101
|
Figainin 1, a Novel Amphibian Skin Peptide with Antimicrobial and Antiproliferative Properties. Antibiotics (Basel) 2020; 9:antibiotics9090625. [PMID: 32967114 PMCID: PMC7559428 DOI: 10.3390/antibiotics9090625] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 01/10/2023] Open
Abstract
Amphibian skin secretions are abundant in bioactive compounds, especially antimicrobial peptides. These molecules are generally cationic and rich in hydrophobic amino acids, have an amphipathic structure and adopt an α-helical conformation when in contact with microorganisms membranes. In this work, we purified and characterized Figainin 1, a novel antimicrobial and antiproliferative peptide from the cutaneous secretion of the frog Boana raniceps. Figainin 1 is a cationic peptide with eighteen amino acid residues—rich in leucine and isoleucine, with an amidated C-terminus—and adopts an α-helical conformation in the presence of trifluoroethanol (TFE). It displayed activity against Gram-negative and especially Gram-positive bacteria, with MIC values ranging from 2 to 16 µM, and showed an IC50 value of 15.9 µM against epimastigote forms of T. cruzi; however, Figanin 1 did not show activity against Candida species. This peptide also showed cytolytic effects against human erythrocytes with an HC50 of 10 µM, in addition to antiproliferative activity against cancer cells and murine fibroblasts, with IC50 values ranging from 10.5 to 13.7 µM. Despite its adverse effects on noncancerous cells, Figainin 1 exhibits interesting properties for the development of new anticancer agents and anti-infective drugs against pathogenic microorganisms.
Collapse
|
102
|
Kumari T, Verma DP, Afshan T, Verma NK, Pant G, Ali M, Shukla PK, Mitra K, Ghosh JK. A Noncytotoxic Temporin L Analogue with In Vivo Antibacterial and Antiendotoxin Activities and a Nonmembrane-Lytic Mode of Action. ACS Infect Dis 2020; 6:2369-2385. [PMID: 32786286 DOI: 10.1021/acsinfecdis.0c00022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cytotoxic frog antimicrobial peptide Temporin L (TempL) is an attractive molecule for the design of lead antimicrobial agents due to its short size and versatile biological activities. However, noncytotoxic TempL variants with desirable biological activities have rarely been reported. TempL analogue Q3K,TempL is water-soluble and possesses a significant antiendotoxin property along with comparable cytotoxicity to TempL. A phenylalanine residue, located at the hydrophobic face of Q3K,TempL and the "d" position of its phenylalanine zipper sequence, was replaced with a cationic lysine residue. This analogue, Q3K,F8K,TempL, showed reduced hydrophobic moment and was noncytotoxic with lower antimicrobial activity. Interestingly, swapping between tryptophan at the fourth and serine at the sixth positions turned Q3K,F8K,TempL totally amphipathic as reflected by its helical wheel projection with clusters of hydrophobic and hydrophilic residues and the highest hydrophobic moment among these peptides. Surprisingly, this analogue, SW,Q3K,F8K,TempL, was as noncytotoxic as Q3K,F8K,TempL but showed augmented antimicrobial and antiendotoxin properties, comparable to that of TempL and Q3K,TempL. SW,Q3K,F8K,TempL exhibited appreciable survival of mice against P. aeruginosa infection and a lipopolysaccharide (LPS) challenge. Unlike TempL and Q3K,TempL, SW,Q3K,F8K,TempL adopted an unordered secondary structure in bacterial membrane mimetic lipid vesicles and did not permeabilize them or depolarize the bacterial membrane. Overall, the results demonstrate the design of a nontoxic TempL analogue that possesses clusters of hydrophobic and hydrophilic residues with impaired secondary structure and shows a nonmembrane-lytic mechanism and in vivo antiendotoxin and antimicrobial activities. This paradigm of design of antimicrobial peptide with clusters of hydrophobic and hydrophilic residues and high hydrophobic moment but low secondary structure could be attempted further.
Collapse
Affiliation(s)
- Tripti Kumari
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Devesh Pratap Verma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Tayyaba Afshan
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Neeraj Kumar Verma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Garima Pant
- Electron Microscopy Unit, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Mehmood Ali
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - P. K. Shukla
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Kalyan Mitra
- Electron Microscopy Unit, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Jimut Kanti Ghosh
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| |
Collapse
|
103
|
Balatti GE, Domene C, Martini MF, Pickholz M. Differential Stability of Aurein 1.2 Pores in Model Membranes of Two Probiotic Strains. J Chem Inf Model 2020; 60:5142-5152. [PMID: 32815723 DOI: 10.1021/acs.jcim.0c00855] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Aurein 1.2 is an antimicrobial peptide from the skin secretion of an Australian frog. In the previous experimental work, we reported a differential action of aurein 1.2 on two probiotic strains Lactobacillus delbrueckii subsp. bulgaricus (CIDCA 331) and Lactobacillus delbrueckii subsp. lactis (CIDCA 133). The differences found were attributed to the bilayer compositions. Cell cultures and CIDCA 331-derived liposomes showed higher susceptibility than the ones derived from the CIDCA 133 strain, leading to content leakage and structural disruption. Here, we used molecular dynamics simulations to explore these systems at the atomistic level. We hypothesize that if the antimicrobial peptides organized themselves to form a pore, it will be more stable in membranes that emulate the CIDCA 331 strain than in those of the CIDCA 133 strain. To test this hypothesis, we simulated preassembled aurein 1.2 pores embedded into bilayer models that emulate the two probiotic strains. It was found that the general behavior of the systems depends on the composition of the membrane rather than the preassemble system characteristics. Overall, it was observed that aurein 1.2 pores are more stable in the CIDCA 331 model membranes. This fact coincides with the high susceptibility of this strain against antimicrobial peptide. In contrast, in the case of the CIDCA 133 model membranes, peptides migrate to the water-lipid interphase, the pore shrinks, and the transport of water through the pore is reduced. The tendency of glycolipids to make hydrogen bonds with peptides destabilizes the pore structures. This feature is observed to a lesser extent in CIDCA 331 due to the presence of anionic lipids. Glycolipid transverse diffusion (flip-flop) between monolayers occurs in the pore surface region in all the cases considered. These findings expand our understanding of the antimicrobial peptide resistance properties of probiotic strains.
Collapse
Affiliation(s)
- Galo E Balatti
- Departamento de Física Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 1, Buenos Aires 1428, Argentina.,IFIBA, CONICET-UBA, Ciudad Universitaria, Pabellón 1, Buenos Aires 1428, Argentina
| | - Carmen Domene
- Department of Chemistry, University of Bath, 1 South Bldg., Claverton Down, Bath BA27AY, The United Kingdom.,Department of Chemistry, University of Oxford, Oxford OX1 3TA, The United Kingdom
| | - M Florencia Martini
- Facultad de Farmacia y Bioquímica, Departamento de Farmacología, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina.,Instituto de Química y Metabolismo del Fármaco, Fac. de Farmacia y Bioquímica, (Universidad de Buenos Aires, IQUIMEFA-CONICET), Junín 956, C1113AAD Buenos Aires, Argentina
| | - Monica Pickholz
- Departamento de Física Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 1, Buenos Aires 1428, Argentina.,IFIBA, CONICET-UBA, Ciudad Universitaria, Pabellón 1, Buenos Aires 1428, Argentina
| |
Collapse
|
104
|
Potent in vitro and in vivo antimicrobial activity of semisynthetic amphiphilic γ-mangostin derivative LS02 against Gram-positive bacteria with destructive effect on bacterial membrane. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183353. [DOI: 10.1016/j.bbamem.2020.183353] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/02/2020] [Accepted: 05/08/2020] [Indexed: 12/24/2022]
|
105
|
Ocampo-Ibáñez ID, Liscano Y, Rivera-Sánchez SP, Oñate-Garzón J, Lugo-Guevara AD, Flórez-Elvira LJ, Lesmes MC. A Novel Cecropin D-Derived Short Cationic Antimicrobial Peptide Exhibits Antibacterial Activity Against Wild-Type and Multidrug-Resistant Strains of Klebsiella pneumoniae and Pseudomonas aeruginosa. Evol Bioinform Online 2020; 16:1176934320936266. [PMID: 32636607 PMCID: PMC7323284 DOI: 10.1177/1176934320936266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022] Open
Abstract
Infections caused by multidrug-resistant (MDR) Pseudomonas aeruginosa and Klebsiella pneumoniae are a serious worldwide public health concern due to the ineffectiveness of empirical antibiotic therapy. Therefore, research and the development of new antibiotic alternatives are urgently needed to control these bacteria. The use of cationic antimicrobial peptides (CAMPs) is a promising candidate alternative therapeutic strategy to antibiotics because they exhibit antibacterial activity against both antibiotic susceptible and MDR strains. In this study, we aimed to investigate the in vitro antibacterial effect of a short synthetic CAMP derived from the ΔM2 analog of Cec D-like (CAMP-CecD) against clinical isolates of K pneumoniae (n = 30) and P aeruginosa (n = 30), as well as its hemolytic activity. Minimal inhibitory concentrations (MICs) and minimal bactericidal concentrations (MBCs) of CAMP-CecD against wild-type and MDR strains were determined by the broth microdilution test. In addition, an in silico molecular dynamic simulation was performed to predict the interaction between CAMP-CecD and membrane models of K pneumoniae and P aeruginosa. The results revealed a bactericidal effect of CAMP-CecD against both wild-type and resistant strains, but MDR P aeruginosa showed higher susceptibility to this peptide with MIC values between 32 and >256 μg/mL. CAMP-CecD showed higher stability in the P aeruginosa membrane model compared with the K pneumoniae model due to the greater number of noncovalent interactions with phospholipid 1-Palmitoyl-2-oleyl-sn-glycero-3-(phospho-rac-(1-glycerol)) (POPG). This may be related to the boosted effectiveness of the peptide against P aeruginosa clinical isolates. Given the antibacterial activity of CAMP-CecD against wild-type and MDR clinical isolates of P aeruginosa and K pneumoniae and its nonhemolytic effects on human erythrocytes, CAMP-CecD may be a promising alternative to conventional antibiotics.
Collapse
Affiliation(s)
- Iván Darío Ocampo-Ibáñez
- Research Group of Microbiology, Industry and Environment, Faculty of Basic Sciences, Universidad Santiago de Cali, Cali, Colombia
| | - Yamil Liscano
- Research Group of Genetic, Regeneration and Cancer, Faculty of Exacts and Natural Sciences, Universidad de Antioquia, Medellín, Colombia
| | - Sandra Patricia Rivera-Sánchez
- Research Group of Microbiology, Industry and Environment, Faculty of Basic Sciences, Universidad Santiago de Cali, Cali, Colombia
| | - José Oñate-Garzón
- Research Group of Chemical and Biotechnology, Faculty of Basic Sciences, Universidad Santiago de Cali, Cali, Colombia
| | - Ashley Dayan Lugo-Guevara
- Research Group of Microbiology, Industry and Environment, Faculty of Basic Sciences, Universidad Santiago de Cali, Cali, Colombia
| | | | - Maria Cristina Lesmes
- Valle del Cauca Departmental Secretary of Health, Gobernación del Valle del Cauca, Cali, Colombia
| |
Collapse
|
106
|
Antifungal In Vitro Activity of Pilosulin- and Ponericin-Like Peptides from the Giant Ant Dinoponera quadriceps and Synergistic Effects with Antimycotic Drugs. Antibiotics (Basel) 2020; 9:antibiotics9060354. [PMID: 32585881 PMCID: PMC7344683 DOI: 10.3390/antibiotics9060354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/17/2020] [Accepted: 06/20/2020] [Indexed: 02/06/2023] Open
Abstract
Venoms from ants comprise a rich source of bioactive peptides, including antimicrobial peptides. From the proteome and peptidome of the giant ant Dinoponera quadriceps venom, members of five known classes of antimicrobial peptides were disclosed (e.g., dermaseptin-, defensin-, ICK-, pilosulin- and ponericin-like types). Based on comparative analysis, these family members have structural determinants that indicate they could display antimicrobial activities. In previous works, pilosulin- and ponericin-like peptides were demonstrated to be active against bacteria, fungi, and parasites. Herein, the antifungal activity of ponericin- and pilosulin-like peptides were assessed, aiming at the expansion of the knowledge about AMPs in predatory ants and the development of new microbicide strategies to deal with difficult-to-treat fungal infections. Synthetic pilosulin- (Dq-2562, Dq-1503, and Dq-1319) and ponericin-like (Dq-3162) peptides were evaluated for their fungicide and fungistatic activities against different species of Candida, including a drug-resistant clinical strain. The MICs and MLCs were determined for all peptides individually and in combination with general antifungal drugs by the microdilution method. The time-kill kinetic curves were set up by means of a luminescent reagent, of which the light signal is proportional to the number of viable cells. The candicidal synergism observed by the combination of subinhibitory concentrations of peptides and general antimycotic drugs were quantified by the checkerboard test and fluorescent dye permeation assay. The influence of ergosterol on the antifungal activity was verified by supplementation of culture medium. The pilosulin- (Dq-2562 and Dq-1503) and ponericin-like (Dq-3162) were the most active peptides, displaying a broad spectrum of antifungal activity in vitro, with MICs in the range of 0.625 to 10 µM. The combination of peptides and conventional antimycotic drugs displayed a synergistic reduction in the MIC values of individual peptides and drugs, while soluble ergosterol in the culture medium increased the MICs. The fungicide and fungistatic activity of the individual peptides and peptides in combination with antimycotics were time-dependent with a rapid onset of action and long-lasting effect, which involved membrane disruption as an underlying mechanism of their action. Altogether, pilosulin- and ponericin-like peptides from the giant ant D. quadriceps venom display a broad-spectrum of candicidal activity, what allows their inclusion in the row of the antifungal peptides and gives support for further studies on the development of strategies to fight candidiasis.
Collapse
|
107
|
Antibiotic Resistance Profiles, Molecular Mechanisms and Innovative Treatment Strategies of Acinetobacter baumannii. Microorganisms 2020; 8:microorganisms8060935. [PMID: 32575913 PMCID: PMC7355832 DOI: 10.3390/microorganisms8060935] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/18/2022] Open
Abstract
Antibiotic resistance is one of the biggest challenges for the clinical sector and industry, environment and societal development. One of the most important pathogens responsible for severe nosocomial infections is Acinetobacter baumannii, a Gram-negative bacterium from the Moraxellaceae family, due to its various resistance mechanisms, such as the β-lactamases production, efflux pumps, decreased membrane permeability and altered target site of the antibiotic. The enormous adaptive capacity of A. baumannii and the acquisition and transfer of antibiotic resistance determinants contribute to the ineffectiveness of most current therapeutic strategies, including last-line or combined antibiotic therapy. In this review, we will present an update of the antibiotic resistance profiles and underlying mechanisms in A. baumannii and the current progress in developing innovative strategies for combating multidrug-resistant A. baumannii (MDRAB) infections.
Collapse
|
108
|
White LJ, Boles JE, Allen N, Alesbrook LS, Sutton JM, Hind CK, Hilton KLF, Blackholly LR, Ellaby RJ, Williams GT, Mulvihill DP, Hiscock JR. Controllable hydrogen bonded self-association for the formation of multifunctional antimicrobial materials. J Mater Chem B 2020; 8:4694-4700. [PMID: 32393938 DOI: 10.1039/d0tb00875c] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
SSAs are a class of supramolecular self-associating amphiphilic salt, the anionic component of which contains a covalently bound hydrogen bond donor-acceptor motif. This results in a monomeric unit which can adopt multiple hydrogen bonding modes simultaneously. Previous investigations have shown examples of SSAs to act as antimicrobial agents against clinically relevant methicillin-resistant Staphylococcus aureus (MRSA). Herein, we report an intrinsically fluorescent SSA which can self-associate producing dimers, spherical aggregates and hydrogels dependent on solvent environment, while retaining antimicrobial activity against both model Gram-positive (MRSA) and Gram-negative (Escherichia coli) bacteria. Finally, we demonstrate the SSA supramolecular hydrogel to tolerate the inclusion of the antibiotic ampicillin, leading to the enhanced inhibition of growth with both model bacteria, and derive initial molecular structure-physicochemical property-antimicrobial activity relationships.
Collapse
Affiliation(s)
- Lisa J White
- School of Physical Sciences, University of Kent, Canterbury, Kent CT2 7NH, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Cancelarich NL, Wilke N, Fanani MAL, Moreira DC, Pérez LO, Alves Barbosa E, Plácido A, Socodato R, Portugal CC, Relvas JB, de la Torre BG, Albericio F, Basso NG, Leite JR, Marani MM. Somuncurins: Bioactive Peptides from the Skin of the Endangered Endemic Patagonian Frog Pleurodema somuncurense. JOURNAL OF NATURAL PRODUCTS 2020; 83:972-984. [PMID: 32134261 DOI: 10.1021/acs.jnatprod.9b00906] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The skin glands of amphibian species hold a major component of their innate immunity, namely a unique set of antimicrobial peptides (AMPs). Although most of them have common characteristics, differences in AMP sequences allow a huge repertoire of biological activity with varying degrees of efficacy. We present the first study of the AMPs from Pleurodema somuncurence (Anura: Leptodactylidae: Leiuperinae). Among the 11 identified mature peptides, three presented antimicrobial activity. Somuncurin-1 (FIIWPLRYRK), somuncurin-2 (FILKRSYPQYY), and thaulin-3 (NLVGSLLGGILKK) inhibited Escherichia coli growth. Somuncurin-1 also showed antimicrobial activity against Staphylococcus aureus. Biophysical membrane model studies revealed that this peptide had a greater permeation effect in prokaryotic-like membranes and capacity to restructure liposomes, suggesting fusogenic activity, which could lead to cell aggregation and disruption of cell morphology. This study contributes to the characterization of peptides with new sequences to enrich the databases for the design of therapeutic agents. Furthermore, it highlights the importance of investing in nature conservation and the power of genetic description as a strategy to identify new compounds.
Collapse
Affiliation(s)
- Natalia L Cancelarich
- Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Consejo Nacional de Investigaciones Cientı́ficas y Técnicas (IPEEC-CONICET), Bv. Almirante Brown 2915, Puerto Madryn U9120ACD, Argentina
| | - Natalia Wilke
- Departamento de Quı́mica Biológica Ranwel Caputto, Facultad de Ciencias Quı́micas, Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Centro de Investigaciones en Quı́mica Biológica de Córdoba, CONICET, Ciudad Universitaria, Haya de la Torre y Medina Allende, Córdoba X5000HUA, Argentina
| | - Marı A L Fanani
- Departamento de Quı́mica Biológica Ranwel Caputto, Facultad de Ciencias Quı́micas, Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Centro de Investigaciones en Quı́mica Biológica de Córdoba, CONICET, Ciudad Universitaria, Haya de la Torre y Medina Allende, Córdoba X5000HUA, Argentina
| | - Daniel C Moreira
- Área de Morfologia, Faculdade de Medicina, Universidade de Brası́lia, Brası́lia 70910-900, Brazil
| | - Luis O Pérez
- Instituto Patagónico de Ciencias Sociales y Humanas, Consejo Nacional de Investigaciones Cientı́ficas y Técnicas (IPCSH-CONICET), Bv. Almirante Brown 2915, Puerto Madryn U9120ACD, Argentina
| | - Eder Alves Barbosa
- Laboratório de Espectrometria de Massa, EMBRAPA Recursos Genéticos e Biotecnologia, Brası́lia 70770-917, Brazil
- Laboratório de Sı́ntese e Análise de Biomoléculas, Instituto de Quı́mica, Universidade de Brası́lia, Brası́lia 70910-900, Brazil
| | - Alexandra Plácido
- LAQV/REQUIMTE, Departamento de Quı́mica e Bioquı́mica, Faculdade de Ciéncias da Universidade do Porto, 4169-007 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal
| | - Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal
| | - Camila C Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal
| | - Beatriz G de la Torre
- KwaZulu-Natal Research Innovation and Sequencing Platform, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Fernando Albericio
- KwaZulu-Natal Research Innovation and Sequencing Platform, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
- CIBER-BBN (Networking Centre on Bioengineering, Biomaterials and Nanomedicine) and Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
| | - Néstor G Basso
- Instituto de Diversidad y Evolución Austral, Consejo Nacional de Investigaciones Cientı́ficas y Técnicas (IDEAus-CONICET), Bv. Almirante Brown 2915, Puerto Madryn U9120ACD, Argentina
| | - José R Leite
- Área de Morfologia, Faculdade de Medicina, Universidade de Brası́lia, Brası́lia 70910-900, Brazil
| | - Mariela M Marani
- Instituto Patagónico para el Estudio de los Ecosistemas Continentales, Consejo Nacional de Investigaciones Cientı́ficas y Técnicas (IPEEC-CONICET), Bv. Almirante Brown 2915, Puerto Madryn U9120ACD, Argentina
| |
Collapse
|
110
|
Lee MW, de Anda J, Kroll C, Bieniossek C, Bradley K, Amrein KE, Wong GCL. How do cyclic antibiotics with activity against Gram-negative bacteria permeate membranes? A machine learning informed experimental study. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183302. [PMID: 32311341 DOI: 10.1016/j.bbamem.2020.183302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/30/2022]
Abstract
All antibiotics have to engage bacterial amphiphilic barriers such as the lipopolysaccharide-rich outer membrane or the phospholipid-based inner membrane in some manner, either by disrupting them outright and/or permeating them and thereby allow the antibiotic to get into bacteria. There is a growing class of cyclic antibiotics, many of which are of bacterial origin, that exhibit activity against Gram-negative bacteria, which constitute an urgent problem in human health. We examine a diverse collection of these cyclic antibiotics, both natural and synthetic, which include bactenecin, polymyxin B, octapeptin, capreomycin, and Kirshenbaum peptoids, in order to identify what they have in common when they interact with bacterial lipid membranes. We find that they virtually all have the ability to induce negative Gaussian curvature (NGC) in bacterial membranes, the type of curvature geometrically required for permeation mechanisms such as pore formation, blebbing, and budding. This is interesting since permeation of membranes is a function usually ascribed to antimicrobial peptides (AMPs) from innate immunity. As prototypical test cases of cyclic antibiotics, we analyzed amino acid sequences of bactenecin, polymyxin B, and capreomycin using our recently developed machine-learning classifier trained on α-helical AMP sequences. Although the original classifier was not trained on cyclic antibiotics, a modified classifier approach correctly predicted that bactenecin and polymyxin B have the ability to induce NGC in membranes, while capreomycin does not. Moreover, the classifier was able to recapitulate empirical structure-activity relationships from alanine scans in polymyxin B surprisingly well. These results suggest that there exists some common ground in the sequence design of hybrid cyclic antibiotics and linear AMPs.
Collapse
Affiliation(s)
- Michelle W Lee
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Jaime de Anda
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Carsten Kroll
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Christoph Bieniossek
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Kenneth Bradley
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Kurt E Amrein
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Gerard C L Wong
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
111
|
Strandberg E, Bentz D, Wadhwani P, Bürck J, Ulrich AS. Terminal charges modulate the pore forming activity of cationic amphipathic helices. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183243. [DOI: 10.1016/j.bbamem.2020.183243] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/14/2020] [Accepted: 02/18/2020] [Indexed: 11/15/2022]
|
112
|
Drug Conjugation Induced Modulation of Structural and Membrane Interaction Features of Cationic Cell-Permeable Peptides. Int J Mol Sci 2020; 21:ijms21062197. [PMID: 32235796 PMCID: PMC7139830 DOI: 10.3390/ijms21062197] [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: 03/03/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
Cell-penetrating peptides might have great potential for enhancing the therapeutic effect of drug molecules against such dangerous pathogens as Mycobacterium tuberculosis (Mtb), which causes a major health problem worldwide. A set of cationic cell-penetration peptides with various hydrophobicity were selected and synthesized as drug carrier of isoniazid (INH), a first-line antibacterial agent against tuberculosis. Molecular interactions between the peptides and their INH-conjugates with cell-membrane-forming lipid layers composed of DPPC and mycolic acid (a characteristic component of Mtb cell wall) were evaluated, using the Langmuir balance technique. Secondary structure of the INH conjugates was analyzed and compared to that of the native peptides by circular dichroism spectroscopic experiments performed in aqueous and membrane mimetic environment. A correlation was found between the conjugation induced conformational and membrane affinity changes of the INH-peptide conjugates. The degree and mode of interaction were also characterized by AFM imaging of penetrated lipid layers. In vitro biological evaluation was performed with Penetratin and Transportan conjugates. Results showed similar internalization rate into EBC-1 human squamous cell carcinoma, but markedly different subcellular localization and activity on intracellular Mtb.
Collapse
|
113
|
Tuerkova A, Kabelka I, Králová T, Sukeník L, Pokorná Š, Hof M, Vácha R. Effect of helical kink in antimicrobial peptides on membrane pore formation. eLife 2020; 9:47946. [PMID: 32167466 PMCID: PMC7069690 DOI: 10.7554/elife.47946] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 02/18/2020] [Indexed: 12/19/2022] Open
Abstract
Every cell is protected by a semipermeable membrane. Peptides with the right properties, for example Antimicrobial peptides (AMPs), can disrupt this protective barrier by formation of leaky pores. Unfortunately, matching peptide properties with their ability to selectively form pores in bacterial membranes remains elusive. In particular, the proline/glycine kink in helical peptides was reported to both increase and decrease antimicrobial activity. We used computer simulations and fluorescence experiments to show that a kink in helices affects the formation of membrane pores by stabilizing toroidal pores but disrupting barrel-stave pores. The position of the proline/glycine kink in the sequence further controls the specific structure of toroidal pore. Moreover, we demonstrate that two helical peptides can form a kink-like connection with similar behavior as one long helical peptide with a kink. The provided molecular-level insight can be utilized for design and modification of pore-forming antibacterial peptides or toxins.
Collapse
Affiliation(s)
- Alzbeta Tuerkova
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice, Czech Republic
| | - Ivo Kabelka
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice, Czech Republic
| | - Tereza Králová
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice, Czech Republic
| | - Lukáš Sukeník
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice, Czech Republic.,Department of Condensed Matter Physics, Faculty of Science, Masaryk University, Kotlářská, Czech Republic
| | - Šárka Pokorná
- J. Heyrovsky Institute of Physical Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Hof
- J. Heyrovsky Institute of Physical Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Robert Vácha
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice, Czech Republic.,Department of Condensed Matter Physics, Faculty of Science, Masaryk University, Kotlářská, Czech Republic
| |
Collapse
|
114
|
The effect of chitosan and PEG polymers on stabilization of GF-17 structure: A molecular dynamics study. Carbohydr Polym 2020; 237:116124. [PMID: 32241401 DOI: 10.1016/j.carbpol.2020.116124] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 01/18/2023]
Abstract
We examine the interactions of chitosan and polyethylene glycol (PEG) with antimicrobial peptide GF-17 to identify a suitable carrier to improve the peptide drug delivery systems. To this end, the molecular dynamics simulations are used to determine the interactions of a typical antimicrobial peptide GF-17 with the chitosan and PEG polymers. The findings indicate the great potential of the peptide to maintain its secondary structure in the adjacent to chitosan polymers. During the interaction with chitosan polymers, the structure of the peptide has smaller fluctuations compared to the PEG polymers. Also, in the presence of both the polymers, the PEG polymers are situated closer to the peptide than chitosan polymers. Moreover, the analysis indicates that the acidic residues and phenylalanine play a crucial role in peptide-polymer interactions. This research provides a valuable insight into the design of polymer surfaces for drug delivery applications such as controlled-release peptide delivery systems.
Collapse
|
115
|
Al-Adwani S, Wallin C, Balhuizen MD, Veldhuizen EJA, Coorens M, Landreh M, Végvári Á, Smith ME, Qvarfordt I, Lindén A, Gräslund A, Agerberth B, Bergman P. Studies on citrullinated LL-37: detection in human airways, antibacterial effects and biophysical properties. Sci Rep 2020; 10:2376. [PMID: 32047184 PMCID: PMC7012854 DOI: 10.1038/s41598-020-59071-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/23/2020] [Indexed: 11/09/2022] Open
Abstract
Arginine residues of the antimicrobial peptide LL-37 can be citrullinated by peptidyl arginine deiminases, which reduce the positive charge of the peptide. Notably, citrullinated LL-37 has not yet been detected in human samples. In addition, functional and biophysical properties of citrullinated LL-37 are not fully explored. The aim of this study was to detect citrullinated LL-37 in human bronchoalveolar lavage (BAL) fluid and to determine antibacterial and biophysical properties of citrullinated LL-37. BAL fluid was obtained from healthy human volunteers after intra-bronchial exposure to lipopolysaccharide. Synthetic peptides were used for bacterial killing assays, transmission electron microscopy, isothermal titration calorimetry, mass-spectrometry and circular dichroism. Using targeted proteomics, we were able to detect both native and citrullinated LL-37 in BAL fluid. The citrullinated peptide did not kill Escherichia coli nor lysed human red blood cells. Both peptides had similar α-helical secondary structures but citrullinated LL-37 was more stable at higher temperatures, as shown by circular dichroism. In conclusion, citrullinated LL-37 is present in the human airways and citrullination impaired bacterial killing, indicating that a net positive charge is important for antibacterial and membrane lysing effects. It is possible that citrullination serves as a homeostatic regulator of AMP-function by alteration of key functions.
Collapse
Affiliation(s)
- Salma Al-Adwani
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden.,Department of Animal and Veterinary Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat, Oman
| | - Cecilia Wallin
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Melanie D Balhuizen
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Edwin J A Veldhuizen
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Maarten Coorens
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
| | - Michael Landreh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ákos Végvári
- Division of Physiological Chemistry I, Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Margaretha E Smith
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingemar Qvarfordt
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Birgitta Agerberth
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bergman
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden. .,Infectious Disease Clinic, Immunodeficiency Unit, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
116
|
Della Pelle G, Perà G, Belardinelli MC, Gerdol M, Felli M, Crognale S, Scapigliati G, Ceccacci F, Buonocore F, Porcelli F. Trematocine, a Novel Antimicrobial Peptide from the Antarctic Fish Trematomus bernacchii: Identification and Biological Activity. Antibiotics (Basel) 2020; 9:E66. [PMID: 32041161 PMCID: PMC7168153 DOI: 10.3390/antibiotics9020066] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 01/02/2023] Open
Abstract
Antimicrobial peptides (AMPs) are short peptides active against a wide range of pathogens and, therefore, they are considered a useful alternative to conventional antibiotics. We have identified a new AMP in a transcriptome derived from the Antarctic fish Trematomus bernacchii. This peptide, named Trematocine, has been investigated for its expression both at the basal level and after in vivo immunization with an endemic Antarctic bacterium (Psychrobacter sp. TAD1). Results agree with the expected behavior of a fish innate immune component, therefore we decided to synthesize the putative mature sequence of Trematocine to determine the structure, the interaction with biological membranes, and the biological activity. We showed that Trematocine folds into a α-helical structure in the presence of both zwitterionic and anionic charged vesicles. We demonstrated that Trematocine has a highly specific interaction with anionic charged vesicles and that it can kill Gram-negative bacteria, possibly via a carpet like mechanism. Moreover, Trematocine showed minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values against selected Gram-positive and Gram-negative bacteria similar to other AMPs isolated from Antarctic fishes. The peptide is a possible candidate for a new drug as it does not show any haemolytic or cytotoxic activity against mammalian cells at the concentration needed to kill the tested bacteria.
Collapse
Affiliation(s)
- Giulia Della Pelle
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (G.D.P.); (G.P.); (M.C.B.); (M.F.); (S.C.); (G.S.); (F.P.)
| | - Giulia Perà
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (G.D.P.); (G.P.); (M.C.B.); (M.F.); (S.C.); (G.S.); (F.P.)
| | - Maria Cristina Belardinelli
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (G.D.P.); (G.P.); (M.C.B.); (M.F.); (S.C.); (G.S.); (F.P.)
| | - Marco Gerdol
- Department of Life Sciences, University of Trieste, Trieste 34128, Italy;
| | - Martina Felli
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (G.D.P.); (G.P.); (M.C.B.); (M.F.); (S.C.); (G.S.); (F.P.)
| | - Silvia Crognale
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (G.D.P.); (G.P.); (M.C.B.); (M.F.); (S.C.); (G.S.); (F.P.)
| | - Giuseppe Scapigliati
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (G.D.P.); (G.P.); (M.C.B.); (M.F.); (S.C.); (G.S.); (F.P.)
| | - Francesca Ceccacci
- CNR—Institute for Biological Systems, Sede Secondaria di Roma-Meccanismi di Reazione, 00185 Rome, Italy;
| | - Francesco Buonocore
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (G.D.P.); (G.P.); (M.C.B.); (M.F.); (S.C.); (G.S.); (F.P.)
| | - Fernando Porcelli
- Department for Innovation in Biological, Agrofood and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (G.D.P.); (G.P.); (M.C.B.); (M.F.); (S.C.); (G.S.); (F.P.)
| |
Collapse
|
117
|
Bertrand B, Munusamy S, Espinosa-Romero JF, Corzo G, Arenas Sosa I, Galván-Hernández A, Ortega-Blake I, Hernández-Adame PL, Ruiz-García J, Velasco-Bolom JL, Garduño-Juárez R, Munoz-Garay C. Biophysical characterization of the insertion of two potent antimicrobial peptides-Pin2 and its variant Pin2[GVG] in biological model membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183105. [DOI: 10.1016/j.bbamem.2019.183105] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/04/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022]
|
118
|
Effective Therapeutic Drug Delivery by GALA3, an Endosomal Escape Peptide with Reduced Hydrophobicity. J Membr Biol 2020; 253:139-152. [DOI: 10.1007/s00232-020-00109-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/20/2020] [Indexed: 12/18/2022]
|
119
|
Kamysz E, Sikorska E, Jaśkiewicz M, Bauer M, Neubauer D, Bartoszewska S, Barańska-Rybak W, Kamysz W. Lipidated Analogs of the LL-37-Derived Peptide Fragment KR12-Structural Analysis, Surface-Active Properties and Antimicrobial Activity. Int J Mol Sci 2020; 21:E887. [PMID: 32019109 PMCID: PMC7036753 DOI: 10.3390/ijms21030887] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 02/06/2023] Open
Abstract
An increasing number of multidrug-resistant pathogens is a serious problem of modern medicine and new antibiotics are highly demanded. In this study, different n-alkyl acids (C2-C14) and aromatic acids (benzoic and trans-cinnamic) were conjugated to the N-terminus of KR12 amide. The effect of this modification on antimicrobial activity (ESKAPE bacteria and biofilm of Staphylococcus aureus) and cytotoxicity (human red blood cells and HaCaT cell line) was examined. The effect of lipophilic modifications on helicity was studied by CD spectroscopy, whereas peptide self-assembly was studied by surface tension measurements and NMR spectroscopy. As shown, conjugation of the KR12-NH2 peptide with C4-C14 fatty acid chains enhanced the antimicrobial activity with an optimum demonstrated by C8-KR12-NH2 (MIC 1-4 μg/mL against ESKAPE strains; MBEC of S. aureus 4-16 μg/mL). Correlation between antimicrobial activity and self-assembly behavior of C14-KR12-NH2 and C8-KR12-NH2 has shown that the former self-assembled into larger aggregated structures, which reduced its antimicrobial activity. In conclusion, N-terminal modification can enhance antimicrobial activity of KR12-NH2; however, at the same time, the cytotoxicity increases. It seems that the selectivity against pathogens over human cells can be achieved through conjugation of peptide N-terminus with appropriate n-alkyl fatty and aromatic acids.
Collapse
Affiliation(s)
- Elżbieta Kamysz
- Laboratory of Chemistry of Biological Macromolecules, Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdańsk, 80-308 Gdańsk, Poland
| | - Emilia Sikorska
- Laboratory of Structural Studies of Biopolymers, Department of Organic Chemistry, Faculty of Chemistry, University of Gdańsk, 80-308 Gdańsk, Poland;
| | - Maciej Jaśkiewicz
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.J.); (M.B.); (D.N.); (S.B.); (W.K.)
| | - Marta Bauer
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.J.); (M.B.); (D.N.); (S.B.); (W.K.)
| | - Damian Neubauer
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.J.); (M.B.); (D.N.); (S.B.); (W.K.)
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.J.); (M.B.); (D.N.); (S.B.); (W.K.)
| | - Wioletta Barańska-Rybak
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdańsk, 80-214 Gdańsk, Poland;
| | - Wojciech Kamysz
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.J.); (M.B.); (D.N.); (S.B.); (W.K.)
| |
Collapse
|
120
|
Feese E, Gracz HS, Boyle PD, Ghiladi RA. Towards microbe-targeted photosensitizers: Synthesis, characterization and in vitro photodynamic inactivation of the tuberculosis model pathogen M. smegmatis by porphyrin-peptide conjugates. J PORPHYR PHTHALOCYA 2020. [DOI: 10.1142/s1088424619501505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Porphyrin-peptide conjugates have a breadth of potential applications, including use in photodynamic therapy, boron neutron capture therapy, as fluorescence imaging tags for tracking subcellular localization, as magnetic resonance imaging (MRI) positive-contrast reagents and as biomimetic catalysts. Here, we have explored three general routes to porphyrin-peptide conjugates using the Cu(I)-catalyzed Huisgen-Medal-Sharpless 1,3-dipolar cycloaddition of peptide-containing azides with a terminal alkyne-containing porphyrin, thereby generating porphyrin-peptide conjugates (PPCs) comprised of a cationic porphyrin coupled to short antimicrobial peptides. In addition to characterizing the PPCs using a variety of spectroscopic (UV-vis, [Formula: see text]H- and [Formula: see text]C-NMR) and mass spectrometric methods, we evaluated their efficacy as photosensitizers for the in vitro photodynamic inactivation of Mycobacterium smegmatis as a model for the pathogen Mycobacterium tuberculosis. Difficulties that needed to be overcome for the efficient synthesis of PPCs were the limited solubility of the quaternized pyridyl porphyrin in common solvents, undesired (de)metallation and transmetallation, and chromatographic purification. Photodynamic inactivation studies of a small library of PPCs against Mycobacterium smegmatis confirmed our hypothesis that the porphyrin-based photosensitizer maintains its ability to efficiently inactivate bacteria when conjugated to a small peptide by upwards of 5–6 log units (99.999[Formula: see text]%) using white light illumination (400–700 nm, 60 mW/cm[Formula: see text], 30 min). Further, hemolysis assays revealed the lack of toxicity of the PPCs against sheep blood at concentrations employed for in vitro photodynamic inactivation. Taken together, the results demonstrated the ability of PPCs to maintain their antimicrobial photodynamic inactivation efficacy when possessing a short cationic peptides for enabling the potential targeting of pathogens in vivo.
Collapse
Affiliation(s)
- Elke Feese
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina, 27695-8204, USA
| | - Hanna S. Gracz
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina, USA
| | - Paul D. Boyle
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina, 27695-8204, USA
| | - Reza A. Ghiladi
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina, 27695-8204, USA
| |
Collapse
|
121
|
Malhotra K, Shankar S, Chauhan N, Rai R, Singh Y. Design, characterization, and evaluation of antibacterial gels, Boc-D-Phe-γ 4-L-Phe-PEA/chitosan and Boc-L-Phe-γ 4-L-Phe-PEA/chitosan, for biomaterial-related infections. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110648. [PMID: 32204079 DOI: 10.1016/j.msec.2020.110648] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 08/29/2019] [Accepted: 01/03/2020] [Indexed: 12/27/2022]
Abstract
Self-assembled peptide gels have generated interest as antibacterial materials to prevent biomaterial-related infections but these peptides are often associated with poor proteolytic stability. Efforts have been made to stabilize peptides by incorporating non-natural amino acids and/or linkages but complexation with polymers have not been explored. Therefore, we developed self-assembled peptide/chitosan gels, Boc-D-Phe-γ4-L-Phe-PEA (NH007)/chitosan and Boc-L-Phe-γ4-L-Phe-PEA (NH009)/chitosan, by complexing dipeptide NH007 or NH009 with chitosan in DMSO:acetic acid. The gels were characterized using SEM, FTIR, contact angle, and rheology data and found to exhibit excellent viscoelastic and self-healing characteristics. Complexation with chitosan led to an increase in stability against proteolytic degradation. Peptide/chitosan gels showed broad spectrum antibacterial activities against Gram-negative and Gram-positive bacteria, such as Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, and Bacillus subtilis at a high inoculum of 107-108 cfu/mL. NH007/chitosan gels showed 70-75% inhibition, whereas NH009/chitosan showed 78-81% inhibition and NH009/chitosan gels, in particular, showed strong antibacterial activity against pathogenic strain of P. aeruginosa. A unique feature of these gels is that the antibacterial activities did not decrease gradually but were sustained for up to 48 h. The mechanistic studies using SEM and HR-TEM indicated interaction of gels with bacterial membrane components, leading to cell lysis. The MTT and LDH assays indicated >90% cell viability and only 8-10% toxicity towards NIH 3T3 fibroblast cells. Thus, peptide/chitosan gels developed in the present work showed improved proteolytic stability and sustained antibacterial activities and, therefore, may be used for preventing biomaterial-related infections.
Collapse
Affiliation(s)
- Kamal Malhotra
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar 140001, Punjab, India
| | - Sudha Shankar
- Medicinal Chemistry Division, CSIR- Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi 180001, Jammu and Kashmir, India; Academy of Scientific and Innovative Research, New Delhi 110001, Delhi, India
| | - Neelam Chauhan
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar 140001, Punjab, India
| | - Rajkishor Rai
- Medicinal Chemistry Division, CSIR- Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi 180001, Jammu and Kashmir, India; Academy of Scientific and Innovative Research, New Delhi 110001, Delhi, India
| | - Yashveer Singh
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar 140001, Punjab, India.
| |
Collapse
|
122
|
Lee Y, Yamada H, Pradipta A, Ma JS, Okamoto M, Nagaoka H, Takashima E, Standley DM, Sasai M, Takei K, Yamamoto M. Initial phospholipid-dependent Irgb6 targeting to Toxoplasma gondii vacuoles mediates host defense. Life Sci Alliance 2019; 3:3/1/e201900549. [PMID: 31852733 PMCID: PMC6925386 DOI: 10.26508/lsa.201900549] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 12/19/2022] Open
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite capable of infecting warm-blooded animals by ingestion. The organism enters host cells and resides in the cytoplasm in a membrane-bound parasitophorous vacuole (PV). Inducing an interferon response enables IFN-γ-inducible immunity-related GTPase (IRG protein) to accumulate on the PV and to restrict parasite growth. However, little is known about the mechanisms by which IRG proteins recognize and destroy T. gondii PV. We characterized the role of IRG protein Irgb6 in the cell-autonomous response against T. gondii, which involves vacuole ubiquitination and breakdown. We show that Irgb6 is capable of binding a specific phospholipid on the PV membrane. Furthermore, the absence of Irgb6 causes reduced targeting of other effector IRG proteins to the PV. This suggests that Irgb6 has a role as a pioneer in the process by which multiple IRG proteins access the PV. Irgb6-deficient mice are highly susceptible to infection by a strain of T. gondii avirulent in wild-type mice.
Collapse
Affiliation(s)
- Youngae Lee
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hiroshi Yamada
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ariel Pradipta
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ji Su Ma
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masaaki Okamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Ehime, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Ehime, Japan
| | - Daron M Standley
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Kohji Takei
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan .,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
123
|
Kintses B, Jangir PK, Fekete G, Számel M, Méhi O, Spohn R, Daruka L, Martins A, Hosseinnia A, Gagarinova A, Kim S, Phanse S, Csörgő B, Györkei Á, Ari E, Lázár V, Nagy I, Babu M, Pál C, Papp B. Chemical-genetic profiling reveals limited cross-resistance between antimicrobial peptides with different modes of action. Nat Commun 2019; 10:5731. [PMID: 31844052 PMCID: PMC6915728 DOI: 10.1038/s41467-019-13618-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/14/2019] [Indexed: 11/09/2022] Open
Abstract
Antimicrobial peptides (AMPs) are key effectors of the innate immune system and promising therapeutic agents. Yet, knowledge on how to design AMPs with minimal cross-resistance to human host-defense peptides remains limited. Here, we systematically assess the resistance determinants of Escherichia coli against 15 different AMPs using chemical-genetics and compare to the cross-resistance spectra of laboratory-evolved AMP-resistant strains. Although generalizations about AMP resistance are common in the literature, we find that AMPs with different physicochemical properties and cellular targets vary considerably in their resistance determinants. As a consequence, cross-resistance is prevalent only between AMPs with similar modes of action. Finally, our screen reveals several genes that shape susceptibility to membrane- and intracellular-targeting AMPs in an antagonistic manner. We anticipate that chemical-genetic approaches could inform future efforts to minimize cross-resistance between therapeutic and human host AMPs.
Collapse
Affiliation(s)
- Bálint Kintses
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
- HCEMM-BRC Translational Microbiology Lab, Szeged, Hungary.
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary.
| | - Pramod K Jangir
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Gergely Fekete
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Mónika Számel
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Orsolya Méhi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Réka Spohn
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ana Martins
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Ali Hosseinnia
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Alla Gagarinova
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Sunyoung Kim
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Sadhna Phanse
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Bálint Csörgő
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Department of Microbiology and Immunology, University of California, San Francisco, USA
| | - Ádám Györkei
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Eszter Ari
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Viktória Lázár
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - István Nagy
- Sequencing Platform, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Mohan Babu
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary.
| |
Collapse
|
124
|
Omolo CA, Megrab NA, Kalhapure RS, Agrawal N, Jadhav M, Mocktar C, Rambharose S, Maduray K, Nkambule B, Govender T. Liposomes with pH responsive 'on and off' switches for targeted and intracellular delivery of antibiotics. J Liposome Res 2019; 31:45-63. [PMID: 31663407 DOI: 10.1080/08982104.2019.1686517] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
pH responsive drug delivery systems are one of the new strategies to address the spread of bacterial resistance to currently used antibiotics. The aim of this study was to formulate liposomes with 'On' and 'Off'' pH responsive switches for infection site targeting. The vancomycin (VCM) loaded liposomes had sizes below 100 nm, at pH 7.4. The QL-liposomes had a negative zeta potential at pH 7.4 that switched to a positive charge at acidic pH. VCM release from the liposome was quicker at pH 6 than pH 7.4. The OA-QL-liposome showed 4-fold lower MIC at pH 7.4 and 8- and 16-fold lower at pH 6.0 against both MSSA and MRSA compared to the bare drug. OA-QL liposome had a 1266.67- and 704.33-fold reduction in the intracellular infection for TPH-1 macrophage and HEK293 cells respectively. In vivo studies showed that the amount of MRSA recovered from mice treated with formulations was 189.67 and 6.33-fold lower than the untreated and bare VCM treated mice respectively. MD simulation of the QL lipid with the phosphatidylcholine membrane (POPC) showed spontaneous binding of the lipid to the bilayer membrane both electrostatic and Van der Waals interactions contributed to the binding. These studies demonstrated that the 'On' and 'Off' pH responsive liposomes enhanced the activity targeted and intracellular delivery VCM.
Collapse
Affiliation(s)
- Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,School of Pharmacy and Health Sciences, United States International University of Africa, Nairobi, Kenya
| | - Nagia A Megrab
- Department of Pharmaceutics and Industrial Pharmacy, Zagazig University, Zagazig, Egypt
| | - Rahul S Kalhapure
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Nikhil Agrawal
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Mahantesh Jadhav
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Chunderika Mocktar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sanjeev Rambharose
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,Division of Emergency Medicine, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Kaminee Maduray
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Bongani Nkambule
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
125
|
Vicente FM, González-Garcia M, Diaz Pico E, Moreno-Castillo E, Garay HE, Rosi PE, Jimenez AM, Campos-Delgado JA, Rivera DG, Chinea G, Pietro RCL, Stenger S, Spellerberg B, Kubiczek D, Bodenberger N, Dietz S, Rosenau F, Paixão MW, Ständker L, Otero-González AJ. Design of a Helical-Stabilized, Cyclic, and Nontoxic Analogue of the Peptide Cm-p5 with Improved Antifungal Activity. ACS OMEGA 2019; 4:19081-19095. [PMID: 31763531 PMCID: PMC6868880 DOI: 10.1021/acsomega.9b02201] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/04/2019] [Indexed: 05/15/2023]
Abstract
Following the information obtained by a rational design study, a cyclic and helical-stabilized analogue of the peptide Cm-p5 was synthetized. The cyclic monomer showed an increased activity in vitro against Candida albicans and Candida parapsilosis, compared to Cm-p5. Initially, 14 mutants of Cm-p5 were synthesized following a rational design to improve the antifungal activity and pharmacological properties. Antimicrobial testing showed that the activity was lost in each of these 14 analogues, suggesting, as a main conclusion, that a Glu-His salt bridge could stabilize Cm-p5 helical conformation during the interaction with the plasma membrane. A derivative, obtained by substitution of Glu and His for Cys, was synthesized and oxidized with the generation of a cyclic monomer with improved antifungal activity. In addition, two dimers were generated during the oxidation procedure, a parallel and antiparallel one. The dimers showed a helical secondary structure in water, whereas the cyclic monomer only showed this conformation in SDS. Molecular dynamic simulations confirmed the helical stabilizations for all of them, therefore indicating the possible essential role of the Glu-His salt bridge. In addition, the antiparallel dimer showed a moderate activity against Pseudomonas aeruginosa and a significant activity against Listeria monocytogenes. Neither the cyclic monomer nor the dimers were toxic against macrophages or THP-1 human cells. Due to its increased capacity for fungal control compared to fluconazole, its low cytotoxicity, together with a stabilized α-helix and disulfide bridges, that may advance its metabolic stability, and in vivo activity, the new cyclic Cm-p5 monomer represents a potential systemic antifungal therapeutic candidate.
Collapse
Affiliation(s)
- Fidel
E. Morales Vicente
- General
Chemistry Department, Faculty of Chemistry and Center for Natural Products Research,
Faculty of Chemistry, University of Havana, Zapata y G, 10400 La Habana, Cuba
- Synthetic
Peptides Group, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, 10600 La Habana, Cuba
- Center
of Excellence for Research in Sustainable Chemistry (CERSusChem),
Department of Chemistry, Federal University
of São Carlos-UFSCar, São Paulo 13565-905, Brazil
| | - Melaine González-Garcia
- Center
for Protein Studies, Faculty of Biology, University of Havana, 25 and I, 10400 La Habana, Cuba
| | - Erbio Diaz Pico
- Synthetic
Peptides Group, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, 10600 La Habana, Cuba
| | - Elena Moreno-Castillo
- General
Chemistry Department, Faculty of Chemistry and Center for Natural Products Research,
Faculty of Chemistry, University of Havana, Zapata y G, 10400 La Habana, Cuba
| | - Hilda E. Garay
- Synthetic
Peptides Group, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, 10600 La Habana, Cuba
| | - Pablo E. Rosi
- Department
of Inorganic Chemistry, Analytical and Physical Chemistry, Facultad
de Ciencias Exactas y Naturales, Universidad
de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Asiel Mena Jimenez
- General
Chemistry Department, Faculty of Chemistry and Center for Natural Products Research,
Faculty of Chemistry, University of Havana, Zapata y G, 10400 La Habana, Cuba
| | - Jose A. Campos-Delgado
- Center
of Excellence for Research in Sustainable Chemistry (CERSusChem),
Department of Chemistry, Federal University
of São Carlos-UFSCar, São Paulo 13565-905, Brazil
| | - Daniel G. Rivera
- General
Chemistry Department, Faculty of Chemistry and Center for Natural Products Research,
Faculty of Chemistry, University of Havana, Zapata y G, 10400 La Habana, Cuba
| | - Glay Chinea
- Synthetic
Peptides Group, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, 10600 La Habana, Cuba
| | - Rosemeire C. L.
R. Pietro
- Laboratory
of Pharmaceutical Biotechnology, Department of Drugs and Medicines,
School of Pharmaceutical Sciences, UNESP, Araraquara 14800-900, Brazil
| | - Steffen Stenger
- Institute
of Medical Microbiology and Hygiene, University
Clinic of Ulm, Robert Koch Str. 8, Ulm D-89081, Germany
| | - Barbara Spellerberg
- Institute
of Medical Microbiology and Hygiene, University
Clinic of Ulm, Robert Koch Str. 8, Ulm D-89081, Germany
| | - Dennis Kubiczek
- Institute
of Pharmaceutical Biotechnology, Ulm University, James-Frank-Ring N27, 89081 Ulm, Germany
| | - Nicholas Bodenberger
- Institute
of Pharmaceutical Biotechnology, Ulm University, James-Frank-Ring N27, 89081 Ulm, Germany
| | - Steffen Dietz
- Institute
of Pharmaceutical Biotechnology, Ulm University, James-Frank-Ring N27, 89081 Ulm, Germany
| | - Frank Rosenau
- Institute
of Pharmaceutical Biotechnology, Ulm University, James-Frank-Ring N27, 89081 Ulm, Germany
| | - Márcio Weber Paixão
- Center
of Excellence for Research in Sustainable Chemistry (CERSusChem),
Department of Chemistry, Federal University
of São Carlos-UFSCar, São Paulo 13565-905, Brazil
- E-mail: (W.P.)
| | - Ludger Ständker
- Core
Facility for Functional Peptidomics, Ulm Peptide Pharmaceuticals (U-PEP),
University Ulm, Faculty of Medicine, Ulm
University, 89081 Ulm, Germany
- E-mail: (L.S.)
| | - Anselmo J. Otero-González
- Center
for Protein Studies, Faculty of Biology, University of Havana, 25 and I, 10400 La Habana, Cuba
- E-mail: (A.J.O.-G.)
| |
Collapse
|
126
|
Mwangi J, Hao X, Lai R, Zhang ZY. Antimicrobial peptides: new hope in the war against multidrug resistance. Zool Res 2019; 40:488-505. [PMID: 31592585 PMCID: PMC6822926 DOI: 10.24272/j.issn.2095-8137.2019.062] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/26/2019] [Indexed: 12/16/2022] Open
Abstract
The discovery of antibiotics marked a golden age in the revolution of human medicine. However, decades later, bacterial infections remain a global healthcare threat, and a return to the pre-antibiotic era seems inevitable if stringent measures are not adopted to curb the rapid emergence and spread of multidrug resistance and the indiscriminate use of antibiotics. In hospital settings, multidrug resistant (MDR) pathogens, including carbapenem-resistant Pseudomonas aeruginosa, vancomycin-resistant enterococci (VRE), methicillin-resistant Staphylococcus aureus (MRSA), and extended-spectrum β-lactamases (ESBL) bearing Acinetobacter baumannii, Escherichia coli, and Klebsiella pneumoniae are amongst the most problematic due to the paucity of treatment options, increased hospital stay, and exorbitant medical costs. Antimicrobial peptides (AMPs) provide an excellent potential strategy for combating these threats. Compared to empirical antibiotics, they show low tendency to select for resistance, rapid killing action, broad-spectrum activity, and extraordinary clinical efficacy against several MDR strains. Therefore, this review highlights multidrug resistance among nosocomial bacterial pathogens and its implications and reiterates the importance of AMPs as next-generation antibiotics for combating MDR superbugs.
Collapse
Affiliation(s)
- James Mwangi
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming Yunnan 650204, China
- Sino-African Joint Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China
| | - Xue Hao
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China
| | - Ren Lai
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China
- Sino-African Joint Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan Hubei 430071, China
| | - Zhi-Ye Zhang
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China, E-mail:
| |
Collapse
|
127
|
Rončević T, Puizina J, Tossi A. Antimicrobial Peptides as Anti-Infective Agents in Pre-Post-Antibiotic Era? Int J Mol Sci 2019; 20:E5713. [PMID: 31739573 PMCID: PMC6887943 DOI: 10.3390/ijms20225713] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Resistance to antibiotics is one of the main current threats to human health and every year multi-drug resistant bacteria are infecting millions of people worldwide, with many dying as a result. Ever since their discovery, some 40 years ago, the antimicrobial peptides (AMPs) of innate defense have been hailed as a potential alternative to conventional antibiotics due to their relatively low potential to elicit resistance. Despite continued effort by both academia and start-ups, currently there are still no antibiotics based on AMPs in use. In this study, we discuss what we know and what we do not know about these agents, and what we need to know to successfully translate discovery to application. Understanding the complex mechanics of action of these peptides is the main prerequisite for identifying and/or designing or redesigning novel molecules with potent biological activity. However, other aspects also need to be well elucidated, i.e., the (bio)synthetic processes, physiological and pathological contexts of their activity, and a quantitative understanding of how physico-chemical properties affect activity. Research groups worldwide are using biological, biophysical, and algorithmic techniques to develop models aimed at designing molecules with the necessary blend of antimicrobial potency and low toxicity. Shedding light on some open questions may contribute toward improving this process.
Collapse
Affiliation(s)
- Tomislav Rončević
- Department of Biology, Faculty of Science, University of Split, 21000 Split, Croatia;
- Laboratory for Aquaculture, Institute of Oceanography and Fisheries, 21000 Split, Croatia
| | - Jasna Puizina
- Department of Biology, Faculty of Science, University of Split, 21000 Split, Croatia;
| | - Alessandro Tossi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| |
Collapse
|
128
|
Arteaga V, Lamas A, Regal P, Vázquez B, Miranda JM, Cepeda A, Franco CM. Antimicrobial activity of apitoxin from Apis mellifera in Salmonella enterica strains isolated from poultry and its effects on motility, biofilm formation and gene expression. Microb Pathog 2019; 137:103771. [PMID: 31580958 DOI: 10.1016/j.micpath.2019.103771] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 10/25/2022]
Abstract
Salmonella is a major global food-borne pathogen. One of the main concerns related to Salmonella and other food-borne pathogens is their capacity to acquire antimicrobial resistance and produce biofilms. Due to the increased resistance to common antimicrobials used to treat livestock animals and human infections, the discovery of new antimicrobial substances is one of the main challenges in microbiological research. An additional challenge is the development of new methods and substances to inhibit and destruct biofilms. We determined the antimicrobial and antibiofilm activities of apitoxin in 16 Salmonella strains isolated from poultry. In addition, the effect of apitoxin on Salmonella motility and the expression of biofilm- and virulence-related genes was evaluated. The minimum inhibitory concentrations (MIC) of apitoxin ranged from 1,024-256 μg/mL, with 512 μg/mL being the most common. Sub-inhibitory concentrations of apitoxin significantly reduced biofilm formation in 14 of the 16 Salmonella strains tested, with significant increases in motility. MIC concentrations of apitoxin destroyed the pre-formed biofilm by 27.66-68.22% (47.00% ± 10.91). The expression of biofilm- and virulence-related genes and small RNAs was differentially regulated according to the strain and the presence of apitoxin. The transcription of the small RNAs dsrA and csrB, related to antimicrobial resistance, was upregulated in the presence of apitoxin. We suggest that apitoxin is a potential antimicrobial substance that could be used in combination with other substances to develop new drugs and sanitizers against food-borne pathogens.
Collapse
Affiliation(s)
- Vicente Arteaga
- Laboratorio de Microbiología Escuela de Ciencias Agrícolas y Ambientales (ECAA), Universidad Católica del Ecuador-Sede Ibarra (PUCESI), Spain
| | - Alexandre Lamas
- Laboratorio de Higiene Inspección y Control de Alimentos. Departamento de Química Analítica, Nutrición y Bromatología. Universidad de Santiago de Compostela, 27002, Lugo, Spain.
| | - Patricia Regal
- Laboratorio de Higiene Inspección y Control de Alimentos. Departamento de Química Analítica, Nutrición y Bromatología. Universidad de Santiago de Compostela, 27002, Lugo, Spain
| | - Beatriz Vázquez
- Laboratorio de Higiene Inspección y Control de Alimentos. Departamento de Química Analítica, Nutrición y Bromatología. Universidad de Santiago de Compostela, 27002, Lugo, Spain
| | - José Manuel Miranda
- Laboratorio de Higiene Inspección y Control de Alimentos. Departamento de Química Analítica, Nutrición y Bromatología. Universidad de Santiago de Compostela, 27002, Lugo, Spain
| | - Alberto Cepeda
- Laboratorio de Higiene Inspección y Control de Alimentos. Departamento de Química Analítica, Nutrición y Bromatología. Universidad de Santiago de Compostela, 27002, Lugo, Spain
| | - Carlos Manuel Franco
- Laboratorio de Higiene Inspección y Control de Alimentos. Departamento de Química Analítica, Nutrición y Bromatología. Universidad de Santiago de Compostela, 27002, Lugo, Spain
| |
Collapse
|
129
|
Chemical and Biological Characteristics of Antimicrobial α-Helical Peptides Found in Solitary Wasp Venoms and Their Interactions with Model Membranes. Toxins (Basel) 2019; 11:toxins11100559. [PMID: 31554187 PMCID: PMC6832458 DOI: 10.3390/toxins11100559] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/04/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023] Open
Abstract
Solitary wasps use their stinging venoms for paralyzing insect or spider prey and feeding them to their larvae. We have surveyed bioactive substances in solitary wasp venoms, and found antimicrobial peptides together with some other bioactive peptides. Eumenine mastoparan-AF (EMP-AF) was the first to be found from the venom of the solitary eumenine wasp Anterhynchium flavomarginatum micado, showing antimicrobial, histamine-releasing, and hemolytic activities, and adopting an α-helical secondary structure under appropriate conditions. Further survey of solitary wasp venom components revealed that eumenine wasp venoms contained such antimicrobial α-helical peptides as the major peptide component. This review summarizes the results obtained from the studies of these peptides in solitary wasp venoms and some analogs from the viewpoint of (1) chemical and biological characterization; (2) physicochemical properties and secondary structure; and (3) channel-like pore-forming properties.
Collapse
|
130
|
Sruthy KS, Nair A, Antony SP, Puthumana J, Singh ISB, Philip R. A histone H2A derived antimicrobial peptide, Fi-Histin from the Indian White shrimp, Fenneropenaeus indicus: Molecular and functional characterization. FISH & SHELLFISH IMMUNOLOGY 2019; 92:667-679. [PMID: 31252047 DOI: 10.1016/j.fsi.2019.06.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/22/2019] [Accepted: 06/23/2019] [Indexed: 06/09/2023]
Abstract
Antimicrobial peptides (AMPs) derived from histone proteins form an important category of peptide antibiotics. Present study deals with the molecular and functional characterization of a 27-amino acid histone H2A derived AMP from the Indian White shrimp, Fenneropenaeus indicus designated as Fi-Histin. This peptide displayed distinctive features of AMPs such as amphiphilic alpha helical structure and a net charge of +6. The synthetic peptide exhibited significant antimicrobial activity against Gram-negative and Gram-positive bacteria especially against V. vulnificus, P. aeruginosa, V. parahaemolyticus, V. cholera and S. aureus. Disruption of cell membrane and cell content leakage were observed in peptide treated V. vulnificus using scanning electron microscopy. The synthetic peptide Fi-His1-21 exhibited DNA binding activity and found to be non-haemolytic at the tested concentrations. Peptide was also found to possess anticancer activity against NCI-H460 and HEp-2 cell lines with an IC50 of 22.670 ± 13.939 μM and 31.274 ± 24.531 μM respectively. This is the first report of a histone H2A derived peptide from F. indicus with a specific antimicrobial activity and anticancer activity, which could be a new candidate for future applications in aquaculture and medicine.
Collapse
Affiliation(s)
- K S Sruthy
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India
| | - Aishwarya Nair
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India
| | - Swapna P Antony
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India
| | - Jayesh Puthumana
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, 682016, Kerala, India
| | - I S Bright Singh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, 682016, Kerala, India
| | - Rosamma Philip
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, 682016, Kerala, India.
| |
Collapse
|
131
|
Small-Molecule Morphogenesis Modulators Enhance the Ability of 14-Helical β-Peptides To Prevent Candida albicans Biofilm Formation. Antimicrob Agents Chemother 2019; 63:AAC.02653-18. [PMID: 31209011 DOI: 10.1128/aac.02653-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/03/2019] [Indexed: 02/03/2023] Open
Abstract
Candida albicans is an opportunistic fungal pathogen responsible for mucosal candidiasis and systemic candidemia in humans. Often, these infections are associated with the formation of drug-resistant biofilms on the surfaces of tissues or medical devices. Increased incidence of C. albicans resistance to current antifungals has heightened the need for new strategies to prevent or eliminate biofilm-related fungal infections. In prior studies, we designed 14-helical β-peptides to mimic the structural properties of natural antimicrobial α-peptides (AMPs) in an effort to develop active and selective antifungal compounds. These amphiphilic, cationic, helical β-peptides exhibited antifungal activity against planktonic C. albicans cells and inhibited biofilm formation in vitro and in vivo Recent studies have suggested the use of antivirulence agents in combination with antifungals. In this study, we investigated the use of compounds that target C. albicans polymorphism, such as 1-dodecanol, isoamyl alcohol, and farnesol, to attempt to improve β-peptide efficacy for preventing C. albicans biofilms. Isoamyl alcohol, which prevents hyphal formation, reduced the minimum biofilm prevention concentrations (MBPCs) of β-peptides by up to 128-fold. Combinations of isoamyl alcohol and antifungal β-peptides resulted in less than 10% hemolysis at the antifungal MBPCs. Overall, our results suggest potential benefits of combination therapies comprised of morphogenesis modulators and antifungal AMP peptidomimetics for preventing C. albicans biofilm formation.
Collapse
|
132
|
Soundrarajan N, Park S, Le Van Chanh Q, Cho HS, Raghunathan G, Ahn B, Song H, Kim JH, Park C. Protegrin-1 cytotoxicity towards mammalian cells positively correlates with the magnitude of conformational changes of the unfolded form upon cell interaction. Sci Rep 2019; 9:11569. [PMID: 31399625 PMCID: PMC6689069 DOI: 10.1038/s41598-019-47955-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/25/2019] [Indexed: 12/23/2022] Open
Abstract
Porcine protegrin-1 (PG-1) is a broad-spectrum antimicrobial peptide (AMP) with potent antimicrobial activities. We produced recombinant PG-1 and evaluated its cytotoxicity toward various types of mammalian cell lines, including embryonic fibroblasts, retinal cells, embryonic kidney cells, neuroblastoma cells, alveolar macrophage cells, and neutrophils. The sensitivity of the different mammalian cells to cytotoxic damage induced by PG-1 differed significantly among the cell types, with retinal neuron cells and neutrophils being the most significantly affected. A circular dichroism analysis showed there was a precise correlation between conformational changes in PG-1 and the magnitude of cytotoxicity among the various cell type. Subsequently, a green fluorescent protein (GFP) penetration assay using positively charged GFPs indicated there was a close correlation between the degree of penetration of charged GFP into cells and the magnitude of PG-1 cytotoxicity. Furthermore, we also showed that inhibition of the synthesis of anionic sulphated proteoglycans on the cell surface decreases the cytotoxic damage induced by PG-1 treatment. Taken together, the observed cytotoxicity of PG-1 towards different membrane surfaces is highly driven by the membrane’s anionic properties. Our results reveal a possible mechanism underlying cell-type dependent differences in cytotoxicity of AMPs, such as PG-1, toward mammalian cells.
Collapse
Affiliation(s)
| | - Suhyun Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, South Korea
| | - Quy Le Van Chanh
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, South Korea
| | - Hye-Sun Cho
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, South Korea
| | - Govindan Raghunathan
- Department of Chemistry, University of Konstanz, Universitӓtsstraße 10, 78457, Konstanz, Germany
| | - Byeongyong Ahn
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, South Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, South Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, South Korea
| | - Chankyu Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, South Korea.
| |
Collapse
|
133
|
Wang H, Zhao D, Nguyen LX, Wu H, Li L, Dong D, Troadec E, Zhu Y, Hoang DH, Stein AS, Al Malki M, Aldoss I, Lin A, Ghoda LY, McDonald T, Pichiorri F, Carlesso N, Kuo YH, Zhang B, Jin J, Marcucci G. Targeting cell membrane HDM2: A novel therapeutic approach for acute myeloid leukemia. Leukemia 2019; 34:75-86. [PMID: 31337857 DOI: 10.1038/s41375-019-0522-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/30/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022]
Abstract
The E3 ligase human double minute 2 (HDM2) regulates the activity of the tumor suppressor protein p53. A p53-independent HDM2 expression has been reported on the membrane of cancer cells but not on that of normal cells. Herein, we first showed that membrane HDM2 (mHDM2) is exclusively expressed on human and mouse AML blasts, including leukemia stem cell (LSC)-enriched subpopulations, but not on normal hematopoietic stem cells (HSCs). Higher mHDM2 levels in AML blasts were associated with leukemia-initiating capacity, quiescence, and chemoresistance. We also showed that a synthetic peptide PNC-27 binds to mHDM2 and enhances the interaction of mHDM2 and E-cadherin on the cell membrane; in turn, E-cadherin ubiquitination and degradation lead to membrane damage and cell death of AML blasts by necrobiosis. PNC-27 treatment in vivo resulted in a significant killing of both AML "bulk" blasts and LSCs, as demonstrated respectively in primary and secondary transplant experiments, using both human and murine AML models. Notably, PNC-27 spares normal HSC activity, as demonstrated in primary and secondary BM transplant experiments of wild-type mice. We concluded that mHDM2 represents a novel and unique therapeutic target, and targeting mHDM2 using PNC-27 selectively kills AML cells, including LSCs, with minimal off-target hematopoietic toxicity.
Collapse
Affiliation(s)
- Huafeng Wang
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA.,Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Dandan Zhao
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Le Xuan Nguyen
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA.,Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Herman Wu
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Ling Li
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Dan Dong
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA.,Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, PR China
| | - Estelle Troadec
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Yinghui Zhu
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Dinh Hoa Hoang
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Anthony S Stein
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Monzr Al Malki
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Ibrahim Aldoss
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Allen Lin
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Lucy Y Ghoda
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Tinisha McDonald
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Flavia Pichiorri
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Nadia Carlesso
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Ya-Huei Kuo
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Bin Zhang
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA.
| | - Jie Jin
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China. .,Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, PR China.
| | - Guido Marcucci
- Hematologic Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA, USA.
| |
Collapse
|
134
|
Rodríguez López ADL, Lee MR, Ortiz BJ, Gastfriend BD, Whitehead R, Lynn DM, Palecek SP. Preventing S. aureus biofilm formation on titanium surfaces by the release of antimicrobial β-peptides from polyelectrolyte multilayers. Acta Biomater 2019; 93:50-62. [PMID: 30831325 PMCID: PMC6693497 DOI: 10.1016/j.actbio.2019.02.047] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/28/2019] [Accepted: 02/27/2019] [Indexed: 02/08/2023]
Abstract
Staphylococcus aureus infections represent the major cause of titanium based-orthopaedic implant failure. Current treatments for S. aureus infections involve the systemic delivery of antibiotics and additional surgeries, increasing health-care costs and affecting patient's quality of life. As a step toward the development of new strategies that can prevent these infections, we build upon previous work demonstrating that the colonization of catheters by the fungal pathogen Candida albicans can be prevented by coating them with thin polymer multilayers composed of chitosan (CH) and hyaluronic acid (HA) designed to release a β-amino acid-based peptidomimetic of antimicrobial peptides (AMPs). We demonstrate here that this β-peptide is also potent against S. aureus (MBPC = 4 μg/mL) and characterize its selectivity toward S. aureus biofilms. We demonstrate further that β-peptide-containing CH/HA thin-films can be fabricated on the surfaces of rough planar titanium substrates in ways that allow mammalian cell attachment and permit the long-term release of β-peptide. β-Peptide loading on CH/HA thin-films was then adjusted to achieve release of β-peptide quantities that selectively prevent S. aureus biofilms on titanium substrates in vitro for up to 24 days and remained antimicrobial after being challenged sequentially five times with S. aureus inocula, while causing no significant MC3T3-E1 preosteoblast cytotoxicity compared to uncoated and film-coated controls lacking β-peptide. We conclude that these β-peptide-containing films offer a novel and promising localized delivery approach for preventing orthopaedic implant infections. The facile fabrication and loading of β-peptide-containing films reported here provides opportunities for coating other medical devices prone to biofilm-associated infections. STATEMENT OF SIGNIFICANCE: Titanium (Ti) and its alloys are used widely in orthopaedic devices due to their mechanical strength and long-term biocompatibility. However, these devices are susceptible to bacterial colonization and the subsequent formation of biofilms. Here we report a chitosan and hyaluronic acid polyelectrolyte multilayer-based approach for the localized delivery of helical, cationic, globally amphiphilic β-peptide mimetics of antimicrobial peptides to inhibit S. aureus colonization and biofilm formation. Our results reveal that controlled release of this β-peptide can selectively kill S. aureus cells without exhibiting toxicity toward MC3T3-E1 preosteoblast cells. Further development of this polymer-based coating could result in new strategies for preventing orthopaedic implant-related infections, improving outcomes of these titanium implants.
Collapse
Affiliation(s)
- Angélica de L Rodríguez López
- Department of Materials Science and Engineering, 1509 University Avenue, University of Wisconsin- Madison, Madison, WI 53706, USA
| | - Myung-Ryul Lee
- Department of Chemical and Biological Engineering, 1415 Engineering Drive, University of Wisconsin- Madison, Madison, WI 53706, USA
| | - Benjamín J Ortiz
- Department of Chemical and Biological Engineering, 1415 Engineering Drive, University of Wisconsin- Madison, Madison, WI 53706, USA
| | - Benjamin D Gastfriend
- Department of Chemical and Biological Engineering, 1415 Engineering Drive, University of Wisconsin- Madison, Madison, WI 53706, USA
| | - Riley Whitehead
- Department of Chemical and Biological Engineering, 1415 Engineering Drive, University of Wisconsin- Madison, Madison, WI 53706, USA
| | - David M Lynn
- Department of Materials Science and Engineering, 1509 University Avenue, University of Wisconsin- Madison, Madison, WI 53706, USA; Department of Chemical and Biological Engineering, 1415 Engineering Drive, University of Wisconsin- Madison, Madison, WI 53706, USA; Department of Chemistry, 1101 University Avenue, University of Wisconsin- Madison, Madison, WI 53706, USA.
| | - Sean P Palecek
- Department of Materials Science and Engineering, 1509 University Avenue, University of Wisconsin- Madison, Madison, WI 53706, USA; Department of Chemical and Biological Engineering, 1415 Engineering Drive, University of Wisconsin- Madison, Madison, WI 53706, USA.
| |
Collapse
|
135
|
Mihailescu M, Sorci M, Seckute J, Silin VI, Hammer J, Perrin BS, Hernandez JI, Smajic N, Shrestha A, Bogardus KA, Greenwood AI, Fu R, Blazyk J, Pastor RW, Nicholson LK, Belfort G, Cotten ML. Structure and Function in Antimicrobial Piscidins: Histidine Position, Directionality of Membrane Insertion, and pH-Dependent Permeabilization. J Am Chem Soc 2019; 141:9837-9853. [PMID: 31144503 DOI: 10.1021/jacs.9b00440] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Piscidins are histidine-enriched antimicrobial peptides that interact with lipid bilayers as amphipathic α-helices. Their activity at acidic and basic pH in vivo makes them promising templates for biomedical applications. This study focuses on p1 and p3, both 22-residue-long piscidins with 68% sequence identity. They share three histidines (H3, H4, and H11), but p1, which is significantly more permeabilizing, has a fourth histidine (H17). This study investigates how variations in amphipathic character associated with histidines affect the permeabilization properties of p1 and p3. First, we show that the permeabilization ability of p3, but not p1, is strongly inhibited at pH 6.0 when the conserved histidines are partially charged and H17 is predominantly neutral. Second, our neutron diffraction measurements performed at low water content and neutral pH indicate that the average conformation of p1 is highly tilted, with its C-terminus extending into the opposite leaflet. In contrast, p3 is surface bound with its N-terminal end tilted toward the bilayer interior. The deeper membrane insertion of p1 correlates with its behavior at full hydration: an enhanced ability to tilt, bury its histidines and C-terminus, induce membrane thinning and defects, and alter membrane conductance and viscoelastic properties. Furthermore, its pH-resiliency relates to the neutral state favored by H17. Overall, these results provide mechanistic insights into how differences in the histidine content and amphipathicity of peptides can elicit different directionality of membrane insertion and pH-dependent permeabilization. This work features complementary methods, including dye leakage assays, NMR-monitored titrations, X-ray and neutron diffraction, oriented CD, molecular dynamics, electrochemical impedance spectroscopy, surface plasmon resonance, and quartz crystal microbalance with dissipation.
Collapse
Affiliation(s)
- Mihaela Mihailescu
- Institute for Bioscience and Biotechnology Research , University of Maryland , Rockville , Maryland 20850 , United States
| | - Mirco Sorci
- Department of Chemical and Biological Engineering and Center for Biotechnology and Interdisciplinary Studies , Rensselaer Polytechnic Institute , Troy , New York 12180 , United States
| | - Jolita Seckute
- Department of Molecular Biology and Genetics , Cornell University , Ithaca , New York 14853 , United States
| | - Vitalii I Silin
- Institute for Bioscience and Biotechnology Research , University of Maryland , Rockville , Maryland 20850 , United States
| | - Janet Hammer
- Department of Biomedical Sciences , Ohio University , Athens , Ohio 45701 , United States
| | - B Scott Perrin
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Jorge I Hernandez
- Department of Bioengineering , Clemson University , Clemson , South Carolina 29634 , United States
| | - Nedzada Smajic
- Department of Chemistry , Hamilton College , Clinton , New York 13323 , United States
| | - Akritee Shrestha
- Department of Chemistry , Hamilton College , Clinton , New York 13323 , United States
| | - Kimberly A Bogardus
- Department of Chemistry , Hamilton College , Clinton , New York 13323 , United States
| | - Alexander I Greenwood
- Department of Applied Science , College of William and Mary , Williamsburg , Virginia 23185 , United States
| | - Riqiang Fu
- National High Magnetic Field Laboratory , Tallahassee , Florida 32310 , United States
| | - Jack Blazyk
- Department of Biomedical Sciences , Ohio University , Athens , Ohio 45701 , United States
| | - Richard W Pastor
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Linda K Nicholson
- Department of Molecular Biology and Genetics , Cornell University , Ithaca , New York 14853 , United States
| | - Georges Belfort
- Department of Chemical and Biological Engineering and Center for Biotechnology and Interdisciplinary Studies , Rensselaer Polytechnic Institute , Troy , New York 12180 , United States
| | - Myriam L Cotten
- Department of Applied Science , College of William and Mary , Williamsburg , Virginia 23185 , United States
| |
Collapse
|
136
|
De novo aggregation of Alzheimer's Aβ25-35 peptides in a lipid bilayer. Sci Rep 2019; 9:7161. [PMID: 31073226 PMCID: PMC6509337 DOI: 10.1038/s41598-019-43685-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/27/2019] [Indexed: 11/28/2022] Open
Abstract
A potential mechanism of cytotoxicity attributed to Alzheimer’s Aβ peptides postulates that their aggregation disrupts membrane structure causing uncontrollable permeation of Ca2+ ions. To gain molecular insights into these processes, we have performed all-atom explicit solvent replica exchange with solute tempering molecular dynamics simulations probing aggregation of the naturally occurring Aβ fragment Aβ25-35 within the DMPC lipid bilayer. To compare the impact produced on the bilayer by Aβ25-35 oligomers and monomers, we used as a control our previous simulations, which explored binding of Aβ25-35 monomers to the same bilayer. We found that compared to monomeric species aggregation results in much deeper insertion of Aβ25-35 peptides into the bilayer hydrophobic core causing more pronounced disruption in its structure. Aβ25-35 peptides aggregate by incorporating monomer-like structures with stable C-terminal helix. As a result the Aβ25-35 dimer features unusual helix head-to-tail topology supported by a parallel off-registry interface. Such topology affords further growth of an aggregate by recruiting additional peptides. Free energy landscape reveals that inserted dimers represent the dominant equilibrium state augmented by two metastable states associated with surface bound dimers and inserted monomers. Using the free energy landscape we propose the pathway of Aβ25-35 binding, aggregation, and insertion into the lipid bilayer.
Collapse
|
137
|
Huang T, Gu W, Wang B, Zhang Y, Cui L, Yao Z, Zhao C, Xu G. Identification and expression of the hepcidin gene from brown trout (Salmo trutta) and functional analysis of its synthetic peptide. FISH & SHELLFISH IMMUNOLOGY 2019; 87:243-253. [PMID: 30648626 DOI: 10.1016/j.fsi.2019.01.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 06/09/2023]
Abstract
Hepcidin, a hepatic antimicrobial peptide, is a key player of the nonspecific immune system. The structure of hepcidin gene from brown trout (Bthepc) has been characterized at the molecular level. The 1158-bp mRNA generates a coding sequence (CDS) of 267 bp, which encodes an 88-amino acid protein. Molecular evolution analysis classified Bthepc to the family Salmonidae. Amino acid sequence homologies between Bthepc and hepcidin in other species such as Oncorhynchus mykiss, Salmo salar, and Hucho taimen were found to be 93.18%, 96.59%, and 92.05% respectively. The mature peptide and the signal peptide of Bthepc are made of 25 and 24 amino acids, respectively. Similar to the other species, eight conserved cysteines in the mature peptide of Bthepc are held together by four disulphide bonds. Expression profiling of Bthepc indicated its highest expression in the liver. Further, iron levels or inflammation did not induce the age-dependent expression of Bthepc. Bthepc mRNA expression analysis in six immune tissues (liver, gill, spleen, skin, head kidney and intestine) indicated different levels of increase when challenged with Aeromonas salmonicida and Aeromonas hydrophila. The antimicrobial activity of synthetic Bthepc to typical pathogens was verified in vitro. In addition, Bthepc showed moderate haemolytic activity to mammalian erythrocytes. The antimicrobial activity of Bthepc was attributed to the disruption of the bacterial outer membrane integrity, which was evident from our scanning electron microscopy results. In summary, hepcidin gene of brown trout was characterized, and its antimicrobial activity was verified on different levels.
Collapse
Affiliation(s)
- Tianqing Huang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Wei Gu
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Bingqian Wang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Yuyong Zhang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Lili Cui
- Yunnan Academy of Fishery Sciences, Kunming, PR China; College of Animal Science and Technology, Yunnan Agricultural University, Kunming, PR China
| | - Zuochun Yao
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Cheng Zhao
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Gefeng Xu
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China.
| |
Collapse
|
138
|
Yang Y, Liu Z, He X, Yang J, Wu J, Yang H, Li M, Qian Q, Lai R, Xu W, Wei L. A small mycobacteriophage-derived peptide and its improved isomer restrict mycobacterial infection via dual mycobactericidal-immunoregulatory activities. J Biol Chem 2019; 294:7615-7631. [PMID: 30894414 DOI: 10.1074/jbc.ra118.006968] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/07/2019] [Indexed: 12/14/2022] Open
Abstract
Mycobacteriophages express various peptides/proteins to infect Mycobacterium tuberculosis (M. tb). Particular attention has been paid to mycobacteriophage-derived endolysin proteins. We herein characterized a small mycobacteriophage-derived peptide designated AK15 with potent anti-M. tb activity. AK15 adopted cationic amphiphilic α-helical structure, and on the basis of this structure, we designed six isomers with increased hydrophobic moment by rearranging amino acid residues of the helix. We found that one of these isomers, AK15-6, exhibits enhanced anti-mycobacterial efficiency. Both AK15 and AK15-6 directly inhibited M. tb by trehalose 6,6'-dimycolate (TDM) binding and membrane disruption. They both exhibited bactericidal activity, cell selectivity, and synergistic effects with rifampicin, and neither induced drug resistance to M. tb They efficiently attenuated mycobacterial load in the lungs of M. tb-infected mice. We observed that lysine, arginine, tryptophan, and an α-helix are key structural requirements for their direct anti-mycobacterial action. Of note, they also exhibited immunomodulatory effects, including inhibition of proinflammatory response in TDM-stimulated or M. tb-infected murine bone marrow-derived macrophages (BMDMs) and M.tb-infected mice and induction of only a modest level of cytokine (tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6)) production in murine BMDMs and a T-cell cytokine (interferin-γ (IFN-γ) and TNF-α) response in murine lung and spleen. In summary, characterization of a small mycobacteriophage-derived peptide and its improved isomer revealed that both efficiently restrain M. tb infection via dual mycobactericidal-immunoregulatory activities. Our work provides clues for identifying small mycobacteriophage-derived anti-mycobacterial peptides and improving those that have cationic amphiphilic α-helices.
Collapse
Affiliation(s)
- Yang Yang
- From the Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu
| | - Zhen Liu
- From the Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu
| | - Xiaoqin He
- the Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan.,the National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, Jiangsu Province
| | - Juanjuan Yang
- the Institute of Pharmaceutical Biotechnology and Engineering, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, and
| | - Jing Wu
- the School of Basic Medical Sciences, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Hailong Yang
- the School of Basic Medical Sciences, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Min Li
- From the Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu
| | - Qian Qian
- From the Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu
| | - Ren Lai
- the Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan,
| | - Wei Xu
- From the Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu,
| | - Lin Wei
- From the Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu,
| |
Collapse
|
139
|
Scheenstra MR, van den Belt M, Tjeerdsma-van Bokhoven JLM, Schneider VAF, Ordonez SR, van Dijk A, Veldhuizen EJA, Haagsman HP. Cathelicidins PMAP-36, LL-37 and CATH-2 are similar peptides with different modes of action. Sci Rep 2019; 9:4780. [PMID: 30886247 PMCID: PMC6423055 DOI: 10.1038/s41598-019-41246-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 02/25/2019] [Indexed: 12/30/2022] Open
Abstract
Host defense peptides (HDPs) play a pivotal role in innate immunity and have, in addition to antimicrobial activity, also important immunomodulatory functions. Bacteria are less likely to develop resistance against HDPs because these peptides target and kill bacteria in multiple ways, as well as modulate the immune system. Therefore, HDPs, and derivatives thereof, are promising alternatives to traditional antibiotics. Hardly anything is known about the immunomodulatory functions of porcine cathelicidin PMAP-36. In this study, we aimed to determine both antibacterial and immunomodulatory activities of PMAP-36 comparing the properties of PMAP-36 analogs with two well-studied peptides, human LL-37 and chicken CATH-2. Transmission electron microscopy revealed different killing mechanisms of E. coli for PMAP-36, CATH-2 and LL-37. LL-37 binds LPS very weakly in contrast to PMAP-36, but it inhibits LPS activation of macrophages the strongest. The first 11 amino acids of the N-terminal side of PMAP-36 are dispensable for E. coli killing, LPS-neutralization and binding. Deletion of four additional amino acids resulted in a strong decrease in activity. The activity of full length PMAP-36 was not affected by monomerization, whereas the shorter analogs require dimerization for proper immunomodulatory activity but not for their antibacterial activity.
Collapse
Affiliation(s)
- Maaike R Scheenstra
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Matthias van den Belt
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Johanna L M Tjeerdsma-van Bokhoven
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Viktoria A F Schneider
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Soledad R Ordonez
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Albert van Dijk
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Edwin J A Veldhuizen
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Henk P Haagsman
- Department of Infectious Diseases and Immunology, Division of Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
140
|
Unifying structural signature of eukaryotic α-helical host defense peptides. Proc Natl Acad Sci U S A 2019; 116:6944-6953. [PMID: 30877253 DOI: 10.1073/pnas.1819250116] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Diversity of α-helical host defense peptides (αHDPs) contributes to immunity against a broad spectrum of pathogens via multiple functions. Thus, resolving common structure-function relationships among αHDPs is inherently difficult, even for artificial-intelligence-based methods that seek multifactorial trends rather than foundational principles. Here, bioinformatic and pattern recognition methods were applied to identify a unifying signature of eukaryotic αHDPs derived from amino acid sequence, biochemical, and three-dimensional properties of known αHDPs. The signature formula contains a helical domain of 12 residues with a mean hydrophobic moment of 0.50 and favoring aliphatic over aromatic hydrophobes in 18-aa windows of peptides or proteins matching its semantic definition. The holistic α-core signature subsumes existing physicochemical properties of αHDPs, and converged strongly with predictions of an independent machine-learning-based classifier recognizing sequences inducing negative Gaussian curvature in target membranes. Queries using the α-core formula identified 93% of all annotated αHDPs in proteomic databases and retrieved all major αHDP families. Synthesis and antimicrobial assays confirmed efficacies of predicted sequences having no previously known antimicrobial activity. The unifying α-core signature establishes a foundational framework for discovering and understanding αHDPs encompassing diverse structural and mechanistic variations, and affords possibilities for deterministic design of antiinfectives.
Collapse
|
141
|
Manzo G, Ferguson PM, Gustilo VB, Hind CK, Clifford M, Bui TT, Drake AF, Atkinson RA, Sutton JM, Batoni G, Lorenz CD, Phoenix DA, Mason AJ. Minor sequence modifications in temporin B cause drastic changes in antibacterial potency and selectivity by fundamentally altering membrane activity. Sci Rep 2019; 9:1385. [PMID: 30718667 PMCID: PMC6362004 DOI: 10.1038/s41598-018-37630-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/10/2018] [Indexed: 11/08/2022] Open
Abstract
Antimicrobial peptides (AMPs) are a potential source of new molecules to counter the increase in antimicrobial resistant infections but a better understanding of their properties is required to understand their native function and for effective translation as therapeutics. Details of the mechanism of their interaction with the bacterial plasma membrane are desired since damage or penetration of this structure is considered essential for AMPs activity. Relatively modest modifications to AMPs primary sequence can induce substantial changes in potency and/or spectrum of activity but, hitherto, have not been predicted to substantially alter the mechanism of interaction with the bacterial plasma membrane. Here we use a combination of molecular dynamics simulations, circular dichroism, solid-state NMR and patch clamp to investigate the extent to which temporin B and its analogues can be distinguished both in vitro and in silico on the basis of their interactions with model membranes. Enhancing the hydrophobicity of the N-terminus and cationicity of the C-terminus in temporin B improves its membrane activity and potency against both Gram-negative and Gram-positive bacteria. In contrast, enhancing the cationicity of the N-terminus abrogates its ability to trigger channel conductance and renders it ineffective against Gram-positive bacteria while nevertheless enhancing its potency against Escherichia coli. Our findings suggest even closely related AMPs may target the same bacterium with fundamentally differing mechanisms of action.
Collapse
Affiliation(s)
- Giorgia Manzo
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - Philip M Ferguson
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - V Benjamin Gustilo
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - Charlotte K Hind
- Technology Development Group, National Infection Service, Public Health England, Salisbury, UK
| | - Melanie Clifford
- Technology Development Group, National Infection Service, Public Health England, Salisbury, UK
| | - Tam T Bui
- Centre for Biomolecular Spectroscopy and Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, London, SE1 1UL, United Kingdom
| | - Alex F Drake
- Centre for Biomolecular Spectroscopy and Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, London, SE1 1UL, United Kingdom
| | - R Andrew Atkinson
- Centre for Biomolecular Spectroscopy and Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, London, SE1 1UL, United Kingdom
| | - J Mark Sutton
- Technology Development Group, National Infection Service, Public Health England, Salisbury, UK
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Christian D Lorenz
- Department of Physics, King's College London, London, WC2R 2LS, United Kingdom
| | - David A Phoenix
- School of Applied Science, London South Bank University, 103 Borough Road, London, SE1 0AA, United Kingdom
| | - A James Mason
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom.
| |
Collapse
|
142
|
Irazazabal LN, Porto WF, Fensterseifer IC, Alves ES, Matos CO, Menezes AC, Felício MR, Gonçalves S, Santos NC, Ribeiro SM, Humblot V, Lião LM, Ladram A, Franco OL. Fast and potent bactericidal membrane lytic activity of PaDBS1R1, a novel cationic antimicrobial peptide. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:178-190. [DOI: 10.1016/j.bbamem.2018.08.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 12/20/2022]
|
143
|
Selectivity of Antimicrobial Peptides: A Complex Interplay of Multiple Equilibria. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1117:175-214. [DOI: 10.1007/978-981-13-3588-4_11] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
144
|
Xu G, Huang T, Gu W, Zhang Y, Yao Z, Zhao C, Wang B. Characterization, expression, and functional analysis of the hepcidin gene from Brachymystax lenok. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 89:131-140. [PMID: 30125584 DOI: 10.1016/j.dci.2018.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/16/2018] [Accepted: 08/16/2018] [Indexed: 06/08/2023]
Abstract
Hepcidin, a cysteine-rich antimicrobial peptide, is an important effector molecule in the innate immune system. Recently, Brachymystax lenok has become to be a valuable cold-water fish in China, particularly as the wild resources are rapidly declining. In this study, the hepcidin gene of Brachymystax lenok (Blhepc) has been cloned. The 870-bp mRNA contains a coding sequence (CDS) of 267 bp that encodes 88 amino acid residues. Amino acid sequence identities of Blhepc with hepcidin in Oncorhynchus mykiss, Salmo salar, and Hucho taimen were found to be 93.18%, 89.77% and 93.18%, respectively. Phylogenetic analysis indicated that Blhepc was clustered in the family Salmonidae. The putative signal peptide and the mature peptide contained 24 and 25 amino acid residues, respectively. The RXXR motif for recruitment of propeptide convertase was identified upstream of the mature peptide of Blhepc by sequence analysis. The N-terminal amino acid residues of the mature Blhepc peptide were Q-SH-L, a structure involved in regulating iron metabolism. Eight conserved cysteine residues in the mature peptide were held together by four disulfide bonds. Expression profiling of Blhepc indicated its highest level in the liver; its expression was stronger in males than in similar-aged females. Moreover, its expression in the liver increased significantly with age. Expression of Blhepc in six immune tissues showed increase in various degrees when challenged with Aeromonas salmonicida and Aeromonas hydrophila. A synthetic Blhepc mature peptide was validated to have significant antimicrobial activity against gram-negative and gram-positive bacteria and fungi in vitro. These results show that Blhepc may be an important component in the innate immunity of Brachymystax lenok, which could provide antimicrobial activities against invading pathogens.
Collapse
Affiliation(s)
- Gefeng Xu
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Tianqing Huang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Wei Gu
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Yuyong Zhang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Zuochun Yao
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Cheng Zhao
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China
| | - Bingqian Wang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, PR China.
| |
Collapse
|
145
|
Bioactive properties of Kilka (Clupeonella cultriventris caspi) fish protein hydrolysates. JOURNAL OF FOOD MEASUREMENT AND CHARACTERIZATION 2018. [DOI: 10.1007/s11694-018-9843-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
146
|
Marine Antimicrobial Peptides: A Promising Source of New Generation Antibiotics and Other Bio-active Molecules. Int J Pept Res Ther 2018. [DOI: 10.1007/s10989-018-9789-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
147
|
Siano A, Humpola MV, de Oliveira E, Albericio F, Simonetta AC, Lajmanovich R, Tonarelli GG. Leptodactylus latrans Amphibian Skin Secretions as a Novel Source for the Isolation of Antibacterial Peptides. Molecules 2018; 23:molecules23112943. [PMID: 30423858 PMCID: PMC6278411 DOI: 10.3390/molecules23112943] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 11/06/2018] [Accepted: 11/09/2018] [Indexed: 12/19/2022] Open
Abstract
Amphibians´ skin produces a diverse array of antimicrobial peptides that play a crucial role as the first line of defense against microbial invasion. Despite the immense richness of wild amphibians in Argentina, current knowledge about the presence of peptides with antimicrobial properties is limited to a only few species. Here we used LC-MS-MS to identify antimicrobial peptides with masses ranging from 1000 to 4000 Da from samples of skin secretions of Leptodactylus latrans (Anura: Leptodactylidae). Three novel amino acid sequences were selected for chemical synthesis and further studies. The three synthetic peptides, named P1-Ll-1577, P2-Ll-1298, and P3-Ll-2085, inhibited the growth of two ATCC strains, namely Escherichia coli and Staphylococcus aureus. P3-Ll-2085 was the most active peptide. In the presence of trifluoroethanol (TFE) and anionic liposomes, it adopted an amphipathic α-helical structure. P2-Ll-1298 showed slightly lower activity than P3-Ll-2085. Comparison of the MIC values of these two peptides revealed that the addition of seven amino acid residues (GLLDFLK) on the N-terminal of P2-Ll-1298 significantly improved activity against both strains. P1-Ll-1577, which remarkably is an anionic peptide, showed interesting antimicrobial activity against E. coli and S. aureus strain, showing marked membrane selectivity and non-hemolysis. Due to this, P1-L1-1577 emerges as a potential candidate for the development of new antibacterial drugs.
Collapse
Affiliation(s)
- Alvaro Siano
- Departamento de Química Orgánica, Facultad de Bioquímica y Cs. Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Ciudad Universitaria, 3000 Santa Fe, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1825 Buenos Aires, Argentina.
| | - Maria Veronica Humpola
- Departamento de Química Orgánica, Facultad de Bioquímica y Cs. Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Ciudad Universitaria, 3000 Santa Fe, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1825 Buenos Aires, Argentina.
| | - Eliandre de Oliveira
- Proteomics Platform, Barcelona Science Park, Baldiri Reixac 10, 08028 Barcelona, Spain.
| | - Fernando Albericio
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Barcelona Science Park, Baldiri Reixac 10, 08028 Barcelona, Spain;.
- Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain.
- School of Chemistry and Physics, University of KwaZulu-Natal, 4000 Durban, South Africa.
| | - Arturo C Simonetta
- Cátedras de Microbiología y Biotecnología, Departamento de Ingeniería en Alimentos, Facultad de Ingeniería Química, U.N.L. Santiago del Estero 2829, 3000 Santa Fe, Argentina.
| | - Rafael Lajmanovich
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1825 Buenos Aires, Argentina.
- Cátedra de Ecotoxicología, Escuela Superior de Sanidad. FBCB, U.N.L. Ciudad Universitaria, 3000 Santa Fe, Argentina.
| | - Georgina G Tonarelli
- Departamento de Química Orgánica, Facultad de Bioquímica y Cs. Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Ciudad Universitaria, 3000 Santa Fe, Argentina.
| |
Collapse
|
148
|
A structural perspective of plant antimicrobial peptides. Biochem J 2018; 475:3359-3375. [PMID: 30413680 DOI: 10.1042/bcj20180213] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 10/12/2018] [Accepted: 10/14/2018] [Indexed: 12/26/2022]
Abstract
Among the numerous strategies plants have developed to fend off enemy attack, antimicrobial peptides (AMPs) stand out as one of the most prominent defensive barriers that grant direct and durable resistance against a wide range of pests and pathogens. These small proteins are characterized by a compact structure and an overall positive charge. AMPs have an ancient origin and widespread occurrence in the plant kingdom but show an unusually high degree of variation in their amino acid sequences. Interestingly, there is a strikingly conserved topology among the plant AMP families, suggesting that the defensive properties of these peptides are not determined by their primary sequences but rather by their tridimensional structure. To explore and expand this idea, we here discuss the role of AMPs for plant defense from a structural perspective. We show how specific structural properties, such as length, charge, hydrophobicity, polar angle and conformation, are essential for plant AMPs to act as a chemical shield that hinders enemy attack. Knowledge on the topology of these peptides is facilitating the isolation, classification and even structural redesign of AMPs, thus allowing scientists to develop new peptides with multiple agronomical and pharmacological potential.
Collapse
|
149
|
Santos RS, Figueiredo C, Azevedo NF, Braeckmans K, De Smedt SC. Nanomaterials and molecular transporters to overcome the bacterial envelope barrier: Towards advanced delivery of antibiotics. Adv Drug Deliv Rev 2018; 136-137:28-48. [PMID: 29248479 DOI: 10.1016/j.addr.2017.12.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/10/2017] [Accepted: 12/12/2017] [Indexed: 01/13/2023]
Abstract
With the dramatic consequences of bacterial resistance to antibiotics, nanomaterials and molecular transporters have started to be investigated as alternative antibacterials or anti-infective carrier systems to improve the internalization of bactericidal drugs. However, the capability of nanomaterials/molecular transporters to overcome the bacterial cell envelope is poorly understood. It is critical to consider the sophisticated architecture of bacterial envelopes and reflect how nanomaterials/molecular transporters can interact with these envelopes, being the major aim of this review. The first part of this manuscript overviews the permeability of bacterial envelopes and how it limits the internalization of common antibiotic and novel oligonucleotide drugs. Subsequently we critically discuss the mechanisms that allow nanomaterials/molecular transporters to overcome the bacterial envelopes, focusing on the most promising ones to this end - siderophores, cyclodextrins, metal nanoparticles, antimicrobial/cell-penetrating peptides and fusogenic liposomes. This review may stimulate drug delivery and microbiology scientists in designing effective nanomaterials/molecular transporters against bacterial infections.
Collapse
|
150
|
Crusca E, Basso LGM, Altei WF, Marchetto R. Biophysical characterization and antitumor activity of synthetic Pantinin peptides from scorpion's venom. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:2155-2165. [DOI: 10.1016/j.bbamem.2018.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/24/2018] [Accepted: 08/19/2018] [Indexed: 01/30/2023]
|