, Pathak R, Kumar M, Ramanjaneya S, Kurkure NV, Barbuddhe SB, Rawool DB. Exploiting Lactoferricin (17-30) as a Potential Antimicrobial and Antibiofilm Candidate Against Multi-Drug-Resistant Enteroaggregative Escherichia coli. Front Microbiol 2020; 11:575917. [PMID: 33072040 PMCID: PMC7531601 DOI: 10.3389/fmicb.2020.575917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/17/2020] [Indexed: 12/28/2022] Open
Abstract
The study evaluated the in vitro antimicrobial and antibiofilm efficacy of an antimicrobial peptide (AMP), lactoferricin (17–30) [Lfcin (17–30)], against biofilm-forming multi-drug-resistant (MDR) strains of enteroaggregative Escherichia coli (EAEC), and subsequently, the in vivo antimicrobial efficacy was assessed in a Galleria mellonella larval model. Initially, minimum inhibitory concentration (MIC; 32 μM), minimum bactericidal concentration (MBC; 32 μM), and minimum biofilm eradication concentration (MBEC; 32 μM) of Lfcin (17–30) were determined against MDR-EAEC field isolates (n = 3). Lfcin (17–30) was tested stable against high-end temperatures (70 and 90°C), physiological concentration of cationic salts (150 mM NaCl and 2 mM MgCl2), and proteases (proteinase-K and lysozyme). Further, at lower MIC, Lfcin (17–30) proved to be safe for sheep RBCs, secondary cell lines (HEp-2 and RAW 264.7), and beneficial gut lactobacilli. In the in vitro time-kill assay, Lfcin (17–30) inhibited the MDR-EAEC strains 3 h post-incubation, and the antibacterial effect was due to membrane permeation of Lfcin (17–30) in the inner and outer membranes of MDR-EAEC. Furthermore, in the in vivo experiments, G. mellonella larvae treated with Lfcin (17–30) exhibited an increased survival rate, lower MDR-EAEC counts (P < 0.001), mild to moderate histopathological changes, and enhanced immunomodulatory effect and were safe to larval cells when compared with infection control. Besides, Lfcin (17–30) proved to be an effective antibiofilm agent, as it inhibited and eradicated the preformed biofilm formed by MDR-EAEC strains in a significant (P < 0.05) manner both by microtiter plate assay and live/dead bacterial quantification-based confocal microscopy. We recommend further investigation of Lfcin (17–30) in an appropriate animal model before its application in target host against MDR-EAEC strains.
Collapse
Affiliation(s)
- Jess Vergis
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Satyaveer Singh Malik
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Richa Pathak
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Manesh Kumar
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Sunitha Ramanjaneya
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | | | | | - Deepak Bhiwa Rawool
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Izatnagar, India.,ICAR-National Research Centre on Meat, Hyderabad, India
| |
Collapse
|
9
|
Gourkhede DP, Bhoomika S, Pathak R, Yadav JP, Nishanth D, Vergis J, Malik SVS, Barbuddhe SB, Rawool DB. Antimicrobial efficacy of Cecropin A (1-7)- Melittin and Lactoferricin (17-30) against multi-drug resistant Salmonella Enteritidis. Microb Pathog 2020; 147:104405. [PMID: 32707313 DOI: 10.1016/j.micpath.2020.104405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 12/21/2022]
Abstract
The present study evaluated intracellular antibacterial efficacy of two short-chain cationic antimicrobial peptides (AMPs) namely, Cecropin A (1-7)-Melittin and lactoferricin (17-30) against three field strains of multi-drug resistant Salmonella Enteritidis. Initially, antimicrobial ability of both the AMPs was evaluated for their minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) against multi-drug resistant S. Enteritidis strains. Besides, the AMPs were evaluated for its in vitro stability (high-end temperatures, proteases, physiological concentrations of cationic salts and pH) and safety (haemolytic assay in sheep erythrocytes; cytotoxicity assay in murine macrophage RAW 264.7 cell line and human epithelioma HEp-2 cell line and beneficial gut lactobacilli). Later, a time-kill assay was performed to assess the intracellular antibacterial activity of Cecropin A (1-7)-Melittin and lactoferricin (17-30) against multi-drug resistant S. Enteritidis in RAW 264.7 and HEp-2 cells. The observed MBC values of Cecropin A (1-7)-Melittin and lactoferricin (17-30) against multi-drug resistant S. Enteritidis (128 μM; 256 μM) were generally twice or four-fold greater than the MIC values (64 μM). Further, both the AMPs were found variably stable after exposure at high-end temperatures (70 °C and 90 °C), protease treatment (trypsin, proteinase K, lysozyme), higher concentration of physiological salts (150 mM NaCl and 2 mM MgCl2) and hydrogen ion concentrations (pH 4.0 to 8.0). Both the AMPs were found non-haemolytic on sheep erythrocytes, revealed minimal cytotoxicity in RAW 264.7 and HEp-2 cells, and was tested safe against beneficial gut lactobacilli (L. acidophilus and L. rhamnosus). Intracellular bacteriostatic effect with both cationic AMPs against multi-drug resistant S. Enteritidis was evident in RAW 264.7 cells; however, in both the cell lines, the significant bactericidal effect was not observed (P > 0.05) with both cationic AMPs understudy against multi-drug resistant S. Enteritidis. Based on the results of the present study, both the cationic AMPs understudy may not be useful for the intracellular elimination of multi-drug resistant S. Enteritidis; hence, further studies such as conjugation of these AMPs with either cell-penetrating peptides (CPP) and/or nanoparticles (NPs) are warranted.
Collapse
Affiliation(s)
- Diksha P Gourkhede
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, 243 122, Uttar Pradesh, India
| | - Sirsant Bhoomika
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, 243 122, Uttar Pradesh, India
| | - Richa Pathak
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, 243 122, Uttar Pradesh, India
| | - Jay Prakash Yadav
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, 243 122, Uttar Pradesh, India
| | - Dani Nishanth
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, 243 122, Uttar Pradesh, India
| | - Jess Vergis
- Department of Veterinary Public Health, College of Veterinary and Animal Sciences, Pookode, KVASU, 673 576, Kerala, India
| | - S V S Malik
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, 243 122, Uttar Pradesh, India
| | - S B Barbuddhe
- ICAR- National Research Centre on Meat, Hyderabad, 500 092, Telangana, India
| | - D B Rawool
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, 243 122, Uttar Pradesh, India.
| |
Collapse
|
10
|
Pirkhezranian Z, Tahmoorespur M, Daura X, Monhemi H, Sekhavati MH. Interaction of camel Lactoferrin derived peptides with DNA: a molecular dynamics study. BMC Genomics 2020; 21:60. [PMID: 31959108 PMCID: PMC6971935 DOI: 10.1186/s12864-020-6458-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 01/07/2020] [Indexed: 12/31/2022] Open
Abstract
Background Lactoferrampin (LFampin), Lactoferricin (LFcin), and LFchimera are three well-known antimicrobial peptides derived from Lactoferrin and proposed as alternatives for antibiotics. Although the intracellular activity of these peptides has been previously demonstrated, their mode of action is not yet fully understood. Here, we performed a molecular dynamics simulation study to understand the molecular interactions between camel Lactoferrin derived peptides, including CLFampin, CLFcin, and CLFchimera, and DNA as an important intracellular target. Results Our results indicate that all three peptides bind to DNA, albeit with different propensities, with CLFchimera showing the highest binding affinity. The secondary structures of the peptides, modeled on Lactoferrin, did not undergo significant changes during simulation, supporting their functional relevance. Main residues involved in the peptide-DNA interaction were identified based on binding free energy estimates calculated over 200 ns, which, as expected, confirmed strong electrostatic interactions between DNA phosphate groups and positively charged peptide side chains. Interaction between the different concentrations of CLFchimera and DNA revealed that after binding of four copies of CLFchimera to DNA, hydrogen bonds between the two strands of DNA start to break from one of the termini. Conclusions Importantly, our results revealed that there is no DNA-sequence preference for peptide binding, in line with a broad antimicrobial activity. Moreover, the results showed that the strength of the interaction between DNA and CLFchimera is concentration dependent. The insight provided by these results can be used for the rational redesign of natural antimicrobial peptides targeting the bacterial DNA.
Collapse
Affiliation(s)
- Zana Pirkhezranian
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mojtaba Tahmoorespur
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Xavier Daura
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Hassan Monhemi
- Department of Chemistry, Faculty of Science, University of Neyshabur, Neyshabur, Iran
| | - Mohammad Hadi Sekhavati
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
11
|
Vergis J, Malik SS, Pathak R, Kumar M, Ramanjaneya S, Kurkure NV, Barbuddhe SB, Rawool DB. Antimicrobial Efficacy of Indolicidin Against Multi-Drug Resistant Enteroaggregative Escherichia coli in a Galleria mellonella Model. Front Microbiol 2019; 10:2723. [PMID: 31849877 PMCID: PMC6895141 DOI: 10.3389/fmicb.2019.02723] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial resistance against enteroaggregative Escherichia coli (EAEC), an emerging food-borne pathogen, has been observed in an increasing trend recently. In the recent wake of antimicrobial resistance, alternate strategies especially, cationic antimicrobial peptides (AMPs) have attracted considerable attention to source antimicrobial technology solutions. This study evaluated the in vitro antimicrobial efficacy of Indolicidin against multi-drug resistant enteroaggregative Escherichia coli (MDR-EAEC) strains and further to assess its in vivo antimicrobial efficacy in Galleria mellonella larval model. The minimum inhibitory concentration (MIC; 32 μM) and minimum bactericidal concentration (MBC; 64 μM) of Indolicidin against MDR-EAEC was determined by micro broth dilution method. Indolicidin was also tested for its stability (high-end temperatures, physiological concentration of salts and proteases); safety (sheep RBCs; HEp-2 and RAW 264.7 cell lines); effect on beneficial microflora (Lactobacillus rhamnosus and Lactobacillus acidophilus) and its mode of action (flow cytometry; nitrocefin and ONPG uptake). In vitro time-kill kinetic assay of MDR-EAEC treated with Indolicidin was performed. Further, survival rate, MDR-EAEC count, melanization rate, hemocyte enumeration, cytotoxicity assay and histopathological examination were carried out in G. mellonella model to assess in vivo antimicrobial efficacy of Indolicidin against MDR-EAEC strains. Indolicidin was tested stable at high temperatures (70°C; 90°C), physiological concentration of cationic salts (NaCl; MgCl2) and proteases, except for trypsin and tested safe with sheep RBCs and cell lines (RAW 264.7; HEp-2) at MIC (1X and 2X); the beneficial flora was not inhibited. Indolicidin exhibited outer membrane permeabilization in a concentration- and time-dependent manner. In vitro time-kill assay revealed concentration-cum-time dependent clearance of MDR-EAEC in Indolicidin-treated groups at 120 min, while, in G. mellonella, the infected group treated with Indolicidin revealed an increased survival rate, immunomodulatory effect, reduced MDR-EAEC counts and were tested safe to the larval cells which was concurred histopathologically. To conclude, the results suggests Indolicidin as an effective antimicrobial candidate against MDR-EAEC and we recommend its further investigation in appropriate animal models (mice/piglets) before its application in the target host.
Collapse
Affiliation(s)
- Jess Vergis
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Satyaveer Singh Malik
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Richa Pathak
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Manesh Kumar
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Sunitha Ramanjaneya
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | | | | | - Deepak Bhiwa Rawool
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, India
| |
Collapse
|
12
|
Alexander DB, Vogel HJ, Tsuda H. Lactoferrin researchers descend on Nagoya Castle. Biochem Cell Biol 2018; 95:1-4. [PMID: 28186858 DOI: 10.1139/bcb-2017-0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Hans J Vogel
- b Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Hiroyuki Tsuda
- c Nanotoxicology Project, Nagoya City University, Nagoya, Japan
| |
Collapse
|
13
|
Multifunctional capacity and therapeutic potential of lactoferrin. Life Sci 2018; 195:61-64. [DOI: 10.1016/j.lfs.2018.01.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/30/2017] [Accepted: 01/03/2018] [Indexed: 01/06/2023]
|
14
|
Ruangcharoen S, Suwannarong W, Lachica MRCT, Bolscher JGM, Nazmi K, Khunkitti W, Taweechaisupapong S. Killing activity of LFchimera on periodontopathic bacteria and multispecies oral biofilm formation in vitro. World J Microbiol Biotechnol 2017; 33:167. [DOI: 10.1007/s11274-017-2334-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/12/2017] [Indexed: 10/19/2022]
|
15
|
Uptake Mechanism of Cell-Penetrating Peptides. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1030:255-264. [DOI: 10.1007/978-3-319-66095-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|