1
|
Ukleja M, Kricks L, Torrens G, Peschiera I, Rodrigues-Lopes I, Krupka M, García-Fernández J, Melero R, Del Campo R, Eulalio A, Mateus A, López-Bravo M, Rico AI, Cava F, Lopez D. Flotillin-mediated stabilization of unfolded proteins in bacterial membrane microdomains. Nat Commun 2024; 15:5583. [PMID: 38961085 PMCID: PMC11222466 DOI: 10.1038/s41467-024-49951-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024] Open
Abstract
The function of many bacterial processes depends on the formation of functional membrane microdomains (FMMs), which resemble the lipid rafts of eukaryotic cells. However, the mechanism and the biological function of these membrane microdomains remain unclear. Here, we show that FMMs in the pathogen methicillin-resistant Staphylococcus aureus (MRSA) are dedicated to confining and stabilizing proteins unfolded due to cellular stress. The FMM scaffold protein flotillin forms a clamp-shaped oligomer that holds unfolded proteins, stabilizing them and favoring their correct folding. This process does not impose a direct energy cost on the cell and is crucial to survival of ATP-depleted bacteria, and thus to pathogenesis. Consequently, FMM disassembling causes the accumulation of unfolded proteins, which compromise MRSA viability during infection and cause penicillin re-sensitization due to PBP2a unfolding. Thus, our results indicate that FMMs mediate ATP-independent stabilization of unfolded proteins, which is essential for bacterial viability during infection.
Collapse
Affiliation(s)
- Marta Ukleja
- Department of Microbiology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, 28049, Spain
| | - Lara Kricks
- Department of Microbiology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, 28049, Spain
| | - Gabriel Torrens
- Department of Molecular Biology, Umeå University, Umeå, SE-901 87, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS). Umeå Center for Microbial Research (UCMR). Science for Life Laboratory (SciLifeLab), Umeå, SE-901 87, Sweden
| | - Ilaria Peschiera
- Department of Microbiology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, 28049, Spain
| | - Ines Rodrigues-Lopes
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504, Coimbra, Portugal
| | - Marcin Krupka
- Department of Microbiology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, 28049, Spain
| | - Julia García-Fernández
- Department of Microbiology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, 28049, Spain
| | - Roberto Melero
- Department of Structural Biology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, 28049, Spain
| | - Rosa Del Campo
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ramón y Cajal Hospital, 28034, Madrid, Spain
| | - Ana Eulalio
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504, Coimbra, Portugal
- Department of Life Sciences, Center for Bacterial Resistance Biology, Imperial College London, London, SW7 2AZ, United Kingdom
| | - André Mateus
- The Laboratory for Molecular Infection Medicine Sweden (MIMS). Umeå Center for Microbial Research (UCMR). Science for Life Laboratory (SciLifeLab), Umeå, SE-901 87, Sweden
- Department of Chemistry, Umeå University, Umeå, SE-901 87, Sweden
| | - María López-Bravo
- Department of Microbiology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, 28049, Spain
| | - Ana I Rico
- Department of Microbiology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, 28049, Spain
| | - Felipe Cava
- Department of Molecular Biology, Umeå University, Umeå, SE-901 87, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS). Umeå Center for Microbial Research (UCMR). Science for Life Laboratory (SciLifeLab), Umeå, SE-901 87, Sweden
| | - Daniel Lopez
- Department of Microbiology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, 28049, Spain.
| |
Collapse
|
2
|
Teixeira RG, Mészáros JP, Matos B, Côrte-Real L, Xavier CPR, Fontrodona X, Garcia MH, Romero I, Spengler G, Vasconcelos MH, Tomaz AI, Enyedy ÉA, Valente A. Novel family of [RuCp(N,N)(P)] + compounds with simultaneous anticancer and antibacterial activity: Biological evaluation and solution chemistry studies. Eur J Med Chem 2023; 262:115922. [PMID: 37944388 DOI: 10.1016/j.ejmech.2023.115922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023]
Abstract
A family of ten novel ruthenium(II)-cyclopentadienyl organometallics of general formula [Ru(η5-C5H5)(N,N)(PPh2(C6H4COOR)][CF3SO3] (1-10) in which (N,N) = 4,4'-R'-2,2'-bipyridyl (R = -H or -CH2CH2OH; R' = -H, -CH3, -OCH3, -CH2OH, and -CH2-biotin) was prepared from [Ru(η5-C5H5)(PPh2(C6H4COOH))2Cl]. All compounds were fully characterized by means of several spectroscopic and analytical techniques, and the molecular structures of [Ru(η5-C5H5)(PPh2(C6H4COOH))2Cl], 1, 3 and 4 have been additionally studied by single-crystal X-ray diffraction. The anticancer activity of all compounds was evaluated in sensitive and multidrug-resistant counterpart cell lines from human colorectal cancer (Colo 205 and Colo 320) and non-small cell lung cancer NSCLC (A549, NCI-H460 versus NCI-H460/R) as well. Notably, compounds 6 and 7 (R CH2CH2OH and (N,N) = bipy or Me2bipy, respectively) showed antiproliferative effect against both cell lines with high intrinsic selectivity towards cancer cells. The antibacterial activity of all compounds was also evaluated against both Gram negative and Gram positive strains, and some compounds in the series showed potent antibacterial activity against Staphylococcus aureus strains, including the methicillin-resistant MRSA strains. Solution speciation studies revealed that the complexes bearing the PPh2(C6H4COO-) ligand are neutral at physiological pH (7.4) in contrast with their ethylene glycol derivatives that have a permanent positive charge. While all compounds are lipophilic, the difference in the distribution coefficient for neutral and charged complexes is around one order of magnitude. Complexes 6 and 7 exhibited excellent biological activity and were selected for further studies. Spectrofluorometric methods were used to investigate their interaction with biomolecules such as human serum albumin (HSA) and calf thymus DNA (ct-DNA). For these complexes, binding site II of HSA is a possible binding pocket through non-covalent interactions. The release of ethidium from the DNA adduct by the charged complexes proves their interaction with DNA in contrast to the neutral ones. In conclusion, Ru(II)-cyclopentadienyl complexes with 2,2'-bipyridyl-derivatives and an ethylene glycol moiety tethered to the phenylphosphane co-ligand are very promising from a therapeutic perspective, in particular complexes 6 and 7 that display remarkable antibacterial activity with a high anti-proliferative effect against colon and non-small cell lung cancers, both clinically challenging neoplasias in need of effective solutions.
Collapse
Affiliation(s)
- Ricardo G Teixeira
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - János P Mészáros
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary; MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary
| | - Beatriz Matos
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, 4200-135, Porto, Portugal
| | - Leonor Côrte-Real
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Cristina P R Xavier
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, 4200-135, Porto, Portugal
| | - Xavier Fontrodona
- Departament de Química and Serveis Tècnics de Recerca, Universitat de Girona, C/ M. Aurèlia Campmany, 69, E-17003, Girona, Spain
| | - M Helena Garcia
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Isabel Romero
- Departament de Química and Serveis Tècnics de Recerca, Universitat de Girona, C/ M. Aurèlia Campmany, 69, E-17003, Girona, Spain
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary; Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, 6725, Szeged, Hungary
| | - M Helena Vasconcelos
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, 4200-135, Porto, Portugal; Department of Biological Sciences, Faculty of Pharmacy of the University of Porto (FFUP), Porto, Portugal
| | - Ana Isabel Tomaz
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Éva A Enyedy
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary; MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary
| | - Andreia Valente
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal.
| |
Collapse
|
3
|
Three-dimensional chiral morphodynamics of chemomechanical active shells. Proc Natl Acad Sci U S A 2022; 119:e2206159119. [PMID: 36442097 PMCID: PMC9894169 DOI: 10.1073/pnas.2206159119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Morphogenesis of active shells such as cells is a fundamental chemomechanical process that often exhibits three-dimensional (3D) large deformations and chemical pattern dynamics simultaneously. Here, we establish a chemomechanical active shell theory accounting for mechanical feedback and biochemical regulation to investigate the symmetry-breaking and 3D chiral morphodynamics emerging in the cell cortex. The active bending and stretching of the elastic shells are regulated by biochemical signals like actomyosin and RhoA, which, in turn, exert mechanical feedback on the biochemical events via deformation-dependent diffusion and inhibition. We show that active deformations can trigger chemomechanical bifurcations, yielding pulse spiral waves and global oscillations, which, with increasing mechanical feedback, give way to traveling or standing waves subsequently. Mechanical feedback is also found to contribute to stabilizing the polarity of emerging patterns, thus ensuring robust morphogenesis. Our results reproduce and unravel the experimentally observed solitary and multiple spiral patterns, which initiate asymmetric cleavage in Xenopus and starfish embryogenesis. This study underscores the crucial roles of mechanical feedback in cell development and also suggests a chemomechanical framework allowing for 3D large deformation and chemical signaling to explore complex morphogenesis in living shell-like structures.
Collapse
|
4
|
Schultz JR, Costa SK, Jachak GR, Hegde P, Zimmerman M, Pan Y, Josten M, Ejeh C, Hammerstad T, Sahl HG, Pereira PM, Pinho MG, Dartois V, Cheung A, Aldrich CC. Identification of 5-(Aryl/Heteroaryl)amino-4-quinolones as Potent Membrane-Disrupting Agents to Combat Antibiotic-Resistant Gram-Positive Bacteria. J Med Chem 2022; 65:13910-13934. [PMID: 36219779 PMCID: PMC9826610 DOI: 10.1021/acs.jmedchem.2c01151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Nosocomial infections caused by resistant Gram-positive organisms are on the rise, presumably due to a combination of factors including prolonged hospital exposure, increased use of invasive procedures, and pervasive antibiotic therapy. Although antibiotic stewardship and infection control measures are helpful, newer agents against multidrug-resistant (MDR) Gram-positive bacteria are urgently needed. Here, we describe our efforts that led to the identification of 5-amino-4-quinolone 111 with exceptionally potent Gram-positive activity with minimum inhibitory concentrations (MICs) ≤0.06 μg/mL against numerous clinical isolates. Preliminary mechanism of action and resistance studies demonstrate that the 5-amino-4-quinolones are bacteriostatic, do not select for resistance, and selectively disrupt bacterial membranes. While the precise molecular mechanism has not been elucidated, the lead compound is nontoxic displaying a therapeutic index greater than 500, is devoid of hemolytic activity, and has attractive physicochemical properties (clog P = 3.8, molecular weight (MW) = 441) that warrant further investigation of this promising antibacterial scaffold for the treatment of Gram-positive infections.
Collapse
Affiliation(s)
- John R Schultz
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Stephen K Costa
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, United States
| | - Gorakhnath R Jachak
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Pooja Hegde
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Yan Pan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Michaele Josten
- Institute for Pharmaceutical Microbiology and Institute for Medical Microbiology, Immunology, and Parasitology, University of Bonn, D-53115 Bonn, Germany
| | - Chinedu Ejeh
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, United States
| | - Travis Hammerstad
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Hans Georg Sahl
- Institute for Pharmaceutical Microbiology and Institute for Medical Microbiology, Immunology, and Parasitology, University of Bonn, D-53115 Bonn, Germany
| | - Pedro M Pereira
- Bacterial Cell Biology Laboratory, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2781-901 Oeiras, Portugal
| | - Mariana G Pinho
- Bacterial Cell Biology Laboratory, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2781-901 Oeiras, Portugal
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Ambrose Cheung
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, United States
| | - Courtney C Aldrich
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
5
|
New diarylpentanoids and chalcones as potential antimicrobial adjuvants. Bioorg Med Chem Lett 2022; 67:128743. [PMID: 35447343 DOI: 10.1016/j.bmcl.2022.128743] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 01/27/2023]
Abstract
Antimicrobial resistance arises due to several adaptation mechanisms, being the overexpression of efflux pumps (EPs) one of the most worrisome. In bacteria, EPs can also play important roles in virulence, quorum-sensing (QS) and biofilm formation. To identify new potential antimicrobial adjuvants, a library of diarylpentanoids and chalcones was synthesized and tested. These compounds presented encouraging results in potentiating the activity of antimicrobials, being diarylpentanoid 13 the most promising. Compounds 9, 13, 16, 19, 22, and 23 displayed EP inhibitory effect, mainly in Staphylococcus aureus 272123. Compounds 13, 19, 22, and 23 exhibited inhibitory effect on biofilm formation in S. aureus 272,123 while 13 and 22 inhibited QS in the pair Sphingomonas paucimobilis Ezf 10-17 and Chromobacterium violaceum CV026. The overall results, demonstrated that diarylpentanoid 13 and chalcone 22 were active against all the resistance mechanisms tested, suggesting their potential as antimicrobial adjuvants.
Collapse
|
6
|
Penicillin-Binding Protein 1 (PBP1) of Staphylococcus aureus Has Multiple Essential Functions in Cell Division. mBio 2022; 13:e0066922. [PMID: 35703435 PMCID: PMC9426605 DOI: 10.1128/mbio.00669-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Bacterial cell division is a complex process requiring the coordination of multiple components to allow the appropriate spatial and temporal control of septum formation and cell scission. Peptidoglycan (PG) is the major structural component of the septum, and our recent studies in the human pathogen Staphylococcus aureus have revealed a complex, multistage PG architecture that develops during septation. Penicillin-binding proteins (PBPs) are essential for the final steps of PG biosynthesis; their transpeptidase activity links the peptide side chains of nascent glycan strands. PBP1 is required for cell division in S. aureus, and here, we demonstrate that it has multiple essential functions associated with its enzymatic activity and as a regulator of division. Loss of PBP1, or just its C-terminal PASTA domains, results in cessation of division at the point of septal plate formation. The PASTA domains can bind PG and thereby potentially coordinate the cell division process. The transpeptidase activity of PBP1 is also essential, but its loss leads to a strikingly different phenotype of thickened and aberrant septa, which is phenocopied by the morphological effects of adding the PBP1-specific β-lactam, meropenem. Together, these results lead to a model for septal PG synthesis where PBP1 enzyme activity is required for the characteristic architecture of the septum and PBP1 protein molecules enable the formation of the septal plate.
Collapse
|
7
|
Ghosh S, Gutti S, Chaudhuri D. Pattern formation, localized and running pulsation on active spherical membranes. SOFT MATTER 2021; 17:10614-10627. [PMID: 34605510 DOI: 10.1039/d1sm00937k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Active force generation by an actin-myosin cortex coupled to a cell membrane allows the cell to deform, respond to the environment, and mediate cell motility and division. Several membrane-bound activator proteins move along it and couple to the membrane curvature. Besides, they can act as nucleating sites for the growth of filamentous actin. Actin polymerization can generate a local outward push on the membrane. Inward pull from the contractile actomyosin cortex can propagate along the membrane via actin filaments. We use coupled evolution of fields to perform linear stability analysis and numerical calculations. As activity overcomes the stabilizing factors such as surface tension and bending rigidity, the spherical membrane shows instability towards pattern formation, localized pulsation, and running pulsation between poles. We present our results in terms of phase diagrams and evolutions of the coupled fields. They have relevance for living cells and can be verified in experiments on artificial cell-like constructs.
Collapse
Affiliation(s)
- Subhadip Ghosh
- Department of Physics, Faculty of Science, University of Zagreb, Bijenička cesta 32, 10000 Zagreb, Croatia.
| | - Sashideep Gutti
- BITS Pilani Hyderabad Campus, Hyderabad 500078, Telengana, India.
| | - Debasish Chaudhuri
- Institute of Physics, Sachivalaya Marg, Bhubaneswar 751005, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
8
|
The cell envelope of Staphylococcus aureus selectively controls the sorting of virulence factors. Nat Commun 2021; 12:6193. [PMID: 34702812 PMCID: PMC8548510 DOI: 10.1038/s41467-021-26517-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 10/05/2021] [Indexed: 11/08/2022] Open
Abstract
Staphylococcus aureus bi-component pore-forming leukocidins are secreted toxins that directly target and lyse immune cells. Intriguingly, one of the leukocidins, Leukocidin AB (LukAB), is found associated with the bacterial cell envelope in addition to secreted into the extracellular milieu. Here, we report that retention of LukAB on the bacterial cells provides S. aureus with a pre-synthesized active toxin that kills immune cells. On the bacteria, LukAB is distributed as discrete foci in two distinct compartments: membrane-proximal and surface-exposed. Through genetic screens, we show that a membrane lipid, lysyl-phosphatidylglycerol (LPG), and lipoteichoic acid (LTA) contribute to LukAB deposition and release. Furthermore, by studying non-covalently surface-bound proteins we discovered that the sorting of additional exoproteins, such as IsaB, Hel, ScaH, and Geh, are also controlled by LPG and LTA. Collectively, our study reveals a multistep secretion system that controls exoprotein storage and protein translocation across the S. aureus cell wall.
Collapse
|
9
|
Perez AJ, Boersma MJ, Bruce KE, Lamanna MM, Shaw SL, Tsui HCT, Taguchi A, Carlson EE, VanNieuwenhze MS, Winkler ME. Organization of peptidoglycan synthesis in nodes and separate rings at different stages of cell division of Streptococcus pneumoniae. Mol Microbiol 2020; 115:1152-1169. [PMID: 33269494 DOI: 10.1111/mmi.14659] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/15/2022]
Abstract
Bacterial peptidoglycan (PG) synthesis requires strict spatiotemporal organization to reproduce specific cell shapes. In ovoid-shaped Streptococcus pneumoniae (Spn), septal and peripheral (elongation) PG synthesis occur simultaneously at midcell. To uncover the organization of proteins and activities that carry out these two modes of PG synthesis, we examined Spn cells vertically oriented onto their poles to image the division plane at the high lateral resolution of 3D-SIM (structured-illumination microscopy). Labeling with fluorescent D-amino acids (FDAA) showed that areas of new transpeptidase (TP) activity catalyzed by penicillin-binding proteins (PBPs) separate into a pair of concentric rings early in division, representing peripheral PG (pPG) synthesis (outer ring) and the leading-edge (inner ring) of septal PG (sPG) synthesis. Fluorescently tagged PBP2x or FtsZ locate primarily to the inner FDAA-marked ring, whereas PBP2b and FtsX remain in the outer ring, suggesting roles in sPG or pPG synthesis, respectively. Pulses of FDAA labeling revealed an arrangement of separate regularly spaced "nodes" of TP activity around the division site of predivisional cells. Tagged PBP2x, PBP2b, and FtsX proteins also exhibited nodal patterns with spacing comparable to that of FDAA labeling. Together, these results reveal new aspects of spatially ordered PG synthesis in ovococcal bacteria during cell division.
Collapse
Affiliation(s)
- Amilcar J Perez
- Department of Biology, Indiana University Bloomington, Bloomington, IN, USA
| | - Michael J Boersma
- Department of Biology, Indiana University Bloomington, Bloomington, IN, USA
| | - Kevin E Bruce
- Department of Biology, Indiana University Bloomington, Bloomington, IN, USA
| | - Melissa M Lamanna
- Department of Biology, Indiana University Bloomington, Bloomington, IN, USA
| | - Sidney L Shaw
- Department of Biology, Indiana University Bloomington, Bloomington, IN, USA
| | - Ho-Ching T Tsui
- Department of Biology, Indiana University Bloomington, Bloomington, IN, USA
| | - Atsushi Taguchi
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Erin E Carlson
- Department of Chemistry, University of Minnesota, Minneapolis, MN, USA
| | | | - Malcolm E Winkler
- Department of Biology, Indiana University Bloomington, Bloomington, IN, USA
| |
Collapse
|
10
|
Polyhedral liquid droplets: Recent advances in elucidation and application. Curr Opin Colloid Interface Sci 2020. [DOI: 10.1016/j.cocis.2020.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
11
|
Carlson AL, Daigger GT, Love NG, Hart E. Multi-year diagnosis of unpredictable fouling occurrences in a full-scale membrane bioreactor. WATER SCIENCE AND TECHNOLOGY : A JOURNAL OF THE INTERNATIONAL ASSOCIATION ON WATER POLLUTION RESEARCH 2020; 82:524-536. [PMID: 32960797 DOI: 10.2166/wst.2020.354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The membrane bioreactor (MBR) at the Traverse City Regional Wastewater Treatment Plant has experienced sudden and unpredictable periods of substantial permeability decline since 2011. Early observations detected irregularly-shaped Gram-positive bacteria that correlated with plant upsets. Use of biomolecular techniques, such as DNA sequencing of laboratory isolates and the mixed liquor microbial community, and fluorescent in situ hybridization, identified the dispersed organisms as members of the genus Staphylococcus. However, Staphylococcus species were consistently present during normal operation and therefore were more likely to be an indicator of the upset, not the cause. The results suggest that these microorganisms are responding to specific influent wastewater constituents. We chemically analysed seven mixed liquor samples from periods of permeability decline in 2017 and 2018, and four samples from a period of normal operation. During upset conditions, the total carbohydrate content exceeded that of normal operation by 40%. Additionally, mixed liquor calcium concentrations were 65% above normal during the upset in 2017. It is hypothesized and supported through multivariate statistical analysis and estimation of specific resistance to filtration values that a calcium-intermediated polymer bridging mechanism with extracellular polymeric substance constituents is a major contributor to fouling and permeability disruptions in the Traverse City MBR.
Collapse
Affiliation(s)
- A L Carlson
- Department of Environmental Engineering, University of Michigan, 1351 Beal Avenue, Ann Arbor, MI 48109, USA E-mail:
| | - G T Daigger
- Department of Environmental Engineering, University of Michigan, 1351 Beal Avenue, Ann Arbor, MI 48109, USA E-mail:
| | - N G Love
- Department of Environmental Engineering, University of Michigan, 1351 Beal Avenue, Ann Arbor, MI 48109, USA E-mail:
| | - E Hart
- Traverse City Regional Wastewater Treatment Plant, 606 Hannah Avenue, Traverse City, MI 49686, USA
| |
Collapse
|
12
|
Liber SR, Butenko AV, Caspi M, Guttman S, Schultz M, Schofield AB, Deutsch M, Sloutskin E. Precise Self-Positioning of Colloidal Particles on Liquid Emulsion Droplets. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:13053-13061. [PMID: 31502850 DOI: 10.1021/acs.langmuir.9b01833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Decorating emulsion droplets by particles stabilizes foodstuff and pharmaceuticals. Interfacial particles also influence aerosol formation, thus impacting atmospheric CO2 exchange. While studies of particles at disordered droplet interfaces abound in the literature, such studies for ubiquitous ordered interfaces are not available. Here, we report such an experimental study, showing that particles residing at crystalline interfaces of liquid droplets spontaneously self-position to specific surface locations, identified as structural topological defects in the crystalline surface monolayer. This monolayer forms at temperature T = Ts, leaving the droplet liquid and driving at Td < Ts a spontaneous shape-change transition of the droplet from spherical to icosahedral. The particle's surface position remains unchanged in the transition, demonstrating these positions to coincide with the vertices of the sphere-inscribed icosahedron. Upon further cooling, droplet shape-changes to other polyhedra occur, with the particles remaining invariably at the polyhedra's vertices. At still lower temperatures, the particles are spontaneously expelled from the droplets. Our results probe the molecular-scale elasticity of quasi-two-dimensional curved crystals, impacting also other fields, such as protein positioning on cell membranes, controlling essential biological functions. Using ligand-decorated particles, and the precise temperature-tunable surface position control found here, may also allow using these droplets for directed supra-droplet self-assembly into larger structures, with a possible post-assembly structure fixation by UV polymerization of the droplet's liquid.
Collapse
Affiliation(s)
- Shir R Liber
- Physics Department and Institute of Nanotechnology & Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Alexander V Butenko
- Physics Department and Institute of Nanotechnology & Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Moshe Caspi
- Physics Department and Institute of Nanotechnology & Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Shani Guttman
- Physics Department and Institute of Nanotechnology & Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Moty Schultz
- Physics Department and Institute of Nanotechnology & Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Andrew B Schofield
- The School of Physics and Astronomy , University of Edinburgh , Edinburgh EH9 3FD , U.K
| | - Moshe Deutsch
- Physics Department and Institute of Nanotechnology & Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Eli Sloutskin
- Physics Department and Institute of Nanotechnology & Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| |
Collapse
|
13
|
Mouwakeh A, Kincses A, Nové M, Mosolygó T, Mohácsi-Farkas C, Kiskó G, Spengler G. Nigella sativa essential oil and its bioactive compounds as resistance modifiers against Staphylococcus aureus. Phytother Res 2019; 33:1010-1018. [PMID: 30672036 DOI: 10.1002/ptr.6294] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/04/2018] [Accepted: 12/31/2018] [Indexed: 11/09/2022]
Abstract
Nigella sativa essential oil (EO) and its compounds (thymoquinone, carvacrol, and p-cymene) have a broad antimicrobial spectrum. The aim of this study was to investigate the antimicrobial and resistance modifying activity of N. sativa EO, thymoquinone, carvacrol, and p-cymene against one methicillin susceptible and one methicillin resistant Staphylococcus aureus strain. N. sativa EO, thymoquinone, carvacrol, and p-cymene were assessed for antimicrobial activity and modulation of antimicrobial resistance (by broth microdilution), inhibition of antimicrobial efflux (by ethidium bromide [EtBr] accumulation assay), relative expression of mepA gene (by real-time reverse transcriptase quantitative polymerase chain reaction), membrane disrupting effect (by LIVE/DEAD BacLight™ Kit), and finally antibiofilm activity (by the crystal violet assay). Both strains of S. aureus were susceptible to N. sativa EO, thymoquinone, and carvacrol. N. sativa EO and carvacrol induced the increase of EtBr accumulated by both S. aureus strains. Membrane integrity of ATCC strain was disrupted by carvacrol and p-cymene, whereas for the methicillin resistant S. aureus (MRSA) strain the membrane integrity was disrupted by each compound. N. sativa EO and its bioactive compounds such as carvacrol and p-cymene could be applied as resistance modifiers in MRSA strains.
Collapse
Affiliation(s)
- Ahmad Mouwakeh
- Department of Microbiology and Biotechnology, Szent István University, Budapest, Hungary
| | - Annamária Kincses
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | - Márta Nové
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | - Tímea Mosolygó
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | - Csilla Mohácsi-Farkas
- Department of Microbiology and Biotechnology, Szent István University, Budapest, Hungary
| | - Gabriella Kiskó
- Department of Microbiology and Biotechnology, Szent István University, Budapest, Hungary
| | - Gabriella Spengler
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| |
Collapse
|
14
|
Nagendra Prasad HS, Karthik CS, Manukumar HM, Mallesha L, Mallu P. New approach to address antibiotic resistance: Miss loading of functional membrane microdomains (FMM) of methicillin-resistant Staphylococcus aureus (MRSA). Microb Pathog 2018; 127:106-115. [PMID: 30503959 DOI: 10.1016/j.micpath.2018.11.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 12/13/2022]
Abstract
The synthesized potent piperazine analog ChDiPiCa was characterised by various spectroscopic techniques and for the first time evaluated functional membrane microdomain (FMM) disassembly in methicillin-resistant Staphylococcus aureus (MRSA). The ChDiPiCa showed excellent in vitro biocidal activity against MRSA at 26 μg/mL compared to the antibiotic streptomycin and bacitracin 14 μg/mL and 13 μg/mL at 10 μg concentration respectively. The membrane damaging property was confirmed by the SEM analysis. Further, we addressed the new approach for the first time to overcome antibiotic resistance of MRSA through membrane microdomain miss loading to lipids. By which, the ChDiPiCa confirms the significant activity in miss loading of FMM of MRSA which is validated by the fatty acid profile and lipid analysis. The result shows that, altered saturated (Lauric acid and Myristic acid), mono unsaturated (Oleic acid), and poly unsaturated (Linoleic acid and Linolenic acid) fatty acids and hypothesises, altered the membrane functional lipids. For the better understanding of miss loading of FMM by the ChDiPiCa, the in-silico molecular docking studies was analyzed and confirmed the predicted role. This suggests the way to develop ChDiPiCa in medicinal chemistry as anti-MRSA candidates and also this report opens up new window to treat microbial pathogens and infections.
Collapse
Affiliation(s)
- H S Nagendra Prasad
- Department of Chemistry, Sri Jayachamarajendra College of Engineering, JSS Science and Technology University, Mysuru, 570 006, Karnataka, India
| | - C S Karthik
- Department of Chemistry, Sri Jayachamarajendra College of Engineering, JSS Science and Technology University, Mysuru, 570 006, Karnataka, India
| | - H M Manukumar
- Department of Chemistry, Sri Jayachamarajendra College of Engineering, JSS Science and Technology University, Mysuru, 570 006, Karnataka, India
| | - L Mallesha
- PG Department of Chemistry, JSS College of Arts, Commerce and Science, Mysuru, 570025, Karnataka, India
| | - P Mallu
- Department of Chemistry, Sri Jayachamarajendra College of Engineering, JSS Science and Technology University, Mysuru, 570 006, Karnataka, India.
| |
Collapse
|
15
|
Weihs F, Wacnik K, Turner RD, Culley S, Henriques R, Foster SJ. Heterogeneous localisation of membrane proteins in Staphylococcus aureus. Sci Rep 2018; 8:3657. [PMID: 29483609 PMCID: PMC5826919 DOI: 10.1038/s41598-018-21750-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/08/2018] [Indexed: 11/25/2022] Open
Abstract
The bacterial cytoplasmic membrane is the interface between the cell and its environment, with multiple membrane proteins serving its many functions. However, how these proteins are organised to permit optimal physiological processes is largely unknown. Based on our initial findings that 2 phospholipid biosynthetic enzymes (PlsY and CdsA) localise heterogeneously in the membrane of the bacterium Staphylococcus aureus, we have analysed the localisation of other key membrane proteins. A range of protein fusions were constructed and used in conjunction with quantitative image analysis. Enzymes involved in phospholipid biosynthesis as well as the lipid raft marker FloT exhibited a heterogeneous localisation pattern. However, the secretion associated SecY protein, was more homogeneously distributed in the membrane. A FRET-based system also identified novel colocalisation between phospholipid biosynthesis enzymes and the respiratory protein CydB revealing a likely larger network of partners. PlsY localisation was found to be dose dependent but not to be affected by membrane lipid composition. Disruption of the activity of the essential cell division organiser FtsZ, using the inhibitor PC190723 led to loss of PlsY localisation, revealing a link to cell division and a possible role for FtsZ in functions not strictly associated with septum formation.
Collapse
Affiliation(s)
- Felix Weihs
- The Krebs Institute. Department of Molecular Biology and Microbiology, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Katarzyna Wacnik
- The Krebs Institute. Department of Molecular Biology and Microbiology, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Robert D Turner
- The Krebs Institute. Department of Molecular Biology and Microbiology, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Siân Culley
- Quantitative Imaging and Nanobiophysics Group, MRC Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- The Francis Crick Institute, 1 Midland Rd, Kings Cross, London, NW1 1AT, UK
| | - Ricardo Henriques
- Quantitative Imaging and Nanobiophysics Group, MRC Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- The Francis Crick Institute, 1 Midland Rd, Kings Cross, London, NW1 1AT, UK
| | - Simon J Foster
- The Krebs Institute. Department of Molecular Biology and Microbiology, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
16
|
Van der Leek AP, Yanishevsky Y, Kozyrskyj AL. The Kynurenine Pathway As a Novel Link between Allergy and the Gut Microbiome. Front Immunol 2017; 8:1374. [PMID: 29163472 PMCID: PMC5681735 DOI: 10.3389/fimmu.2017.01374] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022] Open
Abstract
In the past few decades, the indoleamine 2,3 dioxygenase (IDO) subset of the kynurenine (KYN) pathway of tryptophan (TRP) metabolism has been the subject of much research in the area of immune tolerance. In this review, we aim to incorporate new findings on this pathway in relation to allergy and the gut microbiome, while providing a comprehensive overview of the pathway itself. Stimulated by interferon gamma, IDO acts as a tolerogenic, immunosuppressive enzyme to attenuate allergic responses by the induction of the KYN-IDO pathway, resultant depletion of TRP, and elevation in KYN metabolites. Acting through the aryl hydrocarbon receptor, KYN metabolites cause T-cell anergy and apoptosis, proliferation of Treg and Th17 cells, and deviation of the Th1/Th2 response, although the outcome is highly dependent on the microenvironment. Moreover, new evidence from germ-free mice and human infants shows that gut microbiota and breast milk are key in determining the functioning of the KYN-IDO pathway. As such, we recommend further research on how this pathway may be a critical link between the microbiome and development of allergy.
Collapse
Affiliation(s)
| | | | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada.,Department of Public Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
17
|
García-Fernández E, Koch G, Wagner RM, Fekete A, Stengel ST, Schneider J, Mielich-Süss B, Geibel S, Markert SM, Stigloher C, Lopez D. Membrane Microdomain Disassembly Inhibits MRSA Antibiotic Resistance. Cell 2017; 171:1354-1367.e20. [PMID: 29103614 PMCID: PMC5720476 DOI: 10.1016/j.cell.2017.10.012] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/18/2017] [Accepted: 10/06/2017] [Indexed: 12/21/2022]
Abstract
A number of bacterial cell processes are confined functional membrane microdomains (FMMs), structurally and functionally similar to lipid rafts of eukaryotic cells. How bacteria organize these intricate platforms and what their biological significance is remain important questions. Using the pathogen methicillin-resistant Staphylococcus aureus (MRSA), we show here that membrane-carotenoid interaction with the scaffold protein flotillin leads to FMM formation, which can be visualized using super-resolution array tomography. These membrane platforms accumulate multimeric protein complexes, for which flotillin facilitates efficient oligomerization. One of these proteins is PBP2a, responsible for penicillin resistance in MRSA. Flotillin mutants are defective in PBP2a oligomerization. Perturbation of FMM assembly using available drugs interferes with PBP2a oligomerization and disables MRSA penicillin resistance in vitro and in vivo, resulting in MRSA infections that are susceptible to penicillin treatment. Our study demonstrates that bacteria possess sophisticated cell organization programs and defines alternative therapies to fight multidrug-resistant pathogens using conventional antibiotics. Staphyloxanthin and flotillin preferentially interact and accumulate in FMMs FMMs facilitate efficient oligomerization of multimeric protein complexes PBP2a, which confers β-lactam resistance on S. aureus, is harbored within FMMs FMM disruption disables PBP2a oligomerization and thus, S. aureus antibiotic resistance
Collapse
Affiliation(s)
- Esther García-Fernández
- National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), 28049 Madrid, Spain
| | - Gudrun Koch
- Research Centre for Infectious Diseases (ZINF), University of Würzburg, 97080 Würzburg, Germany; Institute for Molecular Infection Biology (IMIB), University of Würzburg, 97080 Würzburg, Germany
| | - Rabea M Wagner
- National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), 28049 Madrid, Spain; Research Centre for Infectious Diseases (ZINF), University of Würzburg, 97080 Würzburg, Germany; Institute for Molecular Infection Biology (IMIB), University of Würzburg, 97080 Würzburg, Germany
| | - Agnes Fekete
- Julius-von-Sachs-Institute Biocenter, Pharmaceutical Biology, University of Würzburg, 97082 Würzburg, Germany
| | - Stephanie T Stengel
- Research Centre for Infectious Diseases (ZINF), University of Würzburg, 97080 Würzburg, Germany; Institute for Molecular Infection Biology (IMIB), University of Würzburg, 97080 Würzburg, Germany
| | - Johannes Schneider
- Research Centre for Infectious Diseases (ZINF), University of Würzburg, 97080 Würzburg, Germany; Institute for Molecular Infection Biology (IMIB), University of Würzburg, 97080 Würzburg, Germany
| | - Benjamin Mielich-Süss
- Research Centre for Infectious Diseases (ZINF), University of Würzburg, 97080 Würzburg, Germany; Institute for Molecular Infection Biology (IMIB), University of Würzburg, 97080 Würzburg, Germany
| | - Sebastian Geibel
- Research Centre for Infectious Diseases (ZINF), University of Würzburg, 97080 Würzburg, Germany; Institute for Molecular Infection Biology (IMIB), University of Würzburg, 97080 Würzburg, Germany
| | - Sebastian M Markert
- Division of Electron Microscopy, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Christian Stigloher
- Division of Electron Microscopy, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Daniel Lopez
- National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), 28049 Madrid, Spain; Research Centre for Infectious Diseases (ZINF), University of Würzburg, 97080 Würzburg, Germany; Institute for Molecular Infection Biology (IMIB), University of Würzburg, 97080 Würzburg, Germany; National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Darwin 3, Campus de Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
18
|
Straume D, Stamsås GA, Berg KH, Salehian Z, Håvarstein LS. Identification of pneumococcal proteins that are functionally linked to penicillin-binding protein 2b (PBP2b). Mol Microbiol 2016; 103:99-116. [DOI: 10.1111/mmi.13543] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Daniel Straume
- Department of Chemistry, Biotechnology and Food Science; Norwegian University of Life Sciences; Ås NO-1432 Norway
| | - Gro Anita Stamsås
- Department of Chemistry, Biotechnology and Food Science; Norwegian University of Life Sciences; Ås NO-1432 Norway
| | - Kari Helene Berg
- Department of Chemistry, Biotechnology and Food Science; Norwegian University of Life Sciences; Ås NO-1432 Norway
| | - Zhian Salehian
- Department of Chemistry, Biotechnology and Food Science; Norwegian University of Life Sciences; Ås NO-1432 Norway
| | - Leiv Sigve Håvarstein
- Department of Chemistry, Biotechnology and Food Science; Norwegian University of Life Sciences; Ås NO-1432 Norway
| |
Collapse
|