1
|
Van Genechten W, Vergauwen R, Van Dijck P. The intricate link between iron, mitochondria and azoles in Candida species. FEBS J 2024; 291:3568-3580. [PMID: 37846606 DOI: 10.1111/febs.16977] [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: 05/21/2023] [Revised: 09/19/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Invasive fungal infections are rapidly increasing, and the opportunistic pathogenic Candida species are the fourth most common cause of nosocomial systemic infections. The current antifungal classes, of which azoles are the most widely used, all have shortcomings. Azoles are generally considered fungistatic rather than fungicidal, they do not actively kill fungal cells and therefore resistance against azoles can be rapidly acquired. Combination therapies with azoles provide an interesting therapeutic outlook and agents limiting iron are excellent candidates. We summarize how iron is acquired by the host and transported towards both storage and iron-utilizing organelles. We indicate whether these pathways alter azole susceptibility and/or tolerance, to finally link these transport mechanisms to mitochondrial iron availability. In this review, we highlight putative novel intracellular iron shuffling mechanisms and indicate that mitochondrial iron dynamics in relation to azole treatment and iron limitation is a significant knowledge gap.
Collapse
Affiliation(s)
- Wouter Van Genechten
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Belgium
| | - Rudy Vergauwen
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Belgium
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Belgium
| |
Collapse
|
2
|
Mokhtari M, Amiri P, Miller D, Gresham D, Bloor SJ, Munkacsi AB. Chemical genomic analysis reveals the interplay between iron chelation, zinc homeostasis, and retromer function in the bioactivity of an ethanol adduct of the feijoa fruit-derived ellagitannin vescalagin. G3 (BETHESDA, MD.) 2024; 14:jkae098. [PMID: 38805688 PMCID: PMC11228861 DOI: 10.1093/g3journal/jkae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/30/2024]
Abstract
Nature has been a rich source of pharmaceutical compounds, producing 80% of our currently prescribed drugs. The feijoa plant, Acca sellowiana, is classified in the family Myrtaceae, native to South America, and currently grown worldwide to produce feijoa fruit. Feijoa is a rich source of bioactive compounds with anticancer, anti-inflammatory, antibacterial, and antifungal activities; however, the mechanism of action of these compounds is largely not known. Here, we used chemical genetic analyses in the model organism Saccharomyces cerevisiae to investigate the mechanism of action of a feijoa-derived ethanol adduct of vescalagin (EtOH-vescalagin). Genome-wide barcode sequencing analysis revealed yeast strains lacking genes in iron metabolism, zinc metabolism, retromer function, or mitochondrial function were hypersensitive to 0.3 µM EtOH-vescalagin. This treatment increased expression of iron uptake proteins at the plasma membrane, which was a compensatory response to reduced intracellular iron. Likewise, EtOH-vescalagin increased expression of the Cot1 protein in the vacuolar membrane that transports zinc into the vacuole to prevent cytoplasmic accumulation of zinc. Each individual subunit in the retromer complex was required for the iron homeostatic mechanism of EtOH-vescalagin, while only the cargo recognition component in the retromer complex was required for the zinc homeostatic mechanism. Overexpression of either retromer subunits or high-affinity iron transporters suppressed EtOH-vescalagin bioactivity in a zinc-replete condition, while overexpression of only retromer subunits increased EtOH-vescalagin bioactivity in a zinc-deficient condition. Together, these results indicate that EtOH-vescalagin bioactivity begins with extracellular iron chelation and proceeds with intracellular transport of zinc via the retromer complex. More broadly, this is the first report of a bioactive compound to further characterize the poorly understood interaction between zinc metabolism and retromer function.
Collapse
Affiliation(s)
- Mona Mokhtari
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Pegah Amiri
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Darach Miller
- Department of Genetics, Stanford University Medical School, Stanford, CA 94305, USA
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - David Gresham
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | | | - Andrew B Munkacsi
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| |
Collapse
|
3
|
Yang J, Park S, Kim HJ, Lee SJ, Jung WH. The Interkingdom Interaction with Staphylococcus Influences the Antifungal Susceptibility of the Cutaneous Fungus Malassezia. J Microbiol Biotechnol 2023; 33:180-187. [PMID: 36575858 PMCID: PMC9998211 DOI: 10.4014/jmb.2210.10039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022]
Abstract
The skin is a dynamic ecosystem on which diverse microbes reside. The interkingdom interaction between microbial species in the skin microbiota is thought to influence the health and disease of the skin although the roles of the intra- and interkingdom interactions remain to be elucidated. In this context, the interactions between Malassezia and Staphylococcus, the most dominant microorganisms in the skin microbiota, have gained attention. This study investigated how the interaction between Malassezia and Staphylococcus affected the antifungal susceptibility of the fungus to the azole antifungal drug ketoconazole. The susceptibility was significantly decreased when Malassezia was co-cultured with Staphylococcus. We found that acidification of the environment by organic acids produced by Staphylococcus influenced the decrease of the ketoconazole susceptibility of M. restricta in the co-culturing condition. Furthermore, our data demonstrated that the significant increased ergosterol content and cell membrane and wall thickness of the M. restricta cells grown in the acidic environment may be the main cause of the altered azole susceptibility of the fungus. Overall, our study suggests that the interaction between Malassezia and Staphylococcus influences the antifungal susceptibility of the fungus and that pH has a critical role in the polymicrobial interaction in the skin environment.
Collapse
Affiliation(s)
- Juan Yang
- Department of Systems Biotechnology and Institute of Microbiomics, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Sungmin Park
- Department of Systems Biotechnology and Institute of Microbiomics, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Hyun Ju Kim
- Department of Systems Biotechnology and Institute of Microbiomics, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Sang Jun Lee
- Department of Systems Biotechnology and Institute of Microbiomics, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Won Hee Jung
- Department of Systems Biotechnology and Institute of Microbiomics, Chung-Ang University, Anseong 17546, Republic of Korea
| |
Collapse
|
4
|
Ghaffarieh A, Ciolino JB. Potential of Application of Iron Chelating Agents in Ophthalmic Diseases. Semin Ophthalmol 2021; 36:157-161. [PMID: 33621147 DOI: 10.1080/08820538.2021.1887900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The investigations discussed in this review indicate that iron may exacerbate different eye diseases. Therefore, it is plausible that reducing cellular or body iron stores could influence disease pathogenesis, so it is logical to consider the iron chelators' potential protective role in the various ophthalmic diseases in the form of topical eye drops or slow releasing injectable compounds as an adjuvant treatment.
Collapse
Affiliation(s)
- Alireza Ghaffarieh
- Ophthalmology, Cornea, External Disease & Refractive Surgery Fellow, Massachusetts Eye and Ear Infirmary, Harvard University, USA
| | - Joseph B Ciolino
- Ophthalmology, Associate Professor of Ophthalmology, Harvard Medical School Mass. Eye and Ear/Schepens Eye Research Institute, USA
| |
Collapse
|
5
|
Houshmandyar S, Eggleston IM, Bolhuis A. Biofilm-specific uptake of a 4-pyridone-based iron chelator by Pseudomonas aeruginosa. Biometals 2021; 34:315-328. [PMID: 33428087 PMCID: PMC7940164 DOI: 10.1007/s10534-020-00281-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/21/2020] [Indexed: 11/20/2022]
Abstract
Iron is an essential nutrient for virtually all microbes and limiting the concentration of available iron is a potential strategy to be used as an alternative to antibiotic treatment. In this study we analysed the antimicrobial activity of two chelators, specifically 3-hydroxy-1,2-dimethyl-4(1H)-pyridone (deferiprone, DFP), which is clinically approved for the treatment of iron overload disorders, and its 1,2-diethyl homologue, CP94. Both compounds showed moderate activity towards planktonically growing P. aeruginosa cells, and the mechanism of action of these chelators was indeed by limiting the amount of free iron. Surprisingly, the compounds behaved very differently when the cells were grown in biofilms. DFP also showed inhibitory effects on biofilm formation but in contrast, CP94 stimulated this process, in particular at high concentrations. We hypothesised that CP94 behaves as an iron carrier, which was confirmed by our observation that it had antimicrobial synergy with the toxic metals, gallium and copper. This suggests that P. aeruginosa produces a biofilm-specific transport protein that recognises CP94 but not the closely related compound DFP.
Collapse
Affiliation(s)
| | - Ian M Eggleston
- Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, UK
| | - Albert Bolhuis
- Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, UK.
| |
Collapse
|
6
|
Zarzosa-Moreno D, Avalos-Gómez C, Ramírez-Texcalco LS, Torres-López E, Ramírez-Mondragón R, Hernández-Ramírez JO, Serrano-Luna J, de la Garza M. Lactoferrin and Its Derived Peptides: An Alternative for Combating Virulence Mechanisms Developed by Pathogens. Molecules 2020; 25:E5763. [PMID: 33302377 PMCID: PMC7762604 DOI: 10.3390/molecules25245763] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022] Open
Abstract
Due to the emergence of multidrug-resistant pathogens, it is necessary to develop options to fight infections caused by these agents. Lactoferrin (Lf) is a cationic nonheme multifunctional glycoprotein of the innate immune system of mammals that provides numerous benefits. Lf is bacteriostatic and/or bactericidal, can stimulate cell proliferation and differentiation, facilitate iron absorption, improve neural development and cognition, promote bone growth, prevent cancer and exert anti-inflammatory and immunoregulatory effects. Lactoferrin is present in colostrum and milk and is also produced by the secondary granules of polymorphonuclear leukocytes, which store this glycoprotein and release it at sites of infection. Lf is also present in many fluids and exocrine secretions, on the surfaces of the digestive, respiratory and reproductive systems that are commonly exposed to pathogens. Apo-Lf (an iron-free molecule) can be microbiostatic due to its ability to capture ferric iron, blocking the availability of host iron to pathogens. However, apo-Lf is mostly microbicidal via its interaction with the microbial surface, causing membrane damage and altering its permeability function. Lf can inhibit viral entry by binding to cell receptors or viral particles. Lf is also able to counter different important mechanisms evolved by microbial pathogens to infect and invade the host, such as adherence, colonization, invasion, production of biofilms and production of virulence factors such as proteases and toxins. Lf can also cause mitochondrial and caspase-dependent regulated cell death and apoptosis-like in pathogenic yeasts. All of these mechanisms are important targets for treatment with Lf. Holo-Lf (the iron-saturated molecule) can contain up to two ferric ions and can also be microbicidal against some pathogens. On the other hand, lactoferricins (Lfcins) are peptides derived from the N-terminus of Lf that are produced by proteolysis with pepsin under acidic conditions, and they cause similar effects on pathogens to those caused by the parental Lf. Synthetic analog peptides comprising the N-terminus Lf region similarly exhibit potent antimicrobial properties. Importantly, there are no reported pathogens that are resistant to Lf and Lfcins; in addition, Lf and Lfcins have shown a synergistic effect with antimicrobial and antiviral drugs. Due to the Lf properties being microbiostatic, microbicidal, anti-inflammatory and an immune modulator, it represents an excellent natural alternative either alone or as adjuvant in the combat to antibiotic multidrug-resistant bacteria and other pathogens. This review aimed to evaluate the data that appeared in the literature about the effects of Lf and its derived peptides on pathogenic bacteria, protozoa, fungi and viruses and how Lf and Lfcins inhibit the mechanisms developed by these pathogens to cause disease.
Collapse
Affiliation(s)
- Daniela Zarzosa-Moreno
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Zacatenco 07360, CdMx, Mexico; (D.Z.-M.); (C.A.-G.); (J.S.-L.)
| | - Christian Avalos-Gómez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Zacatenco 07360, CdMx, Mexico; (D.Z.-M.); (C.A.-G.); (J.S.-L.)
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Coyoacán 04510, CdMx, Mexico
| | - Luisa Sofía Ramírez-Texcalco
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Estado de México, Mexico; (L.S.R.-T.); (E.T.-L.); (R.R.-M.); (J.O.H.-R.)
| | - Erick Torres-López
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Estado de México, Mexico; (L.S.R.-T.); (E.T.-L.); (R.R.-M.); (J.O.H.-R.)
| | - Ricardo Ramírez-Mondragón
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Estado de México, Mexico; (L.S.R.-T.); (E.T.-L.); (R.R.-M.); (J.O.H.-R.)
| | - Juan Omar Hernández-Ramírez
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Estado de México, Mexico; (L.S.R.-T.); (E.T.-L.); (R.R.-M.); (J.O.H.-R.)
| | - Jesús Serrano-Luna
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Zacatenco 07360, CdMx, Mexico; (D.Z.-M.); (C.A.-G.); (J.S.-L.)
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Zacatenco 07360, CdMx, Mexico; (D.Z.-M.); (C.A.-G.); (J.S.-L.)
| |
Collapse
|
7
|
Stanford FA, Voigt K. Iron Assimilation during Emerging Infections Caused by Opportunistic Fungi with emphasis on Mucorales and the Development of Antifungal Resistance. Genes (Basel) 2020; 11:genes11111296. [PMID: 33143139 PMCID: PMC7693903 DOI: 10.3390/genes11111296] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/23/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Iron is a key transition metal required by most microorganisms and is prominently utilised in the transfer of electrons during metabolic reactions. The acquisition of iron is essential and becomes a crucial pathogenic event for opportunistic fungi. Iron is not readily available in the natural environment as it exists in its insoluble ferric form, i.e., in oxides and hydroxides. During infection, the host iron is bound to proteins such as transferrin, ferritin, and haemoglobin. As such, access to iron is one of the major hurdles that fungal pathogens must overcome in an immunocompromised host. Thus, these opportunistic fungi utilise three major iron acquisition systems to overcome this limiting factor for growth and proliferation. To date, numerous iron acquisition pathways have been fully characterised, with key components of these systems having major roles in virulence. Most recently, proteins involved in these pathways have been linked to the development of antifungal resistance. Here, we provide a detailed review of our current knowledge of iron acquisition in opportunistic fungi, and the role iron may have on the development of resistance to antifungals with emphasis on species of the fungal basal lineage order Mucorales, the causative agents of mucormycosis.
Collapse
Affiliation(s)
- Felicia Adelina Stanford
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology–Hans Knöll Institute, Jena, Adolf-Reichwein-Straße 23, 07745 Jena, Germany;
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich-Schiller University Jena, Neugasse 25, 07743 Jena, Germany
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology–Hans Knöll Institute, Jena, Adolf-Reichwein-Straße 23, 07745 Jena, Germany;
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich-Schiller University Jena, Neugasse 25, 07743 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Jena Microbial Resource Collection Adolf-Reichwein-Straße 23, 07745 Jena, Germany
- Correspondence: ; Tel.: +49-3641-532-1395; Fax: +49-3641-532-2395
| |
Collapse
|
8
|
Caruffo M, Mandakovic D, Mejías M, Chávez-Báez I, Salgado P, Ortiz D, Montt L, Pérez-Valenzuela J, Vera-Tamargo F, Yánez JM, Wacyk J, Pulgar R. Pharmacological iron-chelation as an assisted nutritional immunity strategy against Piscirickettsia salmonis infection. Vet Res 2020; 51:134. [PMID: 33115510 PMCID: PMC7592559 DOI: 10.1186/s13567-020-00845-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Salmonid Rickettsial Septicaemia (SRS), caused by Piscirickettsia salmonis, is a severe bacterial disease in the Chilean salmon farming industry. Vaccines and antibiotics are the current strategies to fight SRS; however, the high frequency of new epizootic events confirms the need to develop new strategies to combat this disease. An innovative opportunity is perturbing the host pathways used by the microorganisms to replicate inside host cells through host-directed antimicrobial drugs (HDAD). Iron is a critical nutrient for P. salmonis infection; hence, the use of iron-chelators becomes an excellent alternative to be used as HDAD. The aim of this work was to use the iron chelator Deferiprone (DFP) as HDAD to treat SRS. Here, we describe the protective effect of the iron chelator DFP over P. salmonis infections at non-antibiotic concentrations, in bacterial challenges both in vitro and in vivo. At the cellular level, our results indicate that DFP reduced the intracellular iron content by 33.1% and P. salmonis relative load during bacterial infections by 78%. These findings were recapitulated in fish, where DFP reduced the mortality of rainbow trout challenged with P. salmonis in 34.9% compared to the non-treated group. This is the first report of the protective capacity of an iron chelator against infection in fish, becoming a potential effective host-directed therapy for SRS and other animals against ferrophilic pathogens.
Collapse
Affiliation(s)
- Mario Caruffo
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Santo Tomás, Santiago, Chile.,Laboratorio Inmunología en Peces, Facultad de Ciencia de la Vida, Universidad Andrés Bello, República 239, Santiago, Chile.,Scimetrica Lab, Santiago, Chile
| | - Dinka Mandakovic
- Laboratorio de Genómica y Genética de Interacciones Biológicas (LG2IB), Instituto de Nutrición y Tecnología de los Alimento, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile.,Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile.,GEMA Center for Genomics, Ecology and Environment, Universidad Mayor, Camino La Pirámide 5750, Huechuraba, Santiago, Chile.,Scimetrica Lab, Santiago, Chile
| | - Madelaine Mejías
- Laboratorio de Genómica y Genética de Interacciones Biológicas (LG2IB), Instituto de Nutrición y Tecnología de los Alimento, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile.,Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile
| | - Ignacio Chávez-Báez
- Laboratorio de Genómica y Genética de Interacciones Biológicas (LG2IB), Instituto de Nutrición y Tecnología de los Alimento, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile.,Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile
| | - Pablo Salgado
- Laboratorio de Genómica y Genética de Interacciones Biológicas (LG2IB), Instituto de Nutrición y Tecnología de los Alimento, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile.,Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile.,Laboratorio de Nutrición Animal (LABNA). Facultad de Ciencias Agronómicas, Producción Animal, Universidad de Chile, Santa Rosa, 11315, La Pintana, Chile
| | - Daniela Ortiz
- Laboratorio de Genómica y Genética de Interacciones Biológicas (LG2IB), Instituto de Nutrición y Tecnología de los Alimento, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile.,Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile.,Laboratorio de Nutrición Animal (LABNA). Facultad de Ciencias Agronómicas, Producción Animal, Universidad de Chile, Santa Rosa, 11315, La Pintana, Chile
| | - Liliana Montt
- Laboratorio de Genómica y Genética de Interacciones Biológicas (LG2IB), Instituto de Nutrición y Tecnología de los Alimento, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile.,Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile
| | - Javiera Pérez-Valenzuela
- Laboratorio de Genómica y Genética de Interacciones Biológicas (LG2IB), Instituto de Nutrición y Tecnología de los Alimento, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile.,Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile
| | - Francisca Vera-Tamargo
- Laboratorio de Genómica y Genética de Interacciones Biológicas (LG2IB), Instituto de Nutrición y Tecnología de los Alimento, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile.,Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile
| | - José Manuel Yánez
- Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile.,Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santa Rosa, 11735, La Pintana, Chile
| | - Jurij Wacyk
- Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile.,Laboratorio de Nutrición Animal (LABNA). Facultad de Ciencias Agronómicas, Producción Animal, Universidad de Chile, Santa Rosa, 11315, La Pintana, Chile
| | - Rodrigo Pulgar
- Laboratorio de Genómica y Genética de Interacciones Biológicas (LG2IB), Instituto de Nutrición y Tecnología de los Alimento, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile. .,Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago, Chile. .,Laboratory for Research in Functional Nutrition, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Av. El Líbano 5524, Macul, 7830490, Santiago, Chile. .,Scimetrica Lab, Santiago, Chile.
| |
Collapse
|
9
|
A copper(II)-binding triazole derivative with ionophore properties is active against Candida spp. J Biol Inorg Chem 2020; 25:1117-1128. [DOI: 10.1007/s00775-020-01828-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/05/2020] [Indexed: 01/12/2023]
|
10
|
Roisin L, Melloul E, Woerther PL, Royer G, Decousser JW, Guillot J, Dannaoui E, Botterel F. Modulated Response of Aspergillus fumigatus and Stenotrophomonas maltophilia to Antimicrobial Agents in Polymicrobial Biofilm. Front Cell Infect Microbiol 2020; 10:574028. [PMID: 33123497 PMCID: PMC7573239 DOI: 10.3389/fcimb.2020.574028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction: The complexity of biofilms constitutes a therapeutic challenge and the antimicrobial susceptibility of fungal-bacterial biofilms remains poorly studied. The filamentous fungus Aspergillus fumigatus (Af) and the Gram-negative bacillus Stenotrophomonas maltophilia (Sm) can form biofilms and can be co-isolated from the airways of cystic fibrosis (CF) patients. We previously developed an in vitro biofilm model which highlighted the antibiosis effect of Sm on Af, which was dependent on the bacterial fitness. The aim of the present study was to investigate the in vitro susceptibility of Af and Sm in mono- or polymicrobial biofilms to five antimicrobial agents alone and in two-drug combinations. Methods: Af and Sm clinical reference strains and two strains from CF sputa were tested through a planktonic and biofilm approaches. Af, Sm, or Af-Sm susceptibilities to amphotericin B (AMB), itraconazole (ITC), voriconazole (VRC), levofloxacin (LVX), and rifampicin (RFN) were evaluated by conventional planktonic techniques, crystal violet, XTT, qPCR, and viable plate count. Results: Af planktonic cells and biofilms in formation were more susceptible to AMB, ITC, and VRC than Af mature biofilms. Af mature biofilms were susceptible to AMB, but not to ITC and VRC. Based on viable plate count, a lower concentration of LVX and RFN was required to reduce Sm cell numbers on biofilms in formation compared with mature biofilms. The antibiosis effect of Sm on Af growth was more pronounced for the association of CF strains that exhibited a higher fitness than the reference strains. In Af-Sm biofilms, the fungal susceptibility to AMB was increased compared with Af biofilms. In contrast, the bacterial susceptibility to LVX decreased in Af-Sm biofilms and was fungal biomass-dependent. The combination of AMB (64 μg/mL) with LVX or RFN (4 μg/mL) was efficient to impair Af and Sm growth in the polymicrobial biofilm. Conclusion: Sm increased the Af susceptibility to AMB, whereas Af protected Sm from LVX. Interactions between Af and Sm within biofilms modulate susceptibility to antimicrobial agents, opening the way to new antimicrobial strategies in CF patients.
Collapse
Affiliation(s)
- Lolita Roisin
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France
| | - Elise Melloul
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France
| | - Paul-Louis Woerther
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Bactériologie-Hygiène, Département de prévention, diagnostic et traitement des infections, Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Guilhem Royer
- Unité de Bactériologie-Hygiène, Département de prévention, diagnostic et traitement des infections, Hôpital Henri Mondor, AP-HP, Créteil, France.,LABGeM, Génomique Métabolique, CEA, Genoscope, Institut François Jacob, Université d'Evry, Université Paris-Saclay, CNRS, Evry, France
| | - Jean-Winoc Decousser
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Bactériologie-Hygiène, Département de prévention, diagnostic et traitement des infections, Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Jacques Guillot
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Parasitologie-Mycologie, Ecole nationale vétérinaire d'Alfort, Maisons-Alfort, France
| | - Eric Dannaoui
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Parasitologie-Mycologie, Service de Microbiologie, Hôpital Européen Georges Pompidou, AP-HP, Université Paris-Descartes, Paris, France
| | - Françoise Botterel
- EA 7380 Dynamyc, Université Paris-Est Créteil, Ecole nationale vétérinaire d'Alfort, USC Anses, Créteil, France.,Unité de Parasitologie-Mycologie, Département de prévention, diagnostic et traitement des infections, Hôpital Henri Mondor, AP-HP, Créteil, France
| |
Collapse
|
11
|
Brilhante RSN, Costa ADC, Pereira VS, Fernandes MR, de Oliveira JS, Rodrigues AM, Camargo ZP, Pereira-Neto WDA, Sidrim JJC, Rocha MFG. Antifungal activity of deferiprone and EDTA against Sporothrix spp.: Effect on planktonic growth and biofilm formation. Med Mycol 2020; 59:myaa073. [PMID: 32838409 DOI: 10.1093/mmy/myaa073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/28/2020] [Accepted: 08/18/2020] [Indexed: 11/14/2022] Open
Abstract
The present study evaluated the antifungal activity of the chelators deferiprone (DFP) and ethylenediaminetetraacetic acid (EDTA) and their effect on biofilm formation of the S. schenckii complex. Eighteen strains of Sporothrix spp. (seven S. brasiliensis, three S. globosa, three S. mexicana and five Sporothrix schenckii sensu stricto) were used. Minimum inhibitory concentration (MIC) values for EDTA and DFP against filamentous forms of Sporothrix spp. ranged from 32 to 128 μg/ml. For antifungal drugs, MIC values ranged from 0.25 to 4 μg/ml for amphotericin B, from 0.25 to 4 μg/ml for itraconazole, and from 0.03 to 0.25 μg/ml for terbinafine. The chelators caused inhibition of Sporothrix spp. in yeast form at concentrations ranging from 16 to 64 μg/ml (for EDTA) and 8 to 32 μg/ml (for DFP). For antifungal drugs, MIC values observed against the yeast varied from 0.03 to 0.5 μg/ml for AMB, 0.03 to 1 μg/ml for ITC, and 0.03 to 0.13 μg/ml for TRB. Both DFP and EDTA presented synergistic interaction with antifungals against Sporothrix spp. in both filamentous and yeast form. Biofilms formed in the presence of the chelators (512 μg/ml) showed a reduction of 47% in biomass and 45% in metabolic activity. Our data reveal that DFP and EDTA reduced the growth of planktonic cells of Sporothrix spp., had synergistic interaction with antifungal drugs against this pathogen, and reduced biofilm formation of Sporothrix spp. LAY SUMMARY Our data reveal that iron chelators deferiprone and ethylenediaminetetraacetic acid reduced the growth of planktonic cells of Sporothrix spp. as well as had synergistic interaction with antifungal drugs against this pathogen and reduced biofilm formation of Sporothrix spp.
Collapse
Affiliation(s)
- Raimunda Sâmia Nogueira Brilhante
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Anderson da Cunha Costa
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Vandbergue Santos Pereira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Mirele Rodrigues Fernandes
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Jonathas Sales de Oliveira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Anderson Messias Rodrigues
- Cellular Biology Division, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo. Rua Botucatu, 862, 6th floor, Medical Sciences Building, CEP: 04023-062, São Paulo, São Paulo, Brazil
| | - Zoilo Pires Camargo
- Cellular Biology Division, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo. Rua Botucatu, 862, 6th floor, Medical Sciences Building, CEP: 04023-062, São Paulo, São Paulo, Brazil
| | - Waldemiro de Aquino Pereira-Neto
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - José Júlio Costa Sidrim
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Marcos Fábio Gadelha Rocha
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
- Postgraduate Program in Veterinary Sciences, College of Veterinary Medicine, State University of Ceará. Av. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
| |
Collapse
|
12
|
Lai YW, Pang CNI, Campbell LT, Chen SCA, Wilkins MR, Carter DA. Different Pathways Mediate Amphotericin-Lactoferrin Drug Synergy in Cryptococcus and Saccharomyces. Front Microbiol 2019; 10:2195. [PMID: 31632362 PMCID: PMC6779777 DOI: 10.3389/fmicb.2019.02195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/06/2019] [Indexed: 12/27/2022] Open
Abstract
Fungal infections are an increasing cause of morbidity and mortality. Current antifungal drugs are limited in spectrum, few new drugs are in development, and resistance is an increasing issue. Drug synergy can enhance available drugs and extend their lifetime, however, few synergistic combinations are in clinical use and mechanistic data on how combinations work is lacking. The multifunctional glycoprotein lactoferrin (LF) acts synergistically with amphotericin B (AMB) in a range of fungal species. Whole LF binds and sequesters iron, and LF can also be digested enzymatically to produce cationic peptides with distinct antimicrobial functions. To understand how LF synergizes AMB, we previously undertook a transcriptomic analysis in Saccharomyces and found a paradoxical down-regulation of iron and stress response, suggesting stress pathway interference was dysregulating an appropriate response, resulting in cell death. To extend this to a fungal pathogen, we here perform the same analysis in Cryptococcus neoformans. While both fungi responded to AMB in a similar way, the addition of LF produced remarkably contrasting results, with the Cryptococcus transcriptome enriched for processes relating to cellular stress, up-regulation of endoplasmic-reticulum-associated protein degradation (ERAD), stress granule disassembly and protein folding, endoplasmic reticulum-Golgi-vacuole trafficking and autophagy, suggesting an overall disruption of protein and lipid biosynthesis. These studies demonstrate that the mechanism of LF-mediated synergy is species-specific, possibly due to differences in the way LF peptides are generated, bind to and enter cells and act on intracellular targets, illustrating how very different cellular processes can underlie what appears to be a similar phenotypic response.
Collapse
Affiliation(s)
- Yu-Wen Lai
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Chi Nam Ignatius Pang
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Kensington, NSW, Australia
| | - Leona T Campbell
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Sharon C A Chen
- Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Sydney, NSW, Australia.,Centre for Infectious Diseases and Microbiology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Sydney Medical School, The University of Sydney, Westmead, NSW, Australia
| | - Marc R Wilkins
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Kensington, NSW, Australia
| | - Dee A Carter
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia.,Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Iron Restriction to Clinical Isolates of Candida albicans by the Novel Chelator DIBI Inhibits Growth and Increases Sensitivity to Azoles In Vitro and In Vivo in a Murine Model of Experimental Vaginitis. Antimicrob Agents Chemother 2018; 62:AAC.02576-17. [PMID: 29844048 DOI: 10.1128/aac.02576-17] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 05/19/2018] [Indexed: 12/22/2022] Open
Abstract
Candida albicans is an important opportunistic pathogen causing various human infections that are often treated with azole antifungals. The U.S. CDC now regards developing candidal antifungal resistance as a threat, creating a need for new and more effective antifungal treatments. Iron is an essential nutrient for all living cells, and there is growing evidence that interference with iron homeostasis of C. albicans can improve its response to antifungals. This study was aimed at establishing whether withholding iron by currently used medical iron chelators and the novel chelator DIBI could restrict growth and also enhance the activity of azoles against clinical isolates of C. albicans DIBI, but not deferoxamine or deferiprone, inhibited the growth of C. albicans at relatively low concentrations in vitro, and this inhibition was reversed by iron addition. DIBI in combination with various azoles demonstrated stronger growth inhibition than the azoles alone and greatly prolonged the inhibition of cell multiplication. In addition, the administration of DIBI along with fluconazole (FLC) to mice inoculated with an FLC-sensitive isolate in a model of experimental C. albicans vaginitis showed a markedly improved clearance of infection. These results suggest that iron chelation by DIBI has the potential to enhance azole efficacy for the treatment of candidiasis.
Collapse
|
14
|
Hsu JL, Manouvakhova OV, Clemons KV, Inayathullah M, Tu AB, Sobel RA, Tian A, Nazik H, Pothineni VR, Pasupneti S, Jiang X, Dhillon GS, Bedi H, Rajadas J, Haas H, Aurelian L, Stevens DA, Nicolls MR. Microhemorrhage-associated tissue iron enhances the risk for Aspergillus fumigatus invasion in a mouse model of airway transplantation. Sci Transl Med 2018; 10:10/429/eaag2616. [PMID: 29467298 PMCID: PMC5841257 DOI: 10.1126/scitranslmed.aag2616] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 02/23/2017] [Accepted: 09/26/2017] [Indexed: 01/25/2023]
Abstract
Invasive pulmonary disease due to the mold Aspergillus fumigatus can be life-threatening in lung transplant recipients, but the risk factors remain poorly understood. To study this process, we used a tracheal allograft mouse model that recapitulates large airway changes observed in patients undergoing lung transplantation. We report that microhemorrhage-related iron content may be a major determinant of A. fumigatus invasion and, consequently, its virulence. Invasive growth was increased during progressive alloimmune-mediated graft rejection associated with high concentrations of ferric iron in the graft. The role of iron in A. fumigatus invasive growth was further confirmed by showing that this invasive phenotype was increased in tracheal transplants from donor mice lacking the hemochromatosis gene (Hfe-/- ). The invasive phenotype was also increased in mouse syngrafts treated with topical iron solution and in allograft recipients receiving deferoxamine, a chelator that increases iron bioavailability to the mold. The invasive growth of the iron-intolerant A. fumigatus double-knockout mutant (ΔsreA/ΔcccA) was lower than that of the wild-type mold. Alloimmune-mediated microvascular damage and iron overload did not appear to impair the host's immune response. In human lung transplant recipients, positive staining for iron in lung transplant tissue was more commonly seen in endobronchial biopsy sections from transplanted airways than in biopsies from the patients' own airways. Collectively, these data identify iron as a major determinant of A. fumigatus invasive growth and a potential target to treat or prevent A. fumigatus infections in lung transplant patients.
Collapse
Affiliation(s)
- Joe L. Hsu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA,Veterans Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA 94304, USA
| | - Olga V. Manouvakhova
- Veterans Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA 94304, USA
| | - Karl V. Clemons
- Infectious Diseases Research Laboratory, California Institute for Medical Research, San Jose, CA 95128, USA,Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammed Inayathullah
- Biomaterials and Advanced Drug Delivery Laboratory, Cardiovascular Pharmacology Division, Cardio-vascular Institute, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Allen B. Tu
- Veterans Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA 94304, USA
| | - Raymond A. Sobel
- Veterans Affairs Palo Alto Health Care System, Pathology and Laboratory Service, Palo Alto, CA 94304, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amy Tian
- Veterans Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA 94304, USA
| | - Hasan Nazik
- Infectious Diseases Research Laboratory, California Institute for Medical Research, San Jose, CA 95128, USA,Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Medical Microbiology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Venkata R. Pothineni
- Biomaterials and Advanced Drug Delivery Laboratory, Cardiovascular Pharmacology Division, Cardio-vascular Institute, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Shravani Pasupneti
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA,Veterans Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA 94304, USA
| | - Xinguo Jiang
- Veterans Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA 94304, USA
| | - Gundeep S. Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Harmeet Bedi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jayakumar Rajadas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA,Biomaterials and Advanced Drug Delivery Laboratory, Cardiovascular Pharmacology Division, Cardio-vascular Institute, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Hubertus Haas
- Division of Molecular Biology, Medical University Innsbruck, Innsbruck, Austria
| | - Laure Aurelian
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - David A. Stevens
- Infectious Diseases Research Laboratory, California Institute for Medical Research, San Jose, CA 95128, USA,Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark R. Nicolls
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA,Veterans Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA 94304, USA,Corresponding author.
| |
Collapse
|
15
|
Bajtai A, Fekete B, Palkó M, Fülöp F, Lindner W, Kohout M, Ilisz I, Péter A. Comparative study on the liquid chromatographic enantioseparation of cyclic β-amino acids and the related cyclic β-aminohydroxamic acids on Cinchona
alkaloid-based zwitterionic chiral stationary phases. J Sep Sci 2017; 41:1216-1223. [DOI: 10.1002/jssc.201701190] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Attila Bajtai
- Department of Inorganic and Analytical Chemistry; University of Szeged; Szeged Hungary
| | - Beáta Fekete
- Institute of Pharmaceutical Chemistry; University of Szeged; Szeged Hungary
| | - Márta Palkó
- Institute of Pharmaceutical Chemistry; University of Szeged; Szeged Hungary
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry; University of Szeged; Szeged Hungary
| | - Wolfgang Lindner
- Department of Analytical Chemistry; University of Vienna; Vienna Austria
| | - Michal Kohout
- Department of Organic Chemistry; University of Chemistry and Technology Prague; Prague Czech Republic
| | - István Ilisz
- Department of Inorganic and Analytical Chemistry; University of Szeged; Szeged Hungary
| | - Antal Péter
- Department of Inorganic and Analytical Chemistry; University of Szeged; Szeged Hungary
| |
Collapse
|
16
|
Identification of membrane proteome of Paracoccidioides lutzii and its regulation by zinc. Future Sci OA 2017; 3:FSO232. [PMID: 29134119 PMCID: PMC5676091 DOI: 10.4155/fsoa-2017-0044] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/21/2017] [Indexed: 01/09/2023] Open
Abstract
Aim: During infection development in the host, Paracoccidioides spp. faces the deprivation of micronutrients, a mechanism called nutritional immunity. This condition induces the remodeling of proteins present in different metabolic pathways. Therefore, we attempted to identify membrane proteins and their regulation by zinc in Paracoccidioides lutzii. Materials & methods: Membranes enriched fraction of yeast cells of P. lutzii were isolated, purified and identified by 2D LC–MS/MS detection and database search. Results & conclusion: Zinc deprivation suppressed the expression of membrane proteins such as glycoproteins, those involved in cell wall synthesis and those related to oxidative phosphorylation. This is the first study describing membrane proteins and the effect of zinc deficiency in their regulation in one member of the genus Paracoccidioides. The methodology of protein identification allows the characterization of biological processes performed by those molecules. Therefore, we performed a membrane proteomic analysis of Paracoccidioides lutzii and further evaluated the responses of the fungus to zinc deprivation. The results obtained in the work allowed the characterization of membrane proteins present in organelles that are related to different cellular functions. Zinc deprivation changes processes related to cellular physiology and metabolism. These results help us to understand the process of pathogen–host interaction, since zinc deprivation is a condition present during infection.
Collapse
|
17
|
Desoubeaux G, Piqueras MDC, Pantin A, Bhattacharya SK, Peschke R, Joachim A, Cray C. Application of mass spectrometry to elucidate the pathophysiology of Encephalitozoon cuniculi infection in rabbits. PLoS One 2017; 12:e0177961. [PMID: 28723944 PMCID: PMC5516978 DOI: 10.1371/journal.pone.0177961] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/05/2017] [Indexed: 11/30/2022] Open
Abstract
Encephalitozoon cuniculi is a microsporidian species which can induce subclinical to serious disease in mammals including rabbits, a definitive natural host. The pathophysiology of infection has not been comprehensively elucidated. In this exploratory study, we utilized two mass spectrometry approaches: first, the analysis of the humoral response by profiling the microsporidian antigens as revealed by Western blot screening, and second, implementing the iTRAQ®-labeling protocol to focus on the changes within the host proteome during infection. Seven E. cuniculi proteins were identified at one-dimensional gel regions where specific seropositive reaction was observed by Western blot, including polar tube protein 3, polar tube protein 2, and for the first time reported: heat shock related 70kDa protein, polysaccharide deacetylase domain-containing protein, zinc finger protein, spore wall and anchoring disk complex protein EnP1, and translation elongation factor 1 alpha. In addition, there was a significant increase of nine host proteins in blood samples from E. cuniculi-diseased rabbits in comparison with non-diseased control subjects undergoing various inflammatory processes. This included serum paraoxonase, alpha-1-antiproteinase F precursor and alpha-1-antiproteinase S-1 which have presumptive catalytic activity likely related to infection control, and cystatin fetuin-B-type, an enzyme regulator that has been poorly studied to date. Notably, 11 proteins were found to be statistically increased in rabbits with neurological versus renal clinical presentation of E. cuniculi infection. Overall, this novel analysis based on mass spectrometry has provided new insights on the inflammatory and humoral responses during E. cuniculi infection in rabbits.
Collapse
Affiliation(s)
- Guillaume Desoubeaux
- University of Miami - Miller School of Medicine, Division of Comparative Pathology, Department of Pathology & Laboratory Medicine, Miami, Florida, United States of America
- CHU de Tours, Service de Parasitologie – Mycologie – Médecine tropicale, Tours, France
- Université François-Rabelais, Faculté de Médecine, CEPR - INSERM U1100 / Équipe 3, Tours, France
| | - Maria del Carmen Piqueras
- University of Miami, Mass Spectrometry Core Facility, Miller School of Medicin–, Miami, Florida, United States of America
| | - Ana Pantin
- University of Miami - Miller School of Medicine, Division of Comparative Pathology, Department of Pathology & Laboratory Medicine, Miami, Florida, United States of America
| | - Sanjoy K. Bhattacharya
- University of Miami, Mass Spectrometry Core Facility, Miller School of Medicin–, Miami, Florida, United States of America
| | - Roman Peschke
- University of Veterinary Medicine, Institute of Parasitology, Department of Pathobiology, Vienna, Austria
| | - Anja Joachim
- University of Veterinary Medicine, Institute of Parasitology, Department of Pathobiology, Vienna, Austria
| | - Carolyn Cray
- University of Miami - Miller School of Medicine, Division of Comparative Pathology, Department of Pathology & Laboratory Medicine, Miami, Florida, United States of America
| |
Collapse
|
18
|
Fekete B, Palkó M, Haukka M, Fülöp F. Synthesis of Pyrrolo[1,2-a]pyrimidine Enantiomers via Domino Ring-Closure followed by Retro Diels-Alder Protocol. Molecules 2017; 22:molecules22040613. [PMID: 28406463 PMCID: PMC6154686 DOI: 10.3390/molecules22040613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 01/14/2023] Open
Abstract
From 2-aminonorbornene hydroxamic acids, a simple and efficient method for the preparation of pyrrolo[1,2-a]pyrimidine enantiomers is reported. The synthesis is based on domino ring-closure followed by microwave-induced retro Diels-Alder (RDA) protocols, where the chirality of the desired products is transferred from norbornene derivatives. The stereochemistry of the synthesized compounds was proven by X-ray crystallography. The absolute configuration of the product is determined by the configuration of the starting amino hydroxamic acid.
Collapse
Affiliation(s)
- Beáta Fekete
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös utca 6, Szeged H-6720, Hungary.
| | - Márta Palkó
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös utca 6, Szeged H-6720, Hungary.
| | - Matti Haukka
- Department of Chemistry, University of Jyväskylä, FIN-40014 Turku, Finland.
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös utca 6, Szeged H-6720, Hungary.
- MTA-SZTE Stereochemistry Research Group, Hungarian Academy of Sciences, Eötvös utca 6, Szeged H-6720, Hungary.
| |
Collapse
|
19
|
Drago-Serrano ME, Campos-Rodríguez R, Carrero JC, de la Garza M. Lactoferrin: Balancing Ups and Downs of Inflammation Due to Microbial Infections. Int J Mol Sci 2017; 18:E501. [PMID: 28257033 PMCID: PMC5372517 DOI: 10.3390/ijms18030501] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 02/13/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Lactoferrin (Lf) is a glycoprotein of the primary innate immune-defense system of mammals present in milk and other mucosal secretions. This protein of the transferrin family has broad antimicrobial properties by depriving pathogens from iron, or disrupting their plasma membranes through its highly cationic charge. Noteworthy, Lf also exhibits immunomodulatory activities performing up- and down-regulation of innate and adaptive immune cells, contributing to the homeostasis in mucosal surfaces exposed to myriad of microbial agents, such as the gastrointestinal and respiratory tracts. Although the inflammatory process is essential for the control of invasive infectious agents, the development of an exacerbated or chronic inflammation results in tissue damage with life-threatening consequences. In this review, we highlight recent findings in in vitro and in vivo models of the gut, lung, oral cavity, mammary gland, and liver infections that provide experimental evidence supporting the therapeutic role of human and bovine Lf in promoting some parameters of inflammation and protecting against the deleterious effects of bacterial, viral, fungal and protozoan-associated inflammation. Thus, this new knowledge of Lf immunomodulation paves the way to more effective design of treatments that include native or synthetic Lf derivatives, which may be useful to reduce immune-mediated tissue damage in infectious diseases.
Collapse
Affiliation(s)
- Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco (UAM-X), CdMx 04960, Mexico.
| | - Rafael Campos-Rodríguez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional (ESM-IPN), CdMx 11340, Mexico.
| | - Julio César Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (IIB-UNAM), CdMx 70228, Mexico.
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), CdMx 07360, Mexico.
| |
Collapse
|
20
|
Pang CNI, Lai YW, Campbell LT, Chen SCA, Carter DA, Wilkins MR. Transcriptome and network analyses in Saccharomyces cerevisiae reveal that amphotericin B and lactoferrin synergy disrupt metal homeostasis and stress response. Sci Rep 2017; 7:40232. [PMID: 28079179 PMCID: PMC5228129 DOI: 10.1038/srep40232] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 12/02/2016] [Indexed: 12/16/2022] Open
Abstract
Invasive fungal infections are difficult to treat. The few available antifungal drugs have problems with toxicity or efficacy, and resistance is increasing. To overcome these challenges, existing therapies may be enhanced by synergistic combination with another agent. Previously, we found amphotericin B (AMB) and the iron chelator, lactoferrin (LF), were synergistic against a range of different fungal pathogens. This study investigates the mechanism of AMB-LF synergy, using RNA-seq and network analyses. AMB treatment resulted in increased expression of genes involved in iron homeostasis and ATP synthesis. Unexpectedly, AMB-LF treatment did not lead to increased expression of iron and zinc homeostasis genes. However, genes involved in adaptive response to zinc deficiency and oxidative stress had decreased expression. The clustering of co-expressed genes and network analysis revealed that many iron and zinc homeostasis genes are targets of transcription factors Aft1p and Zap1p. The aft1Δ and zap1Δ mutants were hypersensitive to AMB and H2O2, suggesting they are key regulators of the drug response. Mechanistically, AMB-LF synergy could involve AMB affecting the integrity of the cell wall and membrane, permitting LF to disrupt intracellular processes. We suggest that Zap1p- and Aft1p-binding molecules could be combined with existing antifungals to serve as synergistic treatments.
Collapse
Affiliation(s)
- Chi Nam Ignatius Pang
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Kensington, New South Wales, Australia
| | - Yu-Wen Lai
- School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Leona T Campbell
- School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Sharon C-A Chen
- Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia.,Centre for Infectious Diseases and Microbiology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Sydney Medical School, University of Sydney, Westmead, NSW, Australia
| | - Dee A Carter
- School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia.,Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
| | - Marc R Wilkins
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Kensington, New South Wales, Australia
| |
Collapse
|
21
|
Balhara M, Chaudhary R, Ruhil S, Singh B, Dahiya N, Parmar VS, Jaiwal PK, Chhillar AK. Siderophores; iron scavengers: the novel & promising targets for pathogen specific antifungal therapy. Expert Opin Ther Targets 2016; 20:1477-1489. [PMID: 27797604 DOI: 10.1080/14728222.2016.1254196] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The recent emergence of resistance, toxicity paradigm and limited efficacy of conventional antifungal drugs necessitate the identification of de novo targets in fungal metabolism. One of the most critical physiological processes during in vivo pathogenesis is maintenance of iron homeostasis. The most life threatening opportunistic human fungal pathogens like Aspergillus, Candida and Cryptococcus exploit the siderophore mediated iron uptake mechanism either for survival, virulence, propagation or resistance to oxidative stress envisaged in vivo during infection. Areas covered: In this review, we will highlight the metabolic pathways; specifically siderophore biosynthesis, uptake and utilisation, triggered in the fungal pathogens in iron starving conditions and the various putative targets viable in these pathways to be recruited as novel therapeutic antidotes either via biosynthetic enzymes catalytic site inhibitors or as drug conjugates through trojan horse approach and further role in the development of fungal specific reliable diagnostic markers. Expert opinion: Siderophores are the weapons released by a pathogen to conquer the battle for iron acquisition. Hence, the fungal siderophore biosynthetic pathways along with their uptake and utilisation mechanisms represent an ideal target for pathogen specific, host friendly therapeutic strategy which would block the proliferation of parasite without causing any harm to the mammalian host.
Collapse
Affiliation(s)
- Meenakshi Balhara
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Renu Chaudhary
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Sonam Ruhil
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Bharat Singh
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Nisha Dahiya
- b Division of Epidemiology and Communicable Diseases , Indian Council of Medical Research , Delhi , India
| | - Virinder S Parmar
- c Bioorganic Laboratory, Department of Chemistry , University of Delhi , Delhi , India
| | - Pawan K Jaiwal
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| | - Anil K Chhillar
- a Centre for Biotechnology , Maharshi Dayanand University , Rohtak , Haryana , India
| |
Collapse
|
22
|
Administration of Zinc Chelators Improves Survival of Mice Infected with Aspergillus fumigatus both in Monotherapy and in Combination with Caspofungin. Antimicrob Agents Chemother 2016; 60:5631-9. [PMID: 27401578 DOI: 10.1128/aac.00324-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 07/01/2016] [Indexed: 12/19/2022] Open
Abstract
Aspergillus fumigatus can infect immunocompromised patients, leading to high mortality rates due to the lack of reliable treatment options. This pathogen requires uptake of zinc from host tissues in order to successfully grow and cause virulence. Reducing the availability of that micronutrient could help treat A. fumigatus infections. In this study, we examined the in vitro effects of seven chelators using a bioluminescent strain of A. fumigatus 1,10-Phenanthroline and N,N,N',N'-tetrakis(2-pyridylmethyl)ethane-1,2-diamine (TPEN) proved to be the chelators most effective at inhibiting fungal growth. Intraperitoneal administration of either phenanthroline or TPEN resulted in a significant improvement in survival and decrease of weight loss and fungal burden for immunosuppressed mice intranasally infected with A. fumigatus In vitro both chelators had an indifferent effect when employed in combination with caspofungin. The use of TPEN in combination with caspofungin also significantly increased survival compared to that when using these drugs individually. Our results suggest that zinc chelation may be a valid strategy for dealing with A. fumigatus infections and that both phenanthroline and TPEN could potentially be used either independently or in combination with caspofungin, indicating that their use in combination with other antifungal treatments might also be applicable.
Collapse
|
23
|
Lai YW, Campbell LT, Wilkins MR, Pang CNI, Chen S, Carter DA. Synergy and antagonism between iron chelators and antifungal drugs in Cryptococcus. Int J Antimicrob Agents 2016; 48:388-94. [PMID: 27474467 DOI: 10.1016/j.ijantimicag.2016.06.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/02/2016] [Accepted: 06/10/2016] [Indexed: 12/27/2022]
Abstract
Fungal infections remain very difficult to treat, and developing new antifungal drugs is difficult and expensive. Recent approaches therefore seek to augment existing antifungals with synergistic agents that can lower the therapeutic dose, increase efficacy and prevent resistance from developing. Iron limitation can inhibit microbial growth, and iron chelators have been employed to treat fungal infections. In this study, chequerboard testing was used to explore combinations of iron chelators with antifungal agents against pathogenic Cryptococcus spp. with the aim of determining how disruption to iron homeostasis affects antifungal susceptibility. The iron chelators ethylenediaminetetraacetic acid (EDTA), deferoxamine (DFO), deferiprone (DFP), deferasirox (DSX), ciclopirox olamine and lactoferrin (LF) were paired with the antifungal agents amphotericin B (AmB), fluconazole, itraconazole, voriconazole and caspofungin. All chelators except for DFO increased the efficacy of AmB, and significant synergy was seen between AmB and LF for all Cryptococcus strains. Addition of exogenous iron rescued cells from the antifungal effect of LF alone but could not prevent inhibition by AmB + LF, indicating that synergy was not due primarily to iron chelation but to other properties of LF that were potentiated in the presence of AmB. Significant synergy was not seen consistently for other antifungal-chelator combinations, and EDTA, DSX and DFP antagonised the activity of azole drugs in strains of Cryptococcus neoformans var. grubii. This study highlights the range of interactions that can be induced by chelators and indicates that most antifungal drugs are not enhanced by iron limitation in Cryptococcus.
Collapse
Affiliation(s)
- Yu-Wen Lai
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Leona T Campbell
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Marc R Wilkins
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW, Australia
| | - Chi Nam Ignatius Pang
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW, Australia
| | - Sharon Chen
- Centre for Infectious Diseases and Microbiology Laboratory Services, ICPMR, Westmead Hospital and the University of Sydney, Sydney, NSW, Australia
| | - Dee A Carter
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
24
|
Fekete B, Palkó M, Mándity I, Haukka M, Fülöp F. A Domino Ring-Closure Followed by Retro-Diels-Alder Reaction for the Preparation of Pyrimido[2,1-a]isoindole Enantiomers. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600434] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Beáta Fekete
- Institute of Pharmaceutical Chemistry; University of Szeged; Eötvös utca 6 6720 Szeged Hungary
| | - Márta Palkó
- Institute of Pharmaceutical Chemistry; University of Szeged; Eötvös utca 6 6720 Szeged Hungary
| | - István Mándity
- Institute of Pharmaceutical Chemistry; University of Szeged; Eötvös utca 6 6720 Szeged Hungary
| | - Matti Haukka
- Department of Chemistry; University of Jyväskylä; 40014 Turku Finland
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry; University of Szeged; Eötvös utca 6 6720 Szeged Hungary
- MTA-SZTE Stereochemistry Research Group; Hungarian Academy of Sciences; Eötvös utca 6 6720 Szeged Hungary
| |
Collapse
|
25
|
High-performance liquid chromatographic enantioseparation of cyclic β-aminohydroxamic acids on zwitterionic chiral stationary phases based on Cinchona alkaloids. Anal Chim Acta 2016; 921:84-94. [DOI: 10.1016/j.aca.2016.03.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 03/22/2016] [Accepted: 03/26/2016] [Indexed: 11/24/2022]
|
26
|
Nazik H, Penner JC, Ferreira JA, Haagensen JAJ, Cohen K, Spormann AM, Martinez M, Chen V, Hsu JL, Clemons KV, Stevens DA. Effects of Iron Chelators on the Formation and Development of Aspergillus fumigatus Biofilm. Antimicrob Agents Chemother 2015; 59:6514-20. [PMID: 26239975 PMCID: PMC4576070 DOI: 10.1128/aac.01684-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 07/30/2015] [Indexed: 12/25/2022] Open
Abstract
Iron acquisition is crucial for the growth of Aspergillus fumigatus. A. fumigatus biofilm formation occurs in vitro and in vivo and is associated with physiological changes. In this study, we assessed the effects of Fe chelators on biofilm formation and development. Deferiprone (DFP), deferasirox (DFS), and deferoxamine (DFM) were tested for MIC against a reference isolate via a broth macrodilution method. The metabolic effects (assessed by XTT [2,3-bis[2-methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxanilide inner salt]) on biofilm formation by conidia were studied upon exposure to DFP, DFM, DFP plus FeCl3, or FeCl3 alone. A preformed biofilm was exposed to DFP with or without FeCl3. The DFP and DFS MIC50 against planktonic A. fumigatus was 1,250 μM, and XTT gave the same result. DFM showed no planktonic inhibition at concentrations of ≤2,500 μM. By XTT testing, DFM concentrations of <1,250 μM had no effect, whereas DFP at 2,500 μM increased biofilms forming in A. fumigatus or preformed biofilms (P < 0.01). DFP at 156 to 2,500 μM inhibited biofilm formation (P < 0.01 to 0.001) in a dose-responsive manner. Biofilm formation with 625 μM DFP plus any concentration of FeCl3 was lower than that in the controls (P < 0.05 to 0.001). FeCl3 at ≥625 μM reversed the DFP inhibitory effect (P < 0.05 to 0.01), but the reversal was incomplete compared to the controls (P < 0.05 to 0.01). For preformed biofilms, DFP in the range of ≥625 to 1,250 μM was inhibitory compared to the controls (P < 0.01 to 0.001). FeCl3 at ≥625 μM overcame inhibition by 625 μM DFP (P < 0.001). FeCl3 alone at ≥156 μM stimulated biofilm formation (P < 0.05 to 0.001). Preformed A. fumigatus biofilm increased with 2,500 μM FeCl3 only (P < 0.05). In a strain survey, various susceptibilities of biofilms of A. fumigatus clinical isolates to DFP were noted. In conclusion, iron stimulates biofilm formation and preformed biofilms. Chelators can inhibit or enhance biofilms. Chelation may be a potential therapy for A. fumigatus, but we show here that chelators must be chosen carefully. Individual isolate susceptibility assessments may be needed.
Collapse
Affiliation(s)
- Hasan Nazik
- California Institute for Medical Research, San Jose, California, USA Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, California, USA Department of Medical Microbiology, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - John C Penner
- California Institute for Medical Research, San Jose, California, USA
| | - Jose A Ferreira
- California Institute for Medical Research, San Jose, California, USA Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, California, USA
| | - Janus A J Haagensen
- Department of Civil and Environmental Engineering, Stanford University, Stanford, California, USA
| | - Kevin Cohen
- California Institute for Medical Research, San Jose, California, USA
| | - Alfred M Spormann
- Department of Civil and Environmental Engineering, Stanford University, Stanford, California, USA
| | - Marife Martinez
- California Institute for Medical Research, San Jose, California, USA
| | - Vicky Chen
- California Institute for Medical Research, San Jose, California, USA
| | - Joe L Hsu
- California Institute for Medical Research, San Jose, California, USA Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Karl V Clemons
- California Institute for Medical Research, San Jose, California, USA Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, California, USA
| | - David A Stevens
- California Institute for Medical Research, San Jose, California, USA Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
27
|
Vicentefranqueira R, Amich J, Laskaris P, Ibrahim-Granet O, Latgé JP, Toledo H, Leal F, Calera JA. Targeting zinc homeostasis to combat Aspergillus fumigatus infections. Front Microbiol 2015; 6:160. [PMID: 25774155 PMCID: PMC4343018 DOI: 10.3389/fmicb.2015.00160] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 02/11/2015] [Indexed: 11/13/2022] Open
Abstract
Aspergillus fumigatus is able to invade and grow in the lungs of immunosuppressed individuals and causes invasive pulmonary aspergillosis. The concentration of free zinc in living tissues is much lower than that required for optimal fungal growth in vitro because most of it is tightly bound to proteins. To obtain efficiently zinc from a living host A. fumigatus uses the zinc transporters ZrfA, ZrfB, and ZrfC. The ZafA transcriptional regulator induces the expression of all these transporters and is essential for virulence. Thus, ZafA could be targeted therapeutically to inhibit fungal growth. The ZrfC transporter plays the major role in zinc acquisition from the host whereas ZrfA and ZrfB rather have a supplementary role to that of ZrfC. In addition, only ZrfC enables A. fumigatus to overcome the inhibitory effect of calprotectin, which is an antimicrobial Zn/Mn-chelating protein synthesized and released by neutrophils within the fungal abscesses of immunosuppressed non-leucopenic animals. Hence, fungal survival in these animals would be undermined upon blocking therapeutically the function of ZrfC. Therefore, both ZafA and ZrfC have emerged as promising targets for the discovery of new antifungals to treat Aspergillus infections.
Collapse
Affiliation(s)
- Rocío Vicentefranqueira
- Instituto de Biología Funcional y Genómica, Centro Mixto del Consejo Superior de Investigaciones Científicas y Universidad de Salamanca , Salamanca, Spain ; Departamento de Microbiología y Genética, Universidad de Salamanca , Salamanca, Spain
| | - Jorge Amich
- Instituto de Biología Funcional y Genómica, Centro Mixto del Consejo Superior de Investigaciones Científicas y Universidad de Salamanca , Salamanca, Spain ; Departamento de Microbiología y Genética, Universidad de Salamanca , Salamanca, Spain
| | - Paris Laskaris
- Unité de Recherche Cytokines and Inflammation, Institut Pasteur , Paris, France
| | | | - Jean P Latgé
- Unité des Aspergillus, Institut Pasteur , Paris, France
| | - Héctor Toledo
- Instituto de Biología Funcional y Genómica, Centro Mixto del Consejo Superior de Investigaciones Científicas y Universidad de Salamanca , Salamanca, Spain ; Departamento de Microbiología y Genética, Universidad de Salamanca , Salamanca, Spain
| | - Fernando Leal
- Instituto de Biología Funcional y Genómica, Centro Mixto del Consejo Superior de Investigaciones Científicas y Universidad de Salamanca , Salamanca, Spain ; Departamento de Microbiología y Genética, Universidad de Salamanca , Salamanca, Spain
| | - José A Calera
- Instituto de Biología Funcional y Genómica, Centro Mixto del Consejo Superior de Investigaciones Científicas y Universidad de Salamanca , Salamanca, Spain ; Departamento de Microbiología y Genética, Universidad de Salamanca , Salamanca, Spain
| |
Collapse
|
28
|
|
29
|
da Costa JP, Cova M, Ferreira R, Vitorino R. Antimicrobial peptides: an alternative for innovative medicines? Appl Microbiol Biotechnol 2015; 99:2023-40. [PMID: 25586583 DOI: 10.1007/s00253-015-6375-x] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/26/2014] [Accepted: 12/28/2014] [Indexed: 12/27/2022]
Abstract
Antimicrobial peptides are small molecules with activity against bacteria, yeasts, fungi, viruses, bacteria, and even tumor cells that make these molecules attractive as therapeutic agents. Due to the alarming increase of antimicrobial resistance, interest in alternative antimicrobial agents has led to the exploitation of antimicrobial peptides, both synthetic and from natural sources. Thus, many peptide-based drugs are currently commercially available for the treatment of numerous ailments, such as hepatitis C, myeloma, skin infections, and diabetes. Initial barriers are being increasingly overcome with the development of cost-effective, more stable peptides. Herein, we review the available strategies for their synthesis, bioinformatics tools for the rational design of antimicrobial peptides with enhanced therapeutic indices, hurdles and shortcomings limiting the large-scale production of AMPs, as well as the challenges that the pharmaceutical industry faces on their use as therapeutic agents.
Collapse
Affiliation(s)
- João Pinto da Costa
- Mass Spectrometry Centre, QOPNA, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | | | | | | |
Collapse
|
30
|
Anti-Cancer Iron(II) Complexes of Pentadentate N-Donor Ligands: Cytotoxicity, Transcriptomics Analyses, and Mechanisms of Action. Chemistry 2014; 21:3062-72. [DOI: 10.1002/chem.201404749] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/10/2014] [Indexed: 01/10/2023]
|
31
|
Tøndervik A, Sletta H, Klinkenberg G, Emanuel C, Powell LC, Pritchard MF, Khan S, Craine KM, Onsøyen E, Rye PD, Wright C, Thomas DW, Hill KE. Alginate oligosaccharides inhibit fungal cell growth and potentiate the activity of antifungals against Candida and Aspergillus spp. PLoS One 2014; 9:e112518. [PMID: 25409186 PMCID: PMC4237368 DOI: 10.1371/journal.pone.0112518] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 10/03/2014] [Indexed: 12/24/2022] Open
Abstract
The oligosaccharide OligoG, an alginate derived from seaweed, has been shown to have anti-bacterial and anti-biofilm properties and potentiates the activity of selected antibiotics against multi-drug resistant bacteria. The ability of OligoG to perturb fungal growth and potentiate conventional antifungal agents was evaluated using a range of pathogenic fungal strains. Candida (n = 11) and Aspergillus (n = 3) spp. were tested using germ tube assays, LIVE/DEAD staining, scanning electron microscopy (SEM), atomic force microscopy (AFM) and high-throughput minimum inhibition concentration assays (MICs). In general, the strains tested showed a significant dose-dependent reduction in cell growth at ≥6% OligoG as measured by optical density (OD600; P<0.05). OligoG (>0.5%) also showed a significant inhibitory effect on hyphal growth in germ tube assays, although strain-dependent variations in efficacy were observed (P<0.05). SEM and AFM both showed that OligoG (≥2%) markedly disrupted fungal biofilm formation, both alone, and in combination with fluconazole. Cell surface roughness was also significantly increased by the combination treatment (P<0.001). High-throughput robotic MIC screening demonstrated the potentiating effects of OligoG (2, 6, 10%) with nystatin, amphotericin B, fluconazole, miconazole, voriconazole or terbinafine with the test strains. Potentiating effects were observed for the Aspergillus strains with all six antifungal agents, with an up to 16-fold (nystatin) reduction in MIC. Similarly, all the Candida spp. showed potentiation with nystatin (up to 16-fold) and fluconazole (up to 8-fold). These findings demonstrate the antifungal properties of OligoG and suggest a potential role in the management of fungal infections and possible reduction of antifungal toxicity.
Collapse
Affiliation(s)
- Anne Tøndervik
- Department of Bioprocess Technology, SINTEF Materials and Chemistry, N-7465 Trondheim, Norway
| | - Håvard Sletta
- Department of Bioprocess Technology, SINTEF Materials and Chemistry, N-7465 Trondheim, Norway
- * E-mail:
| | - Geir Klinkenberg
- Department of Bioprocess Technology, SINTEF Materials and Chemistry, N-7465 Trondheim, Norway
| | - Charlotte Emanuel
- Advanced Therapies Group, Tissue Engineering and Reparative Dentistry, Cardiff University School of Dentistry, Cardiff, CF14 4XY, United Kingdom
| | - Lydia C. Powell
- Advanced Therapies Group, Tissue Engineering and Reparative Dentistry, Cardiff University School of Dentistry, Cardiff, CF14 4XY, United Kingdom
| | - Manon F. Pritchard
- Advanced Therapies Group, Tissue Engineering and Reparative Dentistry, Cardiff University School of Dentistry, Cardiff, CF14 4XY, United Kingdom
| | - Saira Khan
- Advanced Therapies Group, Tissue Engineering and Reparative Dentistry, Cardiff University School of Dentistry, Cardiff, CF14 4XY, United Kingdom
| | - Kieron M. Craine
- Advanced Therapies Group, Tissue Engineering and Reparative Dentistry, Cardiff University School of Dentistry, Cardiff, CF14 4XY, United Kingdom
| | - Edvar Onsøyen
- AlgiPharma AS, Industriveien 33, N-1337 Sandvika, Norway
| | - Phil D. Rye
- AlgiPharma AS, Industriveien 33, N-1337 Sandvika, Norway
| | - Chris Wright
- Centre for NanoHealth, Systems and Process Engineering Centre, College of Engineering, Swansea University, Swansea, SA2 8PP, United Kingdom
| | - David W. Thomas
- Advanced Therapies Group, Tissue Engineering and Reparative Dentistry, Cardiff University School of Dentistry, Cardiff, CF14 4XY, United Kingdom
| | - Katja E. Hill
- Advanced Therapies Group, Tissue Engineering and Reparative Dentistry, Cardiff University School of Dentistry, Cardiff, CF14 4XY, United Kingdom
| |
Collapse
|
32
|
Amich J, Calera JA. Zinc acquisition: a key aspect in Aspergillus fumigatus virulence. Mycopathologia 2014; 178:379-85. [PMID: 24947168 DOI: 10.1007/s11046-014-9764-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 05/28/2014] [Indexed: 10/25/2022]
Abstract
Zinc is an essential micronutrient required for the growth of all microorganisms. To grow in the lungs of a susceptible patient Aspergillus fumigatus must obtain zinc from the surrounding tissues. The concentration of Zn(2+) ions in living tissues is much lower than that required for optimal fungal growth in vitro because most of them are tightly bound to proteins at the physiological pH. However, A. fumigatus has several zinc transporters (ZrfA, ZrfB and ZrfC) that enable it to uptake zinc efficiently under the extreme zinc-limiting conditions provided by a susceptible host. The ZafA transcriptional regulator induces the expression of these transporters and is essential for virulence. ZrfC is required for fungal growth within the host tissues, whereas ZrfA and ZrfB play an accessory role. The zinc-scavenging capacity of ZrfC relies on its unusually long N-terminus. In addition, ZrfC also enables A. fumigatus to overcome the inhibitory effect of calprotectin, which is an antimicrobial Zn/Mn-chelating protein synthesized in high amounts by neutrophils, even in immunosuppressed non-leucopenic animals. In summary, the regulation of zinc homeostasis and zinc acquisition could be promising targets for the discovery and development of a new generation of antifungals for the treatment of invasive pulmonary aspergillosis.
Collapse
Affiliation(s)
- Jorge Amich
- Instituto de Biología Funcional y Genómica (IBFG) (centro mixto CSIC-Universidad de Salamanca) y Departamento de Microbiología y Genética, Universidad de Salamanca, Edificio IBFG, Lab. P1.10. C/Zacarías González nº 2, 37007, Salamanca, Spain
| | | |
Collapse
|
33
|
Sripetchwandee J, Pipatpiboon N, Chattipakorn N, Chattipakorn S. Combined therapy of iron chelator and antioxidant completely restores brain dysfunction induced by iron toxicity. PLoS One 2014; 9:e85115. [PMID: 24400127 PMCID: PMC3882264 DOI: 10.1371/journal.pone.0085115] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 12/02/2013] [Indexed: 12/25/2022] Open
Abstract
Background Excessive iron accumulation leads to iron toxicity in the brain; however the underlying mechanism is unclear. We investigated the effects of iron overload induced by high iron-diet consumption on brain mitochondrial function, brain synaptic plasticity and learning and memory. Iron chelator (deferiprone) and antioxidant (n-acetyl cysteine) effects on iron-overload brains were also studied. Methodology Male Wistar rats were fed either normal diet or high iron-diet consumption for 12 weeks, after which rats in each diet group were treated with vehicle or deferiprone (50 mg/kg) or n-acetyl cysteine (100 mg/kg) or both for another 4 weeks. High iron-diet consumption caused brain iron accumulation, brain mitochondrial dysfunction, impaired brain synaptic plasticity and cognition, blood-brain-barrier breakdown, and brain apoptosis. Although both iron chelator and antioxidant attenuated these deleterious effects, combined therapy provided more robust results. Conclusion In conclusion, this is the first study demonstrating that combined iron chelator and anti-oxidant therapy completely restored brain function impaired by iron overload.
Collapse
Affiliation(s)
- Jirapas Sripetchwandee
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Noppamas Pipatpiboon
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- * E-mail:
| | - Siriporn Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
34
|
Targeting iron acquisition blocks infection with the fungal pathogens Aspergillus fumigatus and Fusarium oxysporum. PLoS Pathog 2013; 9:e1003436. [PMID: 23853581 PMCID: PMC3708856 DOI: 10.1371/journal.ppat.1003436] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 05/02/2013] [Indexed: 12/21/2022] Open
Abstract
Filamentous fungi are an important cause of pulmonary and systemic morbidity and mortality, and also cause corneal blindness and visual impairment worldwide. Utilizing in vitro neutrophil killing assays and a model of fungal infection of the cornea, we demonstrated that Dectin-1 dependent IL-6 production regulates expression of iron chelators, heme and siderophore binding proteins and hepcidin in infected mice. In addition, we show that human neutrophils synthesize lipocalin-1, which sequesters fungal siderophores, and that topical lipocalin-1 or lactoferrin restricts fungal growth in vivo. Conversely, we show that exogenous iron or the xenosiderophore deferroxamine enhances fungal growth in infected mice. By examining mutant Aspergillus and Fusarium strains, we found that fungal transcriptional responses to low iron levels and extracellular siderophores are essential for fungal growth during infection. Further, we showed that targeting fungal iron acquisition or siderophore biosynthesis by topical application of iron chelators or statins reduces fungal growth in the cornea by 60% and that dual therapy with the iron chelator deferiprone and statins further restricts fungal growth by 75%. Together, these studies identify specific host iron-chelating and fungal iron-acquisition mediators that regulate fungal growth, and demonstrate that therapeutic inhibition of fungal iron acquisition can be utilized to treat topical fungal infections.
Collapse
|
35
|
Kronstad JW, Hu G, Jung WH. An encapsulation of iron homeostasis and virulence in Cryptococcus neoformans. Trends Microbiol 2013; 21:457-65. [PMID: 23810126 DOI: 10.1016/j.tim.2013.05.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 05/24/2013] [Accepted: 05/29/2013] [Indexed: 02/07/2023]
Abstract
Vertebrate hosts actively sequester iron, and fungal and other pathogens must therefore adapt to a severe limitation in iron availability to cause disease. Recent studies reveal that the pathogenic fungus Cryptococcus neoformans overcomes iron limitation by multiple mechanisms that target transferrin and heme. The regulation of iron uptake is mediated by an interconnected set of transcription factors that include the master iron regulator Cir1 and the pH-responsive factor Rim101. These factors integrate iron homeostasis with a myriad of other functions including pH sensing, nutrient and stress signaling pathways, virulence factor elaboration, and cell wall biogenesis.
Collapse
Affiliation(s)
- James W Kronstad
- Department of Microbiology and Immunology, The Michael Smith Laboratories, University of British Columbia, Vancouver BC, V6T 1Z4, Canada.
| | | | | |
Collapse
|
36
|
Álvarez F, Fernández-Ruiz M, Aguado JM. [Iron and invasive fungal infection]. Rev Iberoam Micol 2013; 30:217-25. [PMID: 23684655 DOI: 10.1016/j.riam.2013.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 04/08/2013] [Accepted: 04/30/2013] [Indexed: 01/19/2023] Open
Abstract
Iron is an essential factor for both the growth and virulence of most of microorganisms. As a part of the innate (or nutritional) immune system, mammals have developed different mechanisms to store and transport this element in order to limit free iron bioavailability. To survive in this hostile environment, pathogenic fungi have specific uptake systems for host iron sources, one of the most important of which is based on the synthesis of siderophores-soluble, low-molecular-mass, high-affinity iron chelators. The increase in free iron that results from iron-overload conditions is a well-established risk factor for invasive fungal infection (IFI) such as mucormycosis or aspergillosis. Therefore, iron chelation may be an appealing therapeutic option for these infections. Nevertheless, deferoxamine -the first approved iron chelator- paradoxically increases the incidence of IFI, as it serves as a xeno-siderophore to Mucorales. On the contrary, the new oral iron chelators (deferiprone and deferasirox) have shown to exert a deleterious effect on fungal growth both in vitro and in animal models. The present review focuses on the role of iron metabolism in the pathogenesis of IFI and summarises the preclinical data, as well as the limited clinical experience so far, in the use of new iron chelators as treatment for mucormycosis and invasive aspergillosis.
Collapse
Affiliation(s)
- Florencio Álvarez
- Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (i+12), Universidad Complutense, Madrid, España
| | | | | |
Collapse
|
37
|
Hosogaya N, Miyazaki T, Nagi M, Tanabe K, Minematsu A, Nagayoshi Y, Yamauchi S, Nakamura S, Imamura Y, Izumikawa K, Kakeya H, Yanagihara K, Miyazaki Y, Kugiyama K, Kohno S. The heme-binding protein Dap1 links iron homeostasis to azole resistance via the P450 protein Erg11 in Candida glabrata. FEMS Yeast Res 2013; 13:411-21. [PMID: 23496820 DOI: 10.1111/1567-1364.12043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/07/2013] [Accepted: 03/08/2013] [Indexed: 11/29/2022] Open
Abstract
The pathogenic fungus Candida glabrata is relatively resistant to azole antifungals, which target lanosterol 14α-demethylase (Erg11p) in the ergosterol biosynthesis pathway. Our study revealed that C. glabrata exhibits increased azole susceptibility under low-iron conditions. To investigate the molecular basis of this phenomenon, we generated a strain lacking the heme (iron protoporphyrin IX)-binding protein Dap1 in C. glabrata. The Δdap1 mutant displayed growth defects under iron-limited conditions, decreased azole tolerance, decreased production of ergosterol, and increased accumulation of 14α-methylated sterols lanosterol and squalene. All the Δdap1 phenotypes were complemented by wild-type DAP1, but not by DAP1(D91G) , in which a heme-binding site is mutated. Furthermore, azole tolerance of the Δdap1 mutant was rescued by exogenous ergosterol but not by iron supplementation alone. These results suggest that heme binding by Dap1 is crucial for Erg11 activity and ergosterol biosynthesis, thereby being required for azole tolerance. A Dap1-GFP fusion protein predominantly localized to vacuolar membranes and endosomes, and the Δdap1 cells exhibited aberrant vacuole morphologies, suggesting that Dap1 is also involved in the regulation of vacuole structures that could be important for iron storage. Our study demonstrates that Dap1 mediates a functional link between iron homeostasis and azole resistance in C. glabrata.
Collapse
Affiliation(s)
- Naoki Hosogaya
- Department of Molecular Microbiology and Immunology, Nagasaki University School of Medicine, Nagasaki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kim J, Cho YJ, Do E, Choi J, Hu G, Cadieux B, Chun J, Lee Y, Kronstad JW, Jung WH. A defect in iron uptake enhances the susceptibility of Cryptococcus neoformans to azole antifungal drugs. Fungal Genet Biol 2012; 49:955-66. [PMID: 22975303 DOI: 10.1016/j.fgb.2012.08.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 07/16/2012] [Accepted: 08/13/2012] [Indexed: 11/16/2022]
Abstract
The high-affinity reductive iron uptake system that includes a ferroxidase (Cfo1) and an iron permease (Cft1) is critical for the pathogenesis of Cryptococcus neoformans. In addition, a mutant lacking CFO1 or CFT1 not only has reduced iron uptake but also displays a markedly increased susceptibility to azole antifungal drugs. Altered antifungal susceptibility of the mutants was of particular interest because the iron uptake system has been proposed as an alternative target for antifungal treatment. In this study, we used transcriptome analysis to begin exploring the molecular mechanisms of altered antifungal susceptibility in a cfo1 mutant. The wild-type strain and the cfo1 mutant were cultured with or without the azole antifungal drug fluconazole and their transcriptomes were compared following sequencing with Illumina Genome Analyzer IIx (GAIIx) technology. As expected, treatment of both strains with fluconazole caused elevated expression of genes in the ergosterol biosynthetic pathway that includes the target enzyme Erg11. Additionally, genes differentially expressed in the cfo1 mutant were involved in iron uptake and homeostasis, mitochondrial functions and respiration. The cfo1 mutant also displayed phenotypes consistent with these changes including a reduced ratio of NAD(+)/NADH and down-regulation of Fe-S cluster synthesis. Moreover, combination treatment of the wild-type strain with fluconazole and the respiration inhibitor diphenyleneiodonium dramatically increased susceptibility to fluconazole. This result supports the hypothesis that down-regulation of genes required for respiration contributed to the altered fluconazole susceptibility of the cfo1 mutant. Overall, our data suggest that iron uptake and homeostasis play a key role in antifungal susceptibility and could be used as novel targets for combination treatment of cryptococcosis. Indeed, we found that iron chelation in combination with fluconazole treatment synergistically inhibited the growth of C. neoformans.
Collapse
Affiliation(s)
- Jeongmi Kim
- Department of Biotechnology, Chung-Ang University, Anseong 456-756, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
INTRODUCTION Since the first application of antibiotics to treat bacterial infections, the development and spread of resistance has been a persistent threat. An ever evolving pipeline of next-generation therapeutics is required for modern medicine to remain one step ahead of pathogens. AREAS COVERED This review describes recent efforts to develop drugs that interrupt the assimilation of iron by bacteria: a process that is vital to cellular homeostasis and is not currently targeted by antibiotics used in the clinic. This review also covers the mechanisms by which bacteria acquire iron for their environment, and details efforts to intervene in these processes, using small molecule inhibitors that target key steps in these pathways, with a special emphasis on recent advances published during the 2010 - 2012 period. EXPERT OPINION For decades, the routes used by bacteria to assimilate iron from host and environmental settings have been the subject of intense study. While numerous investigations have identified inhibitors of these pathways, many have stopped short of translating the in vitro results to in vivo proof of concept experiments. The extension of preliminary findings in this manner will significantly increase the impact of the field.
Collapse
Affiliation(s)
- Timothy L Foley
- National Institutes of Health, National Center for Advancing Translational Sciences, Division of Preclinical Innovation, 9800 Medical Center Drive, Bethesda, MD 20892-3370, USA
| | | |
Collapse
|
40
|
Antibacterial activities of iron chelators against common nosocomial pathogens. Antimicrob Agents Chemother 2012; 56:5419-21. [PMID: 22850524 DOI: 10.1128/aac.01197-12] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The activities of iron chelators (deferoxamine, deferiprone, Apo6619, and VK28) were evaluated against type strains of Acinetobacter baumannii, Pseudomonas aeruginosa, Staphylococcus aureus, Klebsiella pneumoniae, and Escherichia coli. Deferiprone, Apo6619, and VK28 each inhibited growth in standard and RPMI tissue culture medium, while deferoxamine had no effect. Additionally, time-kill assays revealed that VK28 had a bacteriostatic effect against S. aureus. Therefore, these newly developed iron chelators might provide a nontraditional approach for treatment of bacterial infections.
Collapse
|
41
|
Campbell BC, Chan KL, Kim JH. Chemosensitization as a means to augment commercial antifungal agents. Front Microbiol 2012; 3:79. [PMID: 22393330 PMCID: PMC3289909 DOI: 10.3389/fmicb.2012.00079] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Accepted: 02/15/2012] [Indexed: 11/13/2022] Open
Abstract
Antimycotic chemosensitization and its mode of action are of growing interest. Currently, use of antifungal agents in agriculture and medicine has a number of obstacles. Foremost of these is development of resistance or cross-resistance to one or more antifungal agents. The generally high expense and negative impact, or side effects, associated with antifungal agents are two further issues of concern. Collectively, these problems are exacerbated by efforts to control resistant strains, which can evolve into a treadmill of higher dosages for longer periods. This cycle in turn, inflates cost of treatment, dramatically. A further problem is stagnation in development of new and effective antifungal agents, especially for treatment of human mycoses. Efforts to overcome some of these issues have involved using combinations of available antimycotics (e.g., combination therapy for invasive mycoses). However, this approach has had inconsistent success and is often associated with a marked increase in negative side effects. Chemosensitization by natural compounds to increase effectiveness of commercial antimycotics is a somewhat new approach to dealing with the aforementioned problems. The potential for safe natural products to improve antifungal activity has been observed for over three decades. Chemosensitizing agents possess antifungal activity, but at insufficient levels to serve as antimycotics, alone. Their main function is to disrupt fungal stress response, destabilize the structural integrity of cellular and vacuolar membranes or stimulate production of reactive oxygen species, augmenting oxidative stress and apoptosis. Use of safe chemosensitizing agents has potential benefit to both agriculture and medicine. When co-applied with a commercial antifungal agent, an additive or synergistic interaction may occur, augmenting antifungal efficacy. This augmentation, in turn, lowers effective dosages, costs, negative side effects and, in some cases, countermands resistance.
Collapse
Affiliation(s)
- Bruce C. Campbell
- Plant Mycotoxin Research Unit, Western Regional Research Center, Agricultural Research Service, United States Department of AgricultureAlbany, CA, USA
| | - Kathleen L. Chan
- Plant Mycotoxin Research Unit, Western Regional Research Center, Agricultural Research Service, United States Department of AgricultureAlbany, CA, USA
| | - Jong H. Kim
- Plant Mycotoxin Research Unit, Western Regional Research Center, Agricultural Research Service, United States Department of AgricultureAlbany, CA, USA
| |
Collapse
|
42
|
Haas H. Iron - A Key Nexus in the Virulence of Aspergillus fumigatus. Front Microbiol 2012; 3:28. [PMID: 22347220 PMCID: PMC3272694 DOI: 10.3389/fmicb.2012.00028] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 01/16/2012] [Indexed: 01/01/2023] Open
Abstract
Iron is an essential but, in excess, toxic nutrient. Therefore, fungi evolved fine-tuned mechanisms for uptake and storage of iron, such as the production of siderophores (low-molecular mass iron-specific chelators). In Aspergillus fumigatus, iron starvation causes extensive transcriptional remodeling involving two central transcription factors, which are interconnected in a negative transcriptional feed-back loop: the GATA-factor SreA and the bZip-factor HapX. During iron sufficiency, SreA represses iron uptake, including reductive iron assimilation and siderophore-mediated iron uptake, to avoid toxic effects. During iron starvation, HapX represses iron-consuming pathways, including heme biosynthesis and respiration, to spare iron and activates synthesis of ribotoxin AspF1 and siderophores, the latter partly by ensuring supply of the precursor, ornithine. In accordance with the expression pattern and mode of action, detrimental effects of inactivation of SreA and HapX are confined to growth during iron sufficiency and iron starvation, respectively. Deficiency in HapX, but not SreA, attenuates virulence of A. fumigatus in a murine model of aspergillosis, which underlines the crucial role of adaptation to iron limitation in virulence. Consistently, production of both extra and intracellular siderophores is crucial for virulence of A. fumigatus. Recently, the sterol regulatory element binding protein SrbA was found to be essential for adaptation to iron starvation, thereby linking regulation of iron metabolism, ergosterol biosynthesis, azole drug resistance, and hypoxia adaptation.
Collapse
Affiliation(s)
- Hubertus Haas
- Division of Molecular Biology/Biocenter, Innsbruck Medical University Innsbruck, Austria
| |
Collapse
|
43
|
Mevalonate governs interdependency of ergosterol and siderophore biosyntheses in the fungal pathogen Aspergillus fumigatus. Proc Natl Acad Sci U S A 2011; 109:E497-504. [PMID: 22106303 DOI: 10.1073/pnas.1106399108] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aspergillus fumigatus is the most common airborne fungal pathogen for humans. In this mold, iron starvation induces production of the siderophore triacetylfusarinine C (TAFC). Here we demonstrate a link between TAFC and ergosterol biosynthetic pathways, which are both critical for virulence and treatment of fungal infections. Consistent with mevalonate being a limiting prerequisite for TAFC biosynthesis, we observed increased expression of 3-hydroxy-3-methyl-glutaryl (HMG)-CoA reductase (Hmg1) under iron starvation, reduced TAFC biosynthesis following lovastatin-mediated Hmg1 inhibition, and increased TAFC biosynthesis following Hmg1 overexpression. We identified enzymes, the acyl-CoA ligase SidI and the enoyl-CoA hydratase SidH, linking biosynthesis of mevalonate and TAFC, deficiency of which under iron starvation impaired TAFC biosynthesis, growth, oxidative stress resistance, and murine virulence. Moreover, inactivation of these enzymes alleviated TAFC-derived biosynthetic demand for mevalonate, as evidenced by increased resistance to lovastatin. Concordant with bilateral demand for mevalonate, iron starvation decreased the ergosterol content and composition, a phenotype that is mitigated in TAFC-lacking mutants.
Collapse
|
44
|
Pitman SK, Drew RH, Perfect JR. Addressing current medical needs in invasive fungal infection prevention and treatment with new antifungal agents, strategies and formulations. Expert Opin Emerg Drugs 2011; 16:559-586. [DOI: 10.1517/14728214.2011.607811] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
45
|
Schrettl M, Haas H. Iron homeostasis--Achilles' heel of Aspergillus fumigatus? Curr Opin Microbiol 2011; 14:400-5. [PMID: 21724450 PMCID: PMC3162135 DOI: 10.1016/j.mib.2011.06.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 06/08/2011] [Indexed: 12/16/2022]
Abstract
The opportunistic fungal pathogen Aspergillus fumigatus adapts to iron limitation by upregulation of iron uptake mechanisms including siderophore biosynthesis and downregulation of iron-consuming pathways to spare iron. These metabolic changes depend mainly on the transcription factor HapX. Consistent with the crucial role of iron in pathophysiology, genetic inactivation of either HapX or the siderophore system attenuates virulence of A. fumigatus in a murine model of aspergillosis. The differences in iron handling between mammals and fungi might serve to improve therapy and diagnosis of fungal infections.
Collapse
Affiliation(s)
- Markus Schrettl
- Division of Molecular Biology/Biocenter, Innsbruck Medical University, Fritz-Pregl-Str. 3, A-6020 Innsbruck, Austria
| | | |
Collapse
|
46
|
Gonzales DA, De Torre C, Wang H, Devor CB, Munson PJ, Ying SX, Kern SJ, Petraitiene R, Levens DL, Walsh TJ, Suffredini AF. Protein expression profiles distinguish between experimental invasive pulmonary aspergillosis and Pseudomonas pneumonia. Proteomics 2011; 10:4270-80. [PMID: 21089047 DOI: 10.1002/pmic.200900768] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We hypothesized that invasive pulmonary aspergillosis (IPA) may generate a distinctive proteomic signature in plasma and bronchoalveolar lavage (BAL). Proteins in plasma and BAL from two neutropenic rabbit models of IPA and Pseudomonas pneumonia were analyzed by SELDI-TOF MS. Hierarchical clustering analysis of plasma time course spectra demonstrated two clusters of peaks that were differentially regulated between IPA and Pseudomonas pneumonia (57 and 34 peaks, respectively, p<0.001). PCA of plasma proteins demonstrated a time-dependent separation of the two infections. A random forest analysis that ranked the top 30 spectral points distinguished between late Aspergillus and Pseudomonas pneumonias with 100% sensitivity and specificity. Based on spectral data analysis, three proteins were identified using SDS-PAGE and LC/MS and quantified using reverse phase arrays. Differences in the temporal sequence of plasma haptoglobin (p<0.001), apolipoprotein A1 (p<0.001) and transthyretin (p<0.038) were observed between IPA and Pseudomonas pneumonia, as was C-reactive protein (p<0.001). In summary, proteomic analysis of plasma and BAL proteins of experimental Aspergillus and Pseudomonas pneumonias demonstrates unique protein profiles with principal components and spectral regions that are shared in early infection and diverge at later stages of infection. Haptoglobin, apolipoprotein A1, transthyretin, and C-reactive protein are differentially expressed in these infections suggesting important contributions to host defense against IPA.
Collapse
Affiliation(s)
- Denise A Gonzales
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892-1662, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Pinto E, Afonso C, Duarte S, Vale-Silva L, Costa E, Sousa E, Pinto M. Antifungal activity of xanthones: evaluation of their effect on ergosterol biosynthesis by high-performance liquid chromatography. Chem Biol Drug Des 2011; 77:212-22. [PMID: 21244637 DOI: 10.1111/j.1747-0285.2010.01072.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The increasing resistance of pathogenic fungi to antifungal compounds and the reduced number of available drugs led to the search for therapeutic alternatives among natural products, including xanthones. The antifungal activity of 27 simple oxygenated xanthones was evaluated by determination of their minimal inhibitory concentration on clinical and type strains of Candida, Cryptococcus, Aspergillus and dermatophytes, and their preponderance on the dermatophytic filamentous fungi was observed. Furthermore, a simple and efficient HPLC method with UV detection to study the effect of the active xanthones on the biosynthesis of ergosterol was developed and validated. Using this methodology, the identification and quantification of fungal sterols in whole cells of Candida albicans, Cryptococcus neoformans, Aspergillus fumigatus, and Trichophyton mentagrophytes were accomplished. In summary, 1,2-dihydroxyxanthone was found to be the most active compound against all strains tested, showing its effect on sterol biosynthesis by reducing the amount of ergosterol detected.
Collapse
Affiliation(s)
- Eugénia Pinto
- Research Center of Medicinal Chemistry - University of Porto (CEQUIMED-UP), R. Aníbal Cunha, 164, 4050-047, Porto, Portugal.
| | | | | | | | | | | | | |
Collapse
|
48
|
Kell DB. Towards a unifying, systems biology understanding of large-scale cellular death and destruction caused by poorly liganded iron: Parkinson's, Huntington's, Alzheimer's, prions, bactericides, chemical toxicology and others as examples. Arch Toxicol 2010; 84:825-89. [PMID: 20967426 PMCID: PMC2988997 DOI: 10.1007/s00204-010-0577-x] [Citation(s) in RCA: 286] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 07/14/2010] [Indexed: 12/11/2022]
Abstract
Exposure to a variety of toxins and/or infectious agents leads to disease, degeneration and death, often characterised by circumstances in which cells or tissues do not merely die and cease to function but may be more or less entirely obliterated. It is then legitimate to ask the question as to whether, despite the many kinds of agent involved, there may be at least some unifying mechanisms of such cell death and destruction. I summarise the evidence that in a great many cases, one underlying mechanism, providing major stresses of this type, entails continuing and autocatalytic production (based on positive feedback mechanisms) of hydroxyl radicals via Fenton chemistry involving poorly liganded iron, leading to cell death via apoptosis (probably including via pathways induced by changes in the NF-κB system). While every pathway is in some sense connected to every other one, I highlight the literature evidence suggesting that the degenerative effects of many diseases and toxicological insults converge on iron dysregulation. This highlights specifically the role of iron metabolism, and the detailed speciation of iron, in chemical and other toxicology, and has significant implications for the use of iron chelating substances (probably in partnership with appropriate anti-oxidants) as nutritional or therapeutic agents in inhibiting both the progression of these mainly degenerative diseases and the sequelae of both chronic and acute toxin exposure. The complexity of biochemical networks, especially those involving autocatalytic behaviour and positive feedbacks, means that multiple interventions (e.g. of iron chelators plus antioxidants) are likely to prove most effective. A variety of systems biology approaches, that I summarise, can predict both the mechanisms involved in these cell death pathways and the optimal sites of action for nutritional or pharmacological interventions.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry and the Manchester Interdisciplinary Biocentre, The University of Manchester, Manchester M1 7DN, UK.
| |
Collapse
|
49
|
Kontoghiorghes GJ, Kolnagou A, Skiada A, Petrikkos G. The Role of Iron and Chelators on Infections in Iron Overload and Non Iron Loaded Conditions: Prospects for the Design of New Antimicrobial Therapies. Hemoglobin 2010; 34:227-39. [DOI: 10.3109/03630269.2010.483662] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
50
|
Tunçcan OG, Yegin ZA, Ozkurt ZN, Erbaş G, Akı SZ, Senol E, Yağcı M, Sucak G. High ferritin levels are associated with hepatosplenic candidiasis in hematopoietic stem cell transplant candidates. Int J Infect Dis 2010; 14 Suppl 3:e104-7. [PMID: 20307998 DOI: 10.1016/j.ijid.2009.11.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2009] [Revised: 11/16/2009] [Accepted: 11/17/2009] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES Invasive fungal infections (IFI) are a significant cause of morbidity and mortality in hematopoietic stem cell transplant (HSCT) recipients. Hepatosplenic candidiasis (HSC) is defined as a distinct form of invasive candidiasis, with liver, spleen, and kidney involvement, in patients with hematological disorders. METHODS The charts of 255 patients (male/female 168/87; median age 35 (range 16-71) years) who were evaluated pre-HSCT at the Gazi University Hospital Stem Cell Transplantation Unit between 2003 and 2008, were retrospectively reviewed. RESULTS HSC, which was demonstrated in six (2.3%) patients, was found to be more common in allogeneic HSCT recipients than in autologous HSCT recipients and in patients who had received two or more previous chemotherapy courses than in patients who had received fewer than two (p>0.05). Patients with HSC tended to have a worse performance status than patients without HSC according to the World Health Organization (p=0.001) and Karnofsky scale (p=0.007). Pre-transplantation ferritin (p=0.008) and acute phase reactant levels, including erythrocyte sedimentation rate (p=0.025) and C-reactive protein (p=0.007), were significantly higher in patients with HSC than in patients without HSC. CONCLUSIONS This study shows the predictive role of pre-transplantation ferritin levels in selecting a subset of patients at increased risk for HSC. Pre-transplantation risk assessment and targeted strategies might lower the morbidity and mortality of IFI in HSCT recipients.
Collapse
Affiliation(s)
- Ozlem Güzel Tunçcan
- Department of Clinical Microbiology and Infectious Diseases, Gazi University Faculty of Medicine, Ankara, Turkey
| | | | | | | | | | | | | | | |
Collapse
|