1
|
Boudier A, Mammari N, Lamouroux E, Duval RE. Inorganic Nanoparticles: Tools to Emphasize the Janus Face of Amphotericin B. Antibiotics (Basel) 2023; 12:1543. [PMID: 37887244 PMCID: PMC10604816 DOI: 10.3390/antibiotics12101543] [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: 09/25/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Amphotericin B is the oldest antifungal molecule which is still currently widely used in clinical practice, in particular for the treatment of invasive diseases, even though it is not devoid of side effects (particularly nephrotoxicity). Recently, its redox properties (i.e., both prooxidant and antioxidant) have been highlighted in the literature as mechanisms involved in both its activity and its toxicity. Interestingly, similar properties can be described for inorganic nanoparticles. In the first part of the present review, the redox properties of Amphotericin B and inorganic nanoparticles are discussed. Then, in the second part, inorganic nanoparticles as carriers of the drug are described. A special emphasis is given to their combined redox properties acting either as a prooxidant or as an antioxidant and their connection to the activity against pathogens (i.e., fungi, parasites, and yeasts) and to their toxicity. In a majority of the published studies, inorganic nanoparticles carrying Amphotericin B are described as having a synergistic activity directly related to the rupture of the redox homeostasis of the pathogen. Due to the unique properties of inorganic nanoparticles (e.g., magnetism, intrinsic anti-infectious properties, stimuli-triggered responses, etc.), these nanomaterials may represent a new generation of medicine that can synergistically enhance the antimicrobial properties of Amphotericin B.
Collapse
Affiliation(s)
| | - Nour Mammari
- Université de Lorraine, CNRS, LCM, F-54000 Nancy, France; (N.M.); (E.L.)
| | - Emmanuel Lamouroux
- Université de Lorraine, CNRS, LCM, F-54000 Nancy, France; (N.M.); (E.L.)
| | - Raphaël E. Duval
- Université de Lorraine, CNRS, LCM, F-54000 Nancy, France; (N.M.); (E.L.)
- ABC Platform, F-54505 Vandœuvre-lès-Nancy, France
| |
Collapse
|
2
|
Galinari CB, Biachi TDP, Gonçalves RS, Cesar GB, Bergmann EV, Malacarne LC, Kioshima Cotica ÉS, Bonfim-Mendonça PDS, Svidzinski TIE. Photoactivity of hypericin: from natural product to antifungal application. Crit Rev Microbiol 2023; 49:38-56. [PMID: 35171731 DOI: 10.1080/1040841x.2022.2036100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Considering the multifaceted and increasing application of photodynamic therapy (PDT), in recent years the antimicrobial employment of this therapy has been highlighted, because of the antiviral, antibacterial, antiparasitic, and antifungal activities that have already been demonstrated. In this context, research focussed on antimycological action, especially for treatment of superficial infections, presents promising growth due to the characteristics of these infections that facilitate PDT application as new therapeutic options are needed in the field of medical mycology. Among the more than one hundred classes of photosensitizers the antifungal action of hypericin (Hyp) stands out due to its ability to permeate the lipid membrane and accumulate in different cytoplasmic organelles of eukaryotic cells. In this review, we aim to provide a complete overview of the origin, physicochemical characteristics, and optimal alternative drug deliveries that promote the photodynamic action of Hyp (Hyp-PDT) against fungi. Furthermore, considering the lack of a methodological consensus, we intend to compile the best strategies to guide researchers in the antifungal application of Hyp-PDT. Overall, this review provides a future perspective of new studies and clinical possibilities for the advances of such a technique in the treatment of mycoses in humans.
Collapse
Affiliation(s)
- Camila Barros Galinari
- Department of Analysis Clinics & Biomedicine, State University of Maringá, Paraná, Brazil
| | - Tiago de Paula Biachi
- Department of Analysis Clinics & Biomedicine, State University of Maringá, Paraná, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Rivera-Yañez CR, Ruiz-Hurtado PA, Reyes-Reali J, Mendoza-Ramos MI, Vargas-Díaz ME, Hernández-Sánchez KM, Pozo-Molina G, Méndez-Catalá CF, García-Romo GS, Pedroza-González A, Méndez-Cruz AR, Nieto-Yañez O, Rivera-Yañez N. Antifungal Activity of Mexican Propolis on Clinical Isolates of Candida Species. Molecules 2022; 27:molecules27175651. [PMID: 36080417 PMCID: PMC9457601 DOI: 10.3390/molecules27175651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Infections caused by micro-organisms of the genus Candida are becoming a growing health problem worldwide. These fungi are opportunistic commensals that can produce infections—clinically known as candidiasis—in immunocompromised individuals. The indiscriminate use of different anti-fungal treatments has triggered the resistance of Candida species to currently used therapies. In this sense, propolis has been shown to have potent antimicrobial properties and thus can be used as an approach for the inhibition of Candida species. Therefore, this work aims to evaluate the anti-Candida effects of a propolis extract obtained from the north of Mexico on clinical isolates of Candida species. Candida species were specifically identified from oral lesions, and both the qualitative and quantitative anti-Candida effects of the Mexican propolis were evaluated, as well as its inhibitory effect on C. albicans isolate’s germ tube growth and chemical composition. Three Candida species were identified, and our results indicated that the inhibition halos of the propolis ranged from 7.6 to 21.43 mm, while that of the MFC and FC50 ranged from 0.312 to 1.25 and 0.014 to 0.244 mg/mL, respectively. Moreover, the propolis was found to inhibit germ tube formation (IC50 ranging from 0.030 to 1.291 mg/mL). Chemical composition analysis indicated the presence of flavonoids, including pinocembrin, baicalein, pinobanksin chalcone, rhamnetin, and biochanin A, in the Mexican propolis extract. In summary, our work shows that Mexican propolis presents significant anti-Candida effects related to its chemical composition, and also inhibits germ tube growth. Other Candida species virulence factors should be investigated in future research in order to determine the mechanisms associated with antifungal effects against them.
Collapse
Affiliation(s)
- Claudia Rebeca Rivera-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Porfirio Alonso Ruiz-Hurtado
- Laboratorio de Toxicología de Productos Naturales, Departamento de Farmacia, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Av. Wilfrido Massieu, Gustavo A. Madero 07738, Mexico
| | - Julia Reyes-Reali
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - María Isabel Mendoza-Ramos
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - María Elena Vargas-Díaz
- Laboratorio de Química de Productos Naturales, Departamento de Química Orgánica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala, Ciudad de México 11340, Mexico
| | - Karla Mariela Hernández-Sánchez
- Laboratorio de Química de Productos Naturales, Departamento de Química Orgánica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala, Ciudad de México 11340, Mexico
| | - Glustein Pozo-Molina
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Claudia Fabiola Méndez-Catalá
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Gina Stella García-Romo
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Alexander Pedroza-González
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Adolfo René Méndez-Cruz
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Unidad de Morfofisiología y Función, Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Oscar Nieto-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (O.N.-Y.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| | - Nelly Rivera-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (O.N.-Y.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| |
Collapse
|
4
|
Abstract
Altered lipid metabolism is a characteristic feature and potential driving factor of acute kidney injury (AKI). Of the lipids that accumulate in injured renal tissues, ceramides are potent regulators of metabolism and cell fate. Up-regulation of ceramide synthesis is a common feature shared across several AKI etiologies in vitro and in vivo. Furthermore, ceramide accumulation is an early event in the natural history of AKI that precedes cell death and organ dysfunction. Emerging evidence suggests that inhibition of ceramide accumulation may improve renal outcomes in several models of AKI. This review examines the landscape of ceramide metabolism and regulation in the healthy and injured kidney. Furthermore, we discuss the body of literature regarding ceramides as therapeutic targets for AKI and consider potential mechanisms by which ceramides drive kidney pathogenesis.
Collapse
Affiliation(s)
- Rebekah J Nicholson
- Department of Nutrition and Integrative Physiology, Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT
| | - William L Holland
- Department of Nutrition and Integrative Physiology, Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT.
| |
Collapse
|
5
|
Humelnicu AC, Samoilă P, Cojocaru C, Dumitriu R, Bostănaru AC, Mareș M, Harabagiu V, Simionescu BC. Chitosan-Based Therapeutic Systems for Superficial Candidiasis Treatment. Synergetic Activity of Nystatin and Propolis. Polymers (Basel) 2022; 14:689. [PMID: 35215602 PMCID: PMC8876245 DOI: 10.3390/polym14040689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
The paper deals with new approaches to chitosan (CS)-based antifungal therapeutic formulations designed to fulfill the requirements of specific applications. Gel-like formulations were prepared by mixing CS dissolved in aqueous lactic acid (LA) solution with nystatin (NYS) powder and/or propolis (PRO) aqueous solution dispersed in glycerin, followed by water evaporation to yield flexible mesoporous (pore widths of 2-4 nm) films of high specific surfaces between 1 × 103 and 1.7 × 103 m2/g. Morphological evaluation of the antifungal films showed uniform dispersion and downsizing of NYS crystallites (with initial sizes up to 50 μm). Their mechanical properties were found to be close to those of soft tissues (Young's modulus values between 0.044-0.025 MPa). The films presented hydration capacities in physiological condition depending on their composition, i.e., higher for NYS-charged (628%), as compared with PRO loaded films (118-129%). All NYS charged films presented a quick release for the first 10 min followed by a progressive increase of the release efficiency at 48.6%, for the samples containing NYS alone and decreasing values with increasing amount of PRO to 45.9% and 42.8% after 5 h. By in vitro analysis, the hydrogels with acidic pH values around 3.8 were proven to be active against Candida albicans and Candida glabrata species. The time-killing assay performed during 24 h on Candida albicans in synthetic vagina-simulative medium showed that the hydrogel formulations containing both NYS and PRO presented the faster slowing down of the fungal growth, from colony-forming unit (CFU)/mL of 1.24 × 107 to CFU/mL < 10 (starting from the first 6 h).
Collapse
Affiliation(s)
- Andra-Cristina Humelnicu
- “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (A.-C.H.); (P.S.); (C.C.); (R.D.); (B.C.S.)
| | - Petrișor Samoilă
- “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (A.-C.H.); (P.S.); (C.C.); (R.D.); (B.C.S.)
| | - Corneliu Cojocaru
- “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (A.-C.H.); (P.S.); (C.C.); (R.D.); (B.C.S.)
| | - Raluca Dumitriu
- “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (A.-C.H.); (P.S.); (C.C.); (R.D.); (B.C.S.)
| | - Andra-Cristina Bostănaru
- Laboratory of Antimicrobial Chemotherapy, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania;
| | - Mihai Mareș
- Laboratory of Antimicrobial Chemotherapy, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania;
| | - Valeria Harabagiu
- “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (A.-C.H.); (P.S.); (C.C.); (R.D.); (B.C.S.)
| | - Bogdan C. Simionescu
- “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (A.-C.H.); (P.S.); (C.C.); (R.D.); (B.C.S.)
| |
Collapse
|
6
|
Tocci N, Perenzoni D, Iamonico D, Fava F, Weil T, Mattivi F. Extracts From Hypericum hircinum subsp. majus Exert Antifungal Activity Against a Panel of Sensitive and Drug-Resistant Clinical Strains. Front Pharmacol 2018; 9:382. [PMID: 29755350 PMCID: PMC5932341 DOI: 10.3389/fphar.2018.00382] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/03/2018] [Indexed: 12/27/2022] Open
Abstract
During the last two decades incidences of fungal infections dramatically increased and the often accompanying failure of available antifungal therapies represents a substantial clinical problem. The urgent need for novel antimycotics called particular attention to the study of natural products. The genus Hypericum includes many species that are used in the traditional medicine to treat pathological states like inflammations and infections caused by fungi. However, despite the diffused use of Hypericum-based products the antifungal potential of the genus is still poorly investigated. In this study five Hypericum species autochthonous of Central and Eastern Europe were evaluated regarding their polyphenolic content, their toxicological safety and their antifungal potential against a broad panel of clinical fungal isolates. LC-MS analysis led to the identification and quantification of 52 compounds, revealing that Hypericum extracts are rich sources of flavonols, benzoates and cinnamates, and of flavan-3-ols. An in-depth screen of the biological activity of crude extracts clearly unveiled H. hircinum subsp. majus as a promising candidate species for the search of novel antifungals. H. hircinum is diffused in the Mediterranean basin from Spain to Turkey where it is traditionally used to prepare a herbal tea indicated for the treatment of respiratory tract disorders, several of which are caused by fungi. Noteworthy, the infusion of H. hircinum subsp. majus excreted broad antifungal activity against Penicillium, Aspergillus and non-albicans Candida isolates comprising strains both sensitive and resistant to fluconazole. Additionally, it showed no cytotoxicity on human cells and the chemical characterization of the H. hircinum subsp. majus infusion revealed high amounts of the metabolite hyperoside. These results scientifically support the traditional use of H. hircinum extracts for the treatment of respiratory tract infections and suggest the presence of exploitable antifungal principles for further investigations aimed at developing novel antifungal therapies.
Collapse
Affiliation(s)
- Noemi Tocci
- Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Daniele Perenzoni
- Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Duilio Iamonico
- Laboratory of Phytogeography and Applied Geobotany, Section Environment and Landscape, Department of Planning, Design, and Technology of Architecture, Sapienza University of Rome, Rome, Italy
| | - Francesca Fava
- Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Tobias Weil
- Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Fulvio Mattivi
- Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Italy.,Centre Agriculture Food Environment, University of Trento, Trento, Italy
| |
Collapse
|
7
|
The Role of Signaling via Aqueous Pore Formation in Resistance Responses to Amphotericin B. Antimicrob Agents Chemother 2016; 60:5122-9. [PMID: 27381391 DOI: 10.1128/aac.00878-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Drug resistance studies have played an important role in the validation of antibiotic targets. In the case of the polyene antibiotic amphotericin B (AmB), such studies have demonstrated the essential role that depletion of ergosterol plays in the development of AmB-resistant (AmB-R) organisms. However, AmB-R strains also occur in fungi and parasitic protozoa that maintain a normal level of ergosterol at the plasma membrane. Here, I review evidence that shows not only that there is increased protection against the deleterious consequences of AmB-induced ion leakage across the membrane in these resistant pathogens but also that a set of events are activated that block the cell signaling responses that trigger the oxidative damage produced by the antibiotic. Such signaling events appear to be the consequence of a membrane-thinning effect that is exerted upon lipid-anchored Ras proteins by the aqueous pores formed by AmB. A similar membrane disturbance effect may also explain the activity of AmB on mammalian cells containing Toll-like receptors. These resistance mechanisms expand our current understanding of the role that the formation of AmB aqueous pores plays in triggering signal transduction responses in both pathogens and host immune cells.
Collapse
|
8
|
Davis SA, Vincent BM, Endo MM, Whitesell L, Marchillo K, Andes DR, Lindquist S, Burke MD. Nontoxic antimicrobials that evade drug resistance. Nat Chem Biol 2015; 11:481-7. [PMID: 26030729 PMCID: PMC4472574 DOI: 10.1038/nchembio.1821] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/10/2015] [Indexed: 01/21/2023]
Abstract
Drugs that act more promiscuously provide fewer routes for the emergence of resistant mutants. But this benefit often comes at the cost of serious off-target and dose-limiting toxicities. The classic example is the antifungal amphotericin B (AmB), which has evaded resistance for more than half a century. We report dramatically less toxic amphotericins that nevertheless evade resistance. They are scalably accessed in just three steps from the natural product, and bind their target (the fungal sterol, ergosterol) with far greater selectivity than AmB. Hence, they are less toxic and far more effective in a mouse model of systemic candidiasis. Surprisingly, exhaustive efforts to select for mutants resistant to these more selective compounds revealed that they are just as impervious to resistance as AmB. Thus, highly selective cytocidal action and the evasion of resistance are not mutually exclusive, suggesting practical routes to the discovery of less toxic, resistance-evasive therapies.
Collapse
Affiliation(s)
- Stephen A Davis
- 1] Howard Hughes Medical Institute, Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA. [2] Roger Adam Laboratory, Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Benjamin M Vincent
- 1] Microbiology Graduate Program, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Matthew M Endo
- 1] Howard Hughes Medical Institute, Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA. [2] Roger Adam Laboratory, Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Luke Whitesell
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Karen Marchillo
- 1] Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA. [2] Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, Wisconsin, USA
| | - David R Andes
- 1] Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA. [2] Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, Wisconsin, USA
| | - Susan Lindquist
- 1] Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA. [2] Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Martin D Burke
- 1] Howard Hughes Medical Institute, Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA. [2] Roger Adam Laboratory, Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
9
|
McDaniel BL, Bentley ML. The role of medications and their management in acute kidney injury. INTEGRATED PHARMACY RESEARCH AND PRACTICE 2015; 4:21-29. [PMID: 29354517 PMCID: PMC5741024 DOI: 10.2147/iprp.s52930] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Prior to 2002, the incidence of acute renal failure (ARF) varied as there was no standard definition. To better understand its incidence and etiology and to develop treatment and prevention strategies, while moving research forward, the Acute Dialysis Quality Initiative workgroup developed the RIFLE (risk, injury, failure, loss, end-stage kidney disease) classification. After continued data suggesting that even small increases in serum creatinine lead to worse outcomes, the Acute Kidney Injury Network (AKIN) modified the RIFLE criteria and used the term acute kidney injury (AKI) instead of ARF. These classification and staging systems provide the clinician and researcher a starting point for refining the understanding and treatment of AKI. An important initial step in evaluating AKI is determining the likely location of injury, generally classified as prerenal, renal, or postrenal. There is no single biomarker or test that definitively defines the mechanism of the injury. Identifying the insult(s) requires a thorough assessment of the patient and their medical and medication histories. Prerenal injuries arise primarily due to renal hypoperfusion. This may be the result of systemic or focal conditions or secondary to the effects of drugs such as nonsteroidal anti-inflammatory drugs, calcineurin inhibitors (CIs), and modulators of the renin-angiotensin-aldosterone system. Renal, or intrinsic, injury is an overarching term that represents complex conditions leading to considerable damage to a component of the intrinsic renal system (renal tubules, glomerulus, vascular structures, inter-stitium, or renal tubule obstruction). Acute tubular necrosis and acute interstitial nephritis are the more common types of intrinsic renal injury. Each type of injury has several drugs that are implicated as a possible cause, with antiinfectives being the most common. Postrenal injuries that result from obstruction block the flow of urine, leading to hydronephrosis and subsequent damage to the renal parenchyma. Drugs associated with tubular obstruction include acyclovir, methotrexate, and several antiretrovirals. Renal recovery from drug-induced AKI begins once the offending agent has been removed, if clinically possible, and is complete in most cases. It is uncommon that renal replacement therapy will be needed while recovery occurs. Pharmacists can play a pivotal role in identifying possible causes of drug-induced AKI and limit their toxic effect by identifying those most likely to cause or contribute to injury. Dose adjustment is critical during changes in renal function, and the pharmacist can ensure that optimal therapy is provided during this critical time.
Collapse
Affiliation(s)
| | - Michael L Bentley
- Department of Pharmacy, Carilion Clinic, Roanoke, VA, USA
- Department of Biomedical Science, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| |
Collapse
|
10
|
The interaction of dipole modifiers with amphotericin-ergosterol complexes. Effects of phospholipid and sphingolipid membrane composition. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2014; 43:207-15. [DOI: 10.1007/s00249-014-0946-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/30/2014] [Accepted: 02/05/2014] [Indexed: 10/25/2022]
|
11
|
Shatursky OY, Romanenko OV, Himmelreich NH. Long open amphotericin channels revealed in cholesterol-containing phospholipid membranes are blocked by thiazole derivative. J Membr Biol 2014; 247:211-29. [PMID: 24402241 DOI: 10.1007/s00232-013-9626-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/26/2013] [Indexed: 10/25/2022]
Abstract
The action of antifungal drug, amphotericin B (AmB), on solvent-containing planar lipid bilayers made of sterols (cholesterol, ergosterol) and synthetic C14-C18 tail phospholipids (PCs) or egg PC has been investigated in a voltage-clamp mode. Within the range of PCs tested, a similar increase was achieved in the lifetime of one-sided AmB channels in cholesterol- and ergosterol-containing membranes with the C16 tail PC, DPhPC at sterol/DPhPC molar ratio ≤1. The AmB channel lifetimes decreased only at sterol/DPhPC molar ratio >1 that occurred with sterol/PC molar ratio of target cell membranes at a pathological state. These data obtained on bilayer membranes two times thicker than one-sided AmB channel length are consistent with the accepted AmB pore-forming mechanism, which is associated with membrane thinning around AmB-sterol complex in the lipid rafts. Our results show that AmB can create cytotoxic (long open) channels in cholesterol membrane with C14-C16 tail PCs and nontoxic (short open) channels with C17-C18 tail PCs as the lifetime of one-sided AmB channel depends on ~2-5 Å difference in the thickness of sterol-containing C16 and C18 tail PC membranes. The reduction in toxic AmB channels efficacy can be required at the drug administration because C16 tails in native membrane PCs occur almost as often as C18 tails. The comparative analysis of AmB channel blocking by tetraethylammonium chloride, tetramethylammonium chloride and thiazole derivative of vitamin B1, 3-decyloxycarbonylmethyl-4-methyl-5-(2-hydroxyethyl) thiazole chloride (DMHT), has proved that DMHT is a comparable substitute for both tetraalkylammonia that exhibits a much higher affinity.
Collapse
Affiliation(s)
- Oleg Ya Shatursky
- Department of Neurochemistry, Palladin Institute of Biochemistry, Leontovich Str., 9, Kiev, 01601, Ukraine,
| | | | | |
Collapse
|
12
|
In vitro cytotoxicity and in vivo acute and chronic toxicity of Xanthii Fructus and its processed product. BIOMED RESEARCH INTERNATIONAL 2013; 2013:403491. [PMID: 24371823 PMCID: PMC3858965 DOI: 10.1155/2013/403491] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 10/11/2013] [Accepted: 10/11/2013] [Indexed: 11/18/2022]
Abstract
Xanthii Fructus (XF), the fruit of Xanthium sibiricum Patr., was used in the treatment of rhinitis and related nasal disease. Adverse effects of Xanthii Fructus are frequently reported these years. In the paper, in vitro renal cytotoxicity and in vivo acute and chronic toxicity researches of Xanthii Fructus (XF) and its processed product (processed Xanthii Fructus (PXF)) were carried out. Water extraction of XF displayed no cell membrane damage effects even in the highest concentration (100 μg/mL); however, it might affect the function of renal cell mitochondria. Acute toxicities were observed only in high and middle dosage groups. Fortunately, the single dose administration of XF or PXF was safe even at the highest daily dosage. Twelve-week chronic toxicity assays were performed in SD rats with low, middle, and high dosage. Notable changes in body weight and blood cell and BUN and Scr changes sporadically occurred in middle and high groups after the 9th week. Serum HA and HPCIII values were sustained increasing from the 4th week to the 8th week in Group V male rats, which indicated that the renal fibrosis risks still existed although no fibrosis was found in the pathological examination of the liver and kidney.
Collapse
|
13
|
Matsuo K, Hotokezaka H, Ohara N, Fujimura Y, Yoshimura A, Okada Y, Hara Y, Yoshida N, Nakayama K. Analysis of Amphotericin B-Induced Cell Signaling with Chemical Inhibitors of Signaling Molecules. Microbiol Immunol 2013; 50:337-47. [PMID: 16625056 DOI: 10.1111/j.1348-0421.2006.tb03792.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Although amphotericin B (AmB) is a major polyene antibiotic against invasive fungal infection, administration to patients sometimes causes inflammatory side effects, which limits the usage of the antibiotic. We studied the intracellular signaling that was induced by AmB. p65 (RelA) of nuclear factor-kappaB (NF-kappaB), a well-known signaling molecule as an inducer of proinflammatory cytokines, was phosphorylated by AmB in RAW264.7 cells, a monocyte-like cell line. Among chemical inhibitors of signaling molecules, U-73122 (phospholipase C (PLC) inhibitor), Gö6976 (protein kinase C (PKC) inhibitor), BAPTA-AM (calcium chelator), LFM-A13 (Bruton's tyrosine kinase (Btk)-specific inhibitor), and PP2 (c-Src kinase inhibitor) suppressed AmB-induced phosphorylation of p65 and translocation of p65 into the nucleus. U-73122 and Gö6976 reduced AmB-mediated induction of proinflammatory cytokines (tumor necrosis factor (TNF)-alpha and interleukin (IL)-6) in RAW264.7 cells. Furthermore, AmB-induced activation of NF-kappaB was observed in toll-like receptor (TLR) 2-expressed cells, and the activation of NF-kappaB was inhibited by U-73122, whereas peptidoglycan-induced NF-kappaB activation, which was also dependent on TLR2, was not inhibited by U-73122. Finally, U-73122 partially suppressed in vivo production of TNF-alpha and IL-6 induced by AmB administration in BALB/c mice. These results suggested that the signaling from AmB stimulation to proinflammatory cytokine production is mediated by TLR2, Btk, PLC, PKC, c-Src and NF-kappaB. These signaling molecules may become a target for chemotherapy suppressing AmB-induced proinflammatory cytokine production.
Collapse
Affiliation(s)
- Kenichiro Matsuo
- Microbiology and Oral Infection, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Wilcock BC, Endo MM, Uno BE, Burke MD. C2'-OH of amphotericin B plays an important role in binding the primary sterol of human cells but not yeast cells. J Am Chem Soc 2013; 135:8488-91. [PMID: 23718627 DOI: 10.1021/ja403255s] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Amphotericin B (AmB) is a clinically vital antimycotic but is limited by its severe toxicity. Binding ergosterol, independent of channel formation, is the primary mechanism by which AmB kills yeast, and binding cholesterol may primarily account for toxicity to human cells. The leading structural model predicts that the C2' hydroxyl group on the mycosamine appendage is critical for binding both sterols. To test this, the C2'-OH was synthetically deleted, and the sterol binding capacity of the resulting derivative, C2'deOAmB, was directly characterized via isothermal titration calorimetry. Surprisingly, C2'deOAmB binds ergosterol and, within the limits of detection of this experiment, does not bind cholesterol. Moreover, C2'deOAmB is nearly equipotent to AmB against yeast but, within the limits of detection of our assays, is nontoxic to human cells in vitro. Thus, the leading structural model for AmB/sterol binding interactions is incorrect, and C2'deOAmB is an exceptionally promising new antifungal agent.
Collapse
Affiliation(s)
- Brandon C Wilcock
- Howard Hughes Medical Institute, Roger Adams Laboratory, Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | | | | |
Collapse
|
15
|
Leon CG, Lee J, Bartlett K, Gershkovich P, Wasan EK, Zhao J, Clement JG, Wasan KM. In vitro cytotoxicity of two novel oral formulations of Amphotericin B (iCo-009 and iCo-010) against Candida albicans, human monocytic and kidney cell lines. Lipids Health Dis 2011; 10:144. [PMID: 21854638 PMCID: PMC3173361 DOI: 10.1186/1476-511x-10-144] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 08/20/2011] [Indexed: 11/13/2022] Open
Abstract
Background Invasive fungal infections such as candidiasis constitute an increasingly important medical problem. Drugs currently used for the treatment of candidiasis include polyenes (such as Amphotericin B) and azoles. Amphotericin B (AmpB) presents several limitations such as its nephrotoxicity and limited solubility. We have developed two novel lipid-based AmpB formulations which in vivo show less nephrotoxicity and enhanced solubility compared to Fungizone™ a commercial AmpB formulation. The purpose of this study was to determine the cytotoxicity of Fungizone™, Ambisome™ and two novel AmpB formulations (iCo-009 and iCo-010) against Candida albicans, human kidney (293T) cells and monocytic (THP1) cells. Methods Cell cytotoxicity to the AmpB formulations was evaluated by MTS and LDH assays. In vitro anti-Candida albicans activity was assessed after a 48 h drug incubation. Results None of the AmpB formulations tested showed cytotoxicity against 293T cells. In the case of THP1 cells only Fungizone™ and Ambisome™ showed cytotoxicity at 500 μg/L (n = 4-10, p < 0.05). The calculated EC50 to Candida albicans for the different formulations was as follows: 26.8 ± 2.9 for iCo-010, 74.6 ± 8.9 for iCo-009, 109 ± 31 for Ambisome™ and 87.1 ± 22 for Fungizone™ (μg of AmpB/L, n = 6-12, p < 0.05). Conclusions The AmpB formulations analyzed were not cytotoxic to 293T cells. Cytotoxicity in THP1 cells was observed for Fungizone™ and Ambisome™, but not with the novel AmpB formulations. iCo-010 had higher efficacy compared to other three AmpB formulations in the Candida albicans model. The absence of cytotoxicity as well as its higher efficacy for the Candida model compared to Fungizone™ and Ambisome™ suggest that iCo-010 has potential in treating candidiasis.
Collapse
Affiliation(s)
- Carlos G Leon
- Division of Pharmaceutics and Biopharmaceutics, The University of British Columbia, Vancouver British Columbia, V6T 1Z3, Canada
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Cohen BE. Amphotericin B membrane action: role for two types of ion channels in eliciting cell survival and lethal effects. J Membr Biol 2010; 238:1-20. [PMID: 21085940 DOI: 10.1007/s00232-010-9313-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 10/20/2010] [Indexed: 01/25/2023]
Abstract
The formation of aqueous pores by the polyene antibiotic amphotericin B (AmB) is at the basis of its fungicidal and leishmanicidal action. However, other types of nonlethal and dose-dependent biphasic effects that have been associated with the AmB action in different cells, including a variety of survival responses, are difficult to reconcile with the formation of a unique type of ion channel by the antibiotic. In this respect, there is increasing evidence indicating that AmB forms nonaqueous (cation-selective) channels at concentrations below the threshold at which aqueous pores are formed. The main foci of this review will be (1) to provide a summary of the evidence supporting the formation of cation-selective ion channels and aqueous pores by AmB in lipid membrane models and in the membranes of eukaryotic cells; (2) to discuss the influence of membrane parameters such as thickness fluctuations, the type of sterol present and the existence of sterol-rich specialized lipid raft microdomains in the formation process of such channels; and (3) to develop a cell model that serves as a framework for understanding how the intracellular K(+) and Na(+) concentration changes induced by the cation-selective AmB channels enhance multiple survival response pathways before they are overcome by the more sustained ion fluxes, Ca(2+)-dependent apoptotic events and cell lysis effects that are associated with the formation of AmB aqueous pores.
Collapse
Affiliation(s)
- B Eleazar Cohen
- Division of External Activities, National Institute of Allergy and Infectious Diseases, 6700B Rockledge Drive, Bethesda, MD 20982, USA.
| |
Collapse
|
17
|
Zager RA, Johnson ACM, Naito M, Bomsztyk K. Maleate nephrotoxicity: mechanisms of injury and correlates with ischemic/hypoxic tubular cell death. Am J Physiol Renal Physiol 2007; 294:F187-97. [PMID: 17942567 DOI: 10.1152/ajprenal.00434.2007] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Maleate injection causes dose-dependent injury in proximal tubular cells. This study sought to better define underlying pathogenic mechanisms and to test whether maleate toxicity recapitulates critical components of the hypoxic/ischemic renal injury cascade. CD-1 mice were injected with maleate or used as a source for proximal tubule segments (PTS) for in vitro studies. Maleate induced dose-dependent PTS injury [lactate deydrogenase (LDH) release, ATP reductions, nonesterified fatty acid (NEFA) accumulation]. These changes were partially dependent on maleate metabolism (protection conferred by metabolic inhibitors: succinate, acetoacetate). Maleate toxicity reproduced critical characteristics of the hypoxia/ATP depletion-induced injury cascade: 1) glutathione (GSH) conferred protection, but due to its glycine, not cysteine (antioxidant), content; 2) ATP reductions reflected decreased production, not Na-K-ATPase-driven increased consumption; 3) cell death was completely blocked by extracellular acidosis (pH 6.6); 4) intracellular Ca(2+) chelation (BAPTA) mitigated cell death; 5) maleate and hypoxia each caused plasma membrane cholesterol shedding and in both instances, this was completely glycine suppressible; 6) maleate + hypoxia caused neither additive NEFA accumulation nor LDH release, implying shared pathogenic pathways; and 7) maleate, like ischemia, induced renal cortical cholesterol loading; increased HMG CoA reductase (HMGCR) activity (statin inhibitable), increased HMGCR mRNA levels, and increased RNA polymerase II recruitment to the HMGCR locus (chromatin immunoprecipitation, ChIP, assay) were involved. These results further define critical determinants of maleate nephrotoxicity and suggest that it can serve as a useful adjunct for studies of ischemia/ATP depletion-induced, proximal tubule-specific, cell death.
Collapse
|
18
|
Wen YJ, Su T, Tang JW, Zhang CY, Wang X, Cai SQ, Li XM. Cytotoxicity of Phenanthrenes Extracted from Aristolochia contorta in Human Proximal Tubular Epithelial Cell Line. ACTA ACUST UNITED AC 2006; 103:e95-e102. [PMID: 16554666 DOI: 10.1159/000092194] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2005] [Accepted: 11/08/2005] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Aristolochic acid nephropathy, a progressive tubulointerstitial renal disease, is predominantly a result of aristolochic acid I (AA-I) intoxication. However, other unidentified phytotoxins have indeed been postulated as the cause of this unique interstitial nephropathy. The purpose of this study was to investigate the cytotoxicity of other phenanthrene derivatives extracted from Aristolochia contorta in the human proximal tubular epithelial cell line HK-2. METHODS After HK-2 cells were incubated with an indicated concentration of test compounds for 24 h, cell viability was assessed by lactate dehydrogenase (LDH) leakage assay (cell membrane damage) in combination with MTT assay (metabolic capability). Cellular morphologic assessments were performed with a phase-contrast inverted microscope and transmission electron microscope. RESULTS In all test compounds at 5 microg/ml, AA-I, 7-methoxy-aristololactam IV and aristololactam IVa showed cytotoxic activity in HK-2 cells in both MTT assay and LDH leakage assay (p < 0.01). At high concentration (5-80 microg/ml), these three compounds caused a dose-dependent decrease in MTT reduction and a dose-dependent increase in LDH leakage compared to non-treated cells (p <0.01). In LDH leakage assay, 40 mug/ml 7-methoxy-aristololactam IV induced a 1.58-fold LDH leakage compared to AA-I at the same concentration (p < 0.01). Moreover, the IC50 of these three compounds were 16.675 microg/ml for AA-I, 4.535 microg/ml for 7-methoxy-aristololactam IV, and 30.244 microg/ml for aristololactam IVa in MTT assay. The cellular morphologic assessments suggest interactions with cell membrane and intracellular structures such as lysosome and mitochondria are likely to be involved in cell injury induced by these three compounds. CONCLUSION The potency of cytotoxic activity of aristololactam IVa and 7-methoxy-aristololactam IV extracted from A. contorta is similar to or even stronger than that of AA-I.
Collapse
Affiliation(s)
- Yu-jie Wen
- Renal Division, First Hospital, Peking University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Dabur R, Diwedi SK, Yadav V, Mishra V, Singh R, Singh H, Sharma GL. Efficacy of 2-(3,4-dimethyl-2,5-dihydro-1h-pyrrole-2-yl)-1-methylethyl pentanoate in a murine model of invasive aspergillosis. Antimicrob Agents Chemother 2005; 49:4365-7. [PMID: 16189123 PMCID: PMC1251545 DOI: 10.1128/aac.49.10.4365-4367.2005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
2-(3,4-Dimethyl-2,5-dihydro-1H-pyrrole-2-yl)-1-methylethyl pentanoate, an antifungal compound, was found to be nontoxic to RAW cells up to a concentration of 312.5 microg/ml, whereas amphotericin B was lethal to all cells at 37.5 microg/ml. The treatment of Aspergillus fumigatus-infected mice with a dose of 200.0 mg of compound/kg of body weight increased their survival rate by 60%, with a decrease in CFU in organ tissues. The protection afforded by the compound against experimental aspergillosis was found to be dose dependent.
Collapse
Affiliation(s)
- Rajesh Dabur
- Institute of Biomedical Sciences, Bundelkhand University, Jhansi, India
| | | | | | | | | | | | | |
Collapse
|
20
|
Leon C, Taylor R, Bartlett KH, Wasan KM. Effect of heat-treatment and the role of phospholipases on Fungizone®-induced cytotoxicity within human kidney proximal tubular (HK-2) cells and Aspergillus fumigatus. Int J Pharm 2005; 298:211-8. [PMID: 15950412 DOI: 10.1016/j.ijpharm.2005.04.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2005] [Revised: 04/27/2005] [Accepted: 04/27/2005] [Indexed: 11/17/2022]
Abstract
The objectives of this study were to determine the effects of heat-treatment on Fungizone (FZ)-induced cytotoxicity in human kidney (HK-2) cells and fungal isolates of Aspergillus fumigatus, and to determine the possible role of phospholipases (PLA2 and PLC) on heat-treated FZ (HFZ)-associated renal cell toxicity. HK-2 cells were grown at 37 degrees C in T75 flasks and seeded in 96-well plates at 20,000 cells/well. FZ and HFZ concentrations of 10, 25 and 50 microg/mL of AmpB were prepared. Snake venom PLA2 and PLC (2.15 U/mL) were pre-incubated with HFZ for 1h prior to addition to the cells. After 18 h of incubation, an MTS assay was performed to assess cell viability through mitochondrial respiration. A spore suspension of A. fumigatus was prepared and 96-well plates were seeded at 500,000 spores/well. HFZ and FZ were prepared as above and incubated with the fungi at 35 degrees C. After 72 h, the minimum inhibitory concentration (MIC) was determined as the lowest concentration of drug that inhibited visible growth. Student-Newman-Keuls multiple comparisons tests were conducted to determine statistical significance. FZ-induced cytotoxicity was significantly greater than for HFZ in HK-2 cells at amphotericin B (AmpB) concentrations between 10 and 50 microg AmpB/mL (n = 5-9, p < 0.05). HFZ and FZ were found to have similar minimum inhibitory concentration (MIC) ranges for A. fumigatus (0.225-0.25 microg) AmpB/mL; (n = 6). The addition of PLA2 and PLC to 50 microg heat-treated AmpB/mL significantly enhanced the cytotoxicity compared to controls (n = 6, p < 0.05). The presence of the phospholipases did not alter FZ-associated renal cell toxicity. Taken together, these findings suggest heat-treatment significantly decreased FZ-induced cytotoxicity in HK-2 cells without altering toxicity against a reference strain of A. fumigatus. In addition, PLA2 and PLC enhanced the renal toxicity associated with HFZ, but not that of FZ.
Collapse
Affiliation(s)
- Carlos Leon
- Division of Pharmaceutics and Biopharmaceutics, Faculty of Pharmaceutical Sciences, The University of British Columbia, 2146 East Mall, Vancouver, BC, Canada V6T 1Z3
| | | | | | | |
Collapse
|
21
|
Lemke A, Kiderlen AF, Kayser O. Amphotericin B. Appl Microbiol Biotechnol 2005; 68:151-62. [PMID: 15821914 DOI: 10.1007/s00253-005-1955-9] [Citation(s) in RCA: 223] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2005] [Revised: 02/28/2005] [Accepted: 03/01/2005] [Indexed: 10/25/2022]
Abstract
Invasive fungal infections are a major cause of morbidity and mortality in immunodeficient individuals (such as AIDS patients) and in transplant recipients or tumor patients undergoing immunosuppressive chemotherapy. Amphotericin B is one of the oldest, yet most efficient antimycotic agents. However, its usefulness is limited due to dose-dependent side-effects, notably nephrotoxicity. In order to improve its safety margin, new pharmaceutical formulations of amphotericin B have been designed especially to reduce its detrimental effects on the kidneys. Since the 1980s, a wide variety of new amphotericin B formulations have been brought forward for clinical testing, many of which were approved and reached market value in the 1990s. This review describes and discusses the molecular genetics, pharmacological, toxicological, and clinical aspects of amphotericin B itself and many of its innovative formulations.
Collapse
Affiliation(s)
- A Lemke
- Institute of Pharmacy, Pharmaceutical Technology, Biotechnology, and Quality Management, Freie Universität Berlin, Berlin 12169, Germany
| | | | | |
Collapse
|
22
|
Orce G, Castillo G, Chanampa Y, Bellomio A. Permeability to water in a tight epithelium: possible modulating action of gap junctions. Can J Physiol Pharmacol 2005; 82:417-21. [PMID: 15381966 DOI: 10.1139/y04-037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Osmotic water flow (Jw) across tight distal nephron epithelial membranes increases upon exposure to vasopressin: following binding of the hormone to its receptors, intracellular cyclic AMP concentration increases, leading to insertion of aquaporins in the apical membrane. The involvement of intercellular communication in the process, however, has not been adequately explored. Octanol, 1.2 x 10(-3) M, a gap junction inhibitor, significantly reduced Jw (expressed as mg.20 min(-1)) in isolated toad urinary bladders (a model of the distal nephron) subjected to a transepithelial osmotic gradient and exposed to agents mimicking the vasopressin-triggered mechanism: oxytocin, 50 mIU.mL(-1) (from 185.3 +/- 28.0, P < 0.001, to 69.0 +/- 23.6, P < 0.05; Pdiff < 0.01, n = 6), and cyclic AMP, 2.5 x 10(-3) M (from 98.0 +/- 32.6, P < 0.02, to 31.0 +/- 13.9, NS; Pdiff < 0.05, n = 12), without altering the effect of nystatin, 450 U.mL(-1), which increases Jw via a mechanism unrelated to apical aquaporin insertion (163.2 +/- 16.3, P < 0.001, in controls vs. 150.3 +/- 10.4, P < 0.001, in octanol-treated bladders; Pdiff: NS, n = 6). Another gap junction blocker, carbenoxolone, 2.0 x 10(-4) M (CBX), exerted similar effects on the responses to oxytocin, 100 mIU.mL(-1), reducing the response from 256.7 +/- 33.6, P < 0.001, to 102.7 +/- 10.4, P < 0.001; Pdiff < 0.01, n = 6) and nystatin, which was unaffected (95.0 +/- 20.9, P < 0.01, vs. 132.0 +/- 27.0, P < 0.01; Pdiff: NS, n = 6). Our results suggest that either gap junctions or, alternatively, unopposed gap junction hemichannels, may be important in the regulation of Jw in the isolated toad bladder, by modulating a step in the physiological process leading to increased apical membrane permeability.
Collapse
Affiliation(s)
- Gabriel Orce
- Departamento de Fisiología y Neurociencia, INSIBIO (UNT-CONICET), Instituto de Fisiología-Facultad de Medicina, Tucumán, Argentina.
| | | | | | | |
Collapse
|
23
|
Groll AH, Mickiene D, Petraitis V, Petraitiene R, Alfaro RM, King C, Piscitelli SC, Walsh TJ. Comparative drug disposition, urinary pharmacokinetics, and renal effects of multilamellar liposomal nystatin and amphotericin B deoxycholate in rabbits. Antimicrob Agents Chemother 2004; 47:3917-25. [PMID: 14638502 PMCID: PMC296179 DOI: 10.1128/aac.47.12.3917-3925.2003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The comparative drug dispositions, urinary pharmacokinetics, and effects on renal function of multilamellar liposomal nystatin (LNYS; Nyotran) and amphotericin B deoxycholate (DAMB; Fungizone) were studied in rabbits. Drug concentrations were determined by high-performance liquid chromatography as total concentrations of LNYS and DAMB. In comparison to a standard dose of 1 mg of DAMB/kg of body weight, therapeutic dosages of LNYS, i.e., 2, 4, and 6 mg/kg, resulted in escalating maximum concentrations (Cmax) (17 to 56 microg/ml for LNYS versus 3.36 microg/ml for DAMB; P<0.001) and values for the area under the concentration-time curve from 0 to 24 h (AUC(0-24)) (17 to 77 microg.h/ml for LNYS versus 12 microg.h/ml for DAMB; P<0.001) in plasma but a significantly faster total clearance from plasma (0.117 to 0.080 liter/h/kg for LNYS versus 0.055 liter/h/kg for DAMB; P=0.013) and a < or =8-fold-smaller volume of distribution at steady state (P=0.002). Urinary drug concentration data revealed a > or =10-fold-higher Cmax (16 to 10 microg/ml for LNYS versus 0.96 microg/ml for DAMB; P=0.015) and a 4- to 7-fold-greater AUC(0-24) (63 to 35 microg.h/ml for LNYS versus 8.9 microg.h/ml for DAMB; P=0.015) following the administration of LNYS, with a dose-dependent decrease in the dose-normalized AUC(0-24) in urine (P=0.001) and a trend toward a dose-dependent decrease in renal clearance. Except for the kidneys, the mean concentrations of LNYS in liver, spleen, and lung 24 h after dosing were severalfold lower than those after administration of DAMB (P, <0.002 to <0.001). Less than 1% each of the total dose of LNYS was recovered from the kidneys, liver, spleen, and lungs; in contrast, a quarter of the total dose was recovered from the livers of DAMB-treated animals. LNYS had dose-dependent effects on glomerular filtration and distal, but not proximal, renal tubular function which did not exceed those of DAMB at the highest investigated dosage of 6 mg/kg. The results of this experimental study demonstrate fundamental differences in the dispositions of LNYS and DAMB. Based on its enhanced urinary exposure, LNYS may offer a therapeutic advantage in systemic fungal infections involving the upper and lower urinary tracts that require therapy with antifungal polyenes.
Collapse
Affiliation(s)
- Andreas H Groll
- Immunocompromised Host Section, Pediatric Oncology Branch, National Cancer Institute, Warren Grant Magnuson Clinical Center, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Zager RA, Johnson ACM, Hanson SY, Wasse H. Parenteral iron formulations: a comparative toxicologic analysis and mechanisms of cell injury. Am J Kidney Dis 2002; 40:90-103. [PMID: 12087566 DOI: 10.1053/ajkd.2002.33917] [Citation(s) in RCA: 166] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Multiple parenteral iron (Fe) formulations exist for administration to patients with end-stage renal disease. Although there are concerns regarding their potential toxicities, no direct in vitro comparisons of these agents exist. Thus, the present study contrasted pro-oxidant and cytotoxic potentials of four available Fe preparations: Fe dextran (Fe dext), Fe sucrose (Fe sucr), Fe gluconate (Fe gluc), and Fe oligosaccharide (Fe OS). METHODS Differing dosages (0.06 to 1 mg/mL) of each compound were added to either (1) isolated mouse proximal tubule segments, (2) renal cortical homogenates, or (3) cultured human proximal tubule (HK-2) cells (0.5- to 72-hour incubations). Oxidant injury (malondialdehyde generation) and lethal cell injury (percentage of lactate dehydrogenase release; tetrazolium dye uptake) were assessed. Effects of selected antioxidants (glutathione [GSH], catalase, dimethylthiourea (DMTU), and sodium benzoate also were assessed. RESULTS Each test agent induced massive and similar degrees of lipid peroxidation. Nevertheless, marked differences in cell death resulted (Fe sucr >> Fe gluc > Fe dext approximately Fe OS). This relative toxicity profile also was observed in cultured aortic endothelial cells. Catalase, DMTU, and sodium benzoate conferred no protection. However, GSH and its constituent amino acid glycine blocked Fe sucr-mediated cell death. The latter was mediated by mitochondrial blockade, causing free radical generation and a severe adenosine triphosphate depletion state. CONCLUSIONS (1) parenteral Fes are highly potent pro-oxidants and capable of inducing tubular and endothelial cell death, (2) markedly different toxicity profiles exist among these agents, and (3) GSH can exert protective effects. However, the latter stems from GSH's glycine content, rather than from a direct antioxidant effect.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/pathology
- Cell Division/drug effects
- Cell Line
- Cell Line, Transformed
- Cell Survival/drug effects
- Chlorides
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Ferric Compounds/metabolism
- Ferric Compounds/toxicity
- Ferric Oxide, Saccharated
- Glucaric Acid
- Humans
- Iron/metabolism
- Iron/toxicity
- Iron-Dextran Complex/metabolism
- Iron-Dextran Complex/toxicity
- Kidney Failure, Chronic/metabolism
- Kidney Failure, Chronic/pathology
- Kidney Failure, Chronic/therapy
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Lipid Peroxidation/drug effects
- Male
- Malondialdehyde/blood
- Mice
- Oligosaccharides/metabolism
- Oligosaccharides/toxicity
- Oxidants/metabolism
- Oxidants/toxicity
- Parenteral Nutrition/methods
Collapse
Affiliation(s)
- Richard A Zager
- Department of Medicine, University of Washington, Seattle, WA, USA.
| | | | | | | |
Collapse
|
25
|
Ito Y, Nakayama S, Son M, Kume H, Yamaki K. Protection by tetracyclines against ion transport disruption caused by nystatin in human airway epithelial cells. Toxicol Appl Pharmacol 2001; 177:232-7. [PMID: 11749122 DOI: 10.1006/taap.2001.9313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Polyene antifungal antibiotics like nystatin form monovalent cation pores on the plasma membrane that perturb the intracellular electrolyte milieu, resulting in cell damage. In the present study, we investigated the effects of tetracyclines (minocycline and tetracycline) on ion transporters disrupted by nystatin in cultured human airway Calu-3 cells. Apical application of nystatin (50 microM) on a monolayer of the cells stimulated Na(+)-K(+) pump activity as estimated by ouabain (1 mM)-sensitive short-circuit current (I(sc)). The nystatin-potentiated I(sc) was inhibited by minocycline (IC(50) = 25 microM) or tetracycline (IC(50) = 150 microM) applied only from the apical (nystatin-treated) side. Nystatin increased monolayer conductance that was reversed by the application of tetracyclines. In contrast, ouabain potentiated the nystatin-induced change in the conductance. Further, Na(+)-glucose transport affected by nystatin was also normalized by tetracyclines from the nystatin-treated side of the membrane. These data suggest that tetracyclines may lower the cell permeability potentiated by nystatin, protecting cells against damage.
Collapse
Affiliation(s)
- Y Ito
- Second Division (Respiratory Division), First Department of Physiology, School of Medicine, Nagoya University, Tsurumai-cho, Nagoya, 466-8550, Japan.
| | | | | | | | | |
Collapse
|