1
|
Xisto MIDDS, Rollin-Pinheiro R, Rochetti VP, de Castro-Almeida Y, Borba-Santos LP, dos Santos-Freitas GMP, Cypriano J, Abreu FDÁ, Rozental S, Barreto-Bergter E. Miltefosine: A Repurposing Drug against Mucorales Pathogens. J Fungi (Basel) 2023; 9:1166. [PMID: 38132767 PMCID: PMC10744482 DOI: 10.3390/jof9121166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/27/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
Mucorales are a group of non-septated filamentous fungi widely distributed in nature, frequently associated with human infections, and are intrinsically resistant to many antifungal drugs. For these reasons, there is an urgent need to improve the clinical management of mucormycosis. Miltefosine, which is a phospholipid analogue of alkylphosphocholine, has been considered a promising repurposing drug to be used to treat fungal infections. In the present study, miltefosine displayed antifungal activity against a variety of Mucorales species, and it was also active against biofilms formed by these fungi. Treatment with miltefosine revealed modifications of cell wall components, neutral lipids, mitochondrial membrane potential, cell morphology, and the induction of oxidative stress. Treated Mucorales cells also presented an increased susceptibility to SDS. Purified ergosterol and glucosylceramide added to the culture medium increased miltefosine MIC, suggesting its interaction with fungal lipids. These data contribute to elucidating the effect of a promising drug repurposed to act against some relevant fungal pathogens that significantly impact public health.
Collapse
Affiliation(s)
- Mariana Ingrid Dutra da Silva Xisto
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (V.P.R.); (Y.d.C.-A.); (G.M.P.d.S.-F.)
| | - Rodrigo Rollin-Pinheiro
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (V.P.R.); (Y.d.C.-A.); (G.M.P.d.S.-F.)
| | - Victor Pereira Rochetti
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (V.P.R.); (Y.d.C.-A.); (G.M.P.d.S.-F.)
| | - Yuri de Castro-Almeida
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (V.P.R.); (Y.d.C.-A.); (G.M.P.d.S.-F.)
| | - Luana Pereira Borba-Santos
- Laboratório de Biologia Celular de Fungos, Programa de Biologia Celular e Parasitologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.P.B.-S.); (S.R.)
| | - Giulia Maria Pires dos Santos-Freitas
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (V.P.R.); (Y.d.C.-A.); (G.M.P.d.S.-F.)
| | - Jefferson Cypriano
- Laboratório de Biologia Celular e Magnetotaxia & Unidade de Microscopia Multiusuário, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.C.); (F.d.Á.A.)
| | - Fernanda de Ávila Abreu
- Laboratório de Biologia Celular e Magnetotaxia & Unidade de Microscopia Multiusuário, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.C.); (F.d.Á.A.)
| | - Sonia Rozental
- Laboratório de Biologia Celular de Fungos, Programa de Biologia Celular e Parasitologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.P.B.-S.); (S.R.)
| | - Eliana Barreto-Bergter
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (V.P.R.); (Y.d.C.-A.); (G.M.P.d.S.-F.)
| |
Collapse
|
2
|
Carnielli JB, Dave A, Romano A, Forrester S, de Faria PR, Monti-Rocha R, Costa CH, Dietze R, Graham IA, Mottram JC. 3'Nucleotidase/nuclease is required for Leishmania infantum clinical isolate susceptibility to miltefosine. EBioMedicine 2022; 86:104378. [PMID: 36462405 PMCID: PMC9713291 DOI: 10.1016/j.ebiom.2022.104378] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 10/20/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Miltefosine treatment failure in visceral leishmaniasis in Brazil has been associated with deletion of the miltefosine susceptibility locus (MSL) in Leishmania infantum. The MSL comprises four genes, 3'-nucleotidase/nucleases (NUC1 and NUC2); helicase-like protein (HLP); and 3,2-trans-enoyl-CoA isomerase (TEI). METHODS In this study CRISPR-Cas9 was used to either epitope tag or delete NUC1, NUC2, HLP and TEI, to investigate their role in miltefosine resistance mechanisms. Additionally, miltefosine transporter genes and miltefosine-mediated reactive oxygen species homeostasis were assessed in 26 L. infantum clinical isolates. A comparative lipidomic analysis was also performed to investigate the molecular basis of miltefosine resistance. FINDINGS Deletion of both NUC1, NUC2 from the MSL was associated with a significant decrease in miltefosine susceptibility, which was restored after re-expression. Metabolomic analysis of parasites lacking the MSL or NUC1 and NUC2 identified an increase in the parasite lipid content, including ergosterol; these lipids may contribute to miltefosine resistance by binding the drug in the membrane. Parasites lacking the MSL are more resistant to lipid metabolism perturbation caused by miltefosine and NUC1 and NUC2 are involved in this pathway. Additionally, L. infantum parasites lacking the MSL isolated from patients who relapsed after miltefosine treatment were found to modulate nitric oxide accumulation in host macrophages. INTERPRETATION Altogether, these data indicate that multifactorial mechanisms are involved in natural resistance to miltefosine in L. infantum and that the absence of the 3'nucleotidase/nuclease genes NUC1 and NUC2 contributes to the phenotype. FUNDING MRC GCRF and FAPES.
Collapse
Affiliation(s)
- Juliana B.T. Carnielli
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom,Laboratório de Leishmanioses, Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória-ES, Brazil,Corresponding author. York Biomedical Research Institute, Department of Biology, University of York, Wentworth Way Heslington, York, YO10 5DD, United Kingdom.
| | - Anuja Dave
- Centre for Novel Agricultural Products, Department of Biology, University of York, United Kingdom
| | - Audrey Romano
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Sarah Forrester
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Pedro R. de Faria
- Laboratório de Leishmanioses, Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória-ES, Brazil
| | - Renata Monti-Rocha
- Laboratório de Leishmanioses, Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória-ES, Brazil
| | - Carlos H.N. Costa
- Laboratório de Pesquisas em Leishmanioses, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Teresina-PI, Brazil
| | - Reynaldo Dietze
- Laboratório de Leishmanioses, Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória-ES, Brazil,Global Health & Tropical Medicine—Instituto de Higiene e Medicina Tropical—Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ian A. Graham
- Centre for Novel Agricultural Products, Department of Biology, University of York, United Kingdom
| | - Jeremy C. Mottram
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom,Corresponding author. York Biomedical Research Institute, Department of Biology, University of York, Wentworth Way Heslington, York, YO10 5DD, United Kingdom.
| |
Collapse
|
3
|
Synthesis, Leishmanicidal, Trypanocidal, Antiproliferative Assay and Apoptotic Induction of (2-Phenoxypyridin-3-yl)naphthalene-1(2 H)-one Derivatives. Molecules 2022; 27:molecules27175626. [PMID: 36080388 PMCID: PMC9457600 DOI: 10.3390/molecules27175626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
The coexistence of leishmaniasis, Chagas disease, and neoplasia in endemic areas has been extensively documented. The use of common drugs in the treatment of these pathologies invites us to search for new molecules with these characteristics. In this research, we report 16 synthetic chalcone derivatives that were investigated for leishmanicidal and trypanocidal activities as well as for antiproliferative potential on eight human cancers and two nontumor cell lines. The final compounds 8−23 were obtained using the classical base-catalyzed Claisen−Schmidt condensation. The most potent compounds as parasiticidal were found to be 22 and 23, while compounds 18 and 22 showed the best antiproliferative activity and therapeutic index against CCRF-CEM, K562, A549, and U2OS cancer cell lines and non-toxic VERO, BMDM, MRC-5, and BJ cells. In the case of K562 and the corresponding drug-resistant K562-TAX cell lines, the antiproliferative activity has shown a more significant difference for compound 19 having 10.3 times higher activity against the K562-TAX than K562 cell line. Flow cytometry analysis using K562 and A549 cell lines cultured with compounds 18 and 22 confirmed the induction of apoptosis in treated cells after 24 h. Based on the structural analysis, these chalcones represent new compounds potentially useful for Leishmania, Trypanosoma cruzi, and some cancer treatments.
Collapse
|
4
|
Çetinel ZÖ, Bilge D. The effects of miltefosine on the structure and dynamics of DPPC and DPPS liposomes mimicking normal and cancer cell membranes: FTIR and DSC studies. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
5
|
Fanani ML, Nocelli NE, Zulueta Díaz YDLM. What can we learn about amphiphile-membrane interaction from model lipid membranes? BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2022; 1864:183781. [PMID: 34555419 DOI: 10.1016/j.bbamem.2021.183781] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/30/2021] [Accepted: 09/14/2021] [Indexed: 10/20/2022]
Abstract
Surface-active amphiphiles find applications in a wide range of areas of industry such as agrochemicals, personal care, and pharmaceuticals. In many of these applications, interaction with cell membranes is a key factor for achieving their purpose. How do amphiphiles interact with lipid membranes? What are their bases for membrane specificity? Which biophysical properties of membranes are susceptible to modulation by amphiphilic membrane-effectors? What aspects of this interaction are important for performing their function? In our work on membrane biophysics over the years, questions like these have arisen and we now share some of our findings and discuss them in this review. This topic was approached focusing on the membrane properties and their alterations rather than on the amphiphile structure requirements for their interaction. Here, we do not aim to provide a comprehensive list of the modes of action of amphiphiles of biological interest but to help in understanding them.
Collapse
Affiliation(s)
- Maria Laura Fanani
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Córdoba, Argentina.
| | - Natalia E Nocelli
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Córdoba, Argentina
| | - Yenisleidy de Las Mercedes Zulueta Díaz
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Córdoba, Argentina
| |
Collapse
|
6
|
Bulté D, Van Bockstal L, Dirkx L, Van den Kerkhof M, De Trez C, Timmermans JP, Hendrickx S, Maes L, Caljon G. Miltefosine enhances infectivity of a miltefosine-resistant Leishmania infantum strain by attenuating its innate immune recognition. PLoS Negl Trop Dis 2021; 15:e0009622. [PMID: 34292975 PMCID: PMC8330912 DOI: 10.1371/journal.pntd.0009622] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/03/2021] [Accepted: 07/01/2021] [Indexed: 01/08/2023] Open
Abstract
Background Miltefosine (MIL) is currently the only oral drug available to treat visceral leishmaniasis but its use as first-line monotherapy has been compromised by an increasing treatment failure. Despite the scarce number of resistant clinical isolates, MIL-resistance by mutations in a single aminophospholipid transporter gene can easily be selected in a laboratory environment. These mutations result in a reduced survival in the mammalian host, which can partially be restored by exposure to MIL, suggesting a kind of drug-dependency. Methodology/Principal findings To enable a combined study of the infection dynamics and underlying immunological events for differential in vivo survival, firefly luciferase (PpyRE9) / red fluorescent protein (DsRed) double-reporter strains were generated of MIL-resistant (MIL-R) and syngeneic MIL-sensitive (MIL-S) Leishmania infantum. Results in C57Bl/6 and BALB/c mice show that MIL-R parasites induce an increased innate immune response that is characterized by enhanced influx and infection of neutrophils, monocytes and dendritic cells in the liver and elevated serum IFN-γ levels, finally resulting in a less efficient establishment in liver macrophages. The elevated IFN-γ levels were shown to originate from an increased response of hepatic NK and NKT cells to the MIL-R parasites. In addition, we demonstrated that MIL could increase the in vivo fitness of MIL-R parasites by lowering NK and NKT cell activation, leading to a reduced IFN-γ production. Conclusions/Significance Differential induction of innate immune responses in the liver was found to underlie the attenuated phenotype of a MIL-R parasite and its peculiar feature of drug-dependency. The impact of MIL on hepatic NK and NKT activation and IFN-γ production following recognition of a MIL-R strain indicates that this mechanism may sustain infections with resistant parasites and contribute to treatment failure. Visceral leishmaniasis is a neglected tropical disease that is fatal if left untreated. Miltefosine is currently the only oral drug available but is increasingly failing to cure patients, resulting in its discontinuation as first-line drug in some endemic areas. To understand these treatment failures, we investigated the complex interplay of the parasite with the host immune system in the presence and absence of miltefosine. Our data indicate that miltefosine-resistant Leishmania parasites become severely hampered in their in vivo infectivity, which could be attributed to the induction of a pronounced innate immune response. Interestingly, the infection deficit was partially restored in the presence of miltefosine. Our results further indicate that miltefosine can exacerbate infections with resistant parasites by reducing innate immune recognition. This study provides new insights into the complex interplay between parasite, drug and host and discloses an immune-related mechanism of treatment failure.
Collapse
Affiliation(s)
- Dimitri Bulté
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Lieselotte Van Bockstal
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Laura Dirkx
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Magali Van den Kerkhof
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Carl De Trez
- Vrije Universiteit Brussel, Laboratory for Cellular and Molecular Immunology (CMIM), Brussels, Belgium
| | - Jean-Pierre Timmermans
- University of Antwerp, Department of Veterinary Sciences, Laboratory of Cell biology & Histology, Wilrijk, Belgium
| | - Sarah Hendrickx
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Louis Maes
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Guy Caljon
- University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
- * E-mail:
| |
Collapse
|
7
|
Zulueta Díaz YDLM, Ambroggio EE, Fanani ML. Miltefosine inhibits the membrane remodeling caused by phospholipase action by changing membrane physical properties. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183407. [DOI: 10.1016/j.bbamem.2020.183407] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 01/04/2023]
|
8
|
Ahmed H, Curtis CR, Tur-Gracia S, Olatunji TO, Carter KC, Williams RAM. Drug combinations as effective anti-leishmanials against drug resistant Leishmania mexicana. RSC Med Chem 2020; 11:905-912. [PMID: 33479685 DOI: 10.1039/d0md00101e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022] Open
Abstract
Leishmania is a parasite that causes the disease leishmaniasis, and 700 000 to 1 million new cases occur each year. There are few drugs that treat the disease and drug resistance in the parasite limits the clinical utility of existing drugs. One way to combat drug resistance is to use combination therapy rather than monotherapy. In this study we have compared the effect of single and combination treatments with four different compounds, i.e. alkylphosphocholine analogues APC12 and APC14, miltefosine (MIL), ketoconazole (KTZ), and amphotericin B (AmpB), on the survival of Leishmania mexicana wild-type promastigotes and a cell line derived from the WT with induced resistance to APC12 (C12Rx). The combination treatment with APC14 and APC16 had a synergistic effect in killing the WT while the combination treatment with KTZ and APC12 or APC14 or APC12 and APC14 had a synergistic effect against C12Rx. More than 90% killing efficiency was obtained using APC12 alone at >1 mg ml-1 against the C12Rx strain; however, combinations with APC14 produced a similar killing efficiency using APC12 at 0.063-0.25 mg ml-1 and APC14 at 0.003-0.5 mg ml-1. These results show that combination therapy can negate induced drug resistance in L. mexicana and that the use of this type of screening system could accelerate the development of drug combinations for clinical use.
Collapse
Affiliation(s)
- Humera Ahmed
- University of the West of Scotland Paisley Campus , UK .
| | | | | | | | | | | |
Collapse
|
9
|
Abstract
Outbreaks of trichinellosis caused by Trichinella papuae have been reported in South-East Asia. Mebendazole and thiabendazole are the treatments of choice for trichinellosis; however, both drugs result in significant side effects and are less effective for muscle-stage larvae (L1). An alternative therapeutic agent is needed to improve treatment. Information on lipid composition and metabolic pathways may bridge gaps in our knowledge and lead to new antiparasitics. The T. papuae L1 lipidome was analysed using a mass spectrometry-based approach, and 403 lipid components were identified. Eight lipid classes were found and glycerophospholipids were dominant, corresponding to 63% of total lipids, of which the glycerolipid DG (20:1[11Z]/22:4[7Z,10Z,13Z,16Z]/0:0) (iso2) was the most abundant. Overall, 57% of T. papuae lipids were absent in humans; therefore, lipid metabolism may be dissimilar in the two species. Proteins involved T. papuae lipid metabolism were explored using bioinformatics. We found that 4-hydroxybutyrate coenzyme A transferase, uncharacterized protein (A0A0V1MCB5) and ML-domain-containing protein are not present in humans. T. papuae glycerophospholipid metabolic and phosphatidylinositol dephosphorylation processes contain several proteins that are dissimilar to those in humans. These findings provide insights into T. papuae lipid composition and metabolism, which may facilitate the development of novel trichinellosis treatments.
Collapse
|
10
|
Reimão JQ, Pita Pedro DP, Coelho AC. The preclinical discovery and development of oral miltefosine for the treatment of visceral leishmaniasis: a case history. Expert Opin Drug Discov 2020; 15:647-658. [PMID: 32202449 DOI: 10.1080/17460441.2020.1743674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Visceral leishmaniasis (VL) is a vector-borne disease caused by Leishmania donovani or Leishmania infantum. Closely related to poverty, VL is fatal and represents one of the main burdens on public health in developing countries. Treatment of VL relies exclusively on chemotherapy, a strategy still experiencing numerous limitations. Miltefosine (MF) has been used in the chemotherapy of VL in some endemic areas, and has been expanded to other regions, being considered crucial in eradication programs. AREAS COVERED This article reviews the most relevant preclinical and clinical aspects of MF, its mechanism of action and resistance to Leishmania parasites, as well as its limitations. The authors also give their perspectives on the treatment of VL. EXPERT OPINION The discovery of MF represented an enormous advance in the chemotherapy of VL, since it was the first oral drug for this neglected disease. Beyond selection of resistant parasites due to drug pressure, several other factors can lead to treatment failure such as, for example, factors intrinsic to the host, parasite and the drug itself. Although its efficacy as a monotherapy has reduced over recent years, MF is still an important alternative in VL chemotherapy, especially when used in combination with other drugs.
Collapse
Affiliation(s)
- Juliana Q Reimão
- Departamento de Morfologia e Patologia Básica, Faculdade de Medicina de Jundiaí , Jundiaí, Brazil
| | - Débora P Pita Pedro
- Departamento de Morfologia e Patologia Básica, Faculdade de Medicina de Jundiaí , Jundiaí, Brazil
| | - Adriano C Coelho
- Departamento de Biologia Animal, Instituto de Biologia, Universidade Estadual de Campinas , Campinas, Brazil
| |
Collapse
|
11
|
Chandan S, Umesha S, Prasad KS, Balamurugan V, Chandrashekar S, Santosh Kumar SR, Ramu R, Shirahatti P, Syed A, Elgorban A. Potential antileptospiral constituents from Phyllanthus amarus. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_10_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
12
|
Malli S, Pomel S, Ayadi Y, Deloménie C, Da Costa A, Loiseau PM, Bouchemal K. Topically Applied Chitosan-Coated Poly(isobutylcyanoacrylate) Nanoparticles Are Active Against Cutaneous Leishmaniasis by Accelerating Lesion Healing and Reducing the Parasitic Load. ACS APPLIED BIO MATERIALS 2019; 2:2573-2586. [DOI: 10.1021/acsabm.9b00263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Sophia Malli
- Institut Galien Paris Sud, UMR CNRS 8612, Univ. Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5, rue J.B. Clément, 92296 Cedex Châtenay-Malabry, France
- BioCIS Biomolécules: Conception, Isolement, Synthèse, Chimiothérapie Antiparasitaire, UMR CNRS 8076, Univ. Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5, rue J.B. Clément, 92296 Cedex Châtenay-Malabry, France
| | - Sebastien Pomel
- BioCIS Biomolécules: Conception, Isolement, Synthèse, Chimiothérapie Antiparasitaire, UMR CNRS 8076, Univ. Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5, rue J.B. Clément, 92296 Cedex Châtenay-Malabry, France
| | - Yasmine Ayadi
- Institut Galien Paris Sud, UMR CNRS 8612, Univ. Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5, rue J.B. Clément, 92296 Cedex Châtenay-Malabry, France
- BioCIS Biomolécules: Conception, Isolement, Synthèse, Chimiothérapie Antiparasitaire, UMR CNRS 8076, Univ. Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5, rue J.B. Clément, 92296 Cedex Châtenay-Malabry, France
| | - Claudine Deloménie
- Faculté de Pharmacie, Institut Paris Saclay d’Innovation Thérapeutique, UMS Inserm CNRS UPSud, Université Paris-Saclay, 92296 Cedex Châtenay-Malabry, France
| | - Antonio Da Costa
- Université d’Artois, CNRS, Centrale Lille, ENSCL, Université Lille, UMR 8181, Unité de Catalyse et de Chimie du Solide (UCCS), Faculté Jean-Perrin, Rue Jean Souvras − SP 18, 62307 Lens, France
| | - Philippe M. Loiseau
- BioCIS Biomolécules: Conception, Isolement, Synthèse, Chimiothérapie Antiparasitaire, UMR CNRS 8076, Univ. Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5, rue J.B. Clément, 92296 Cedex Châtenay-Malabry, France
| | - Kawthar Bouchemal
- Institut Galien Paris Sud, Junior Member of the Institut Universitaire de France, UMR CNRS 8612, Univ. Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5, rue J.B. Clément, 92296 Cedex Châtenay-Malabry, France
| |
Collapse
|
13
|
Anti-leishmanial effect of spiro dihydroquinoline-oxindoles on volume regulation decrease and sterol biosynthesis of Leishmania braziliensis. Exp Parasitol 2019; 198:31-38. [PMID: 30690024 DOI: 10.1016/j.exppara.2019.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/03/2018] [Accepted: 01/20/2019] [Indexed: 12/23/2022]
Abstract
Diverse spiro dihydroquinoline-oxindoles (JS series) were prepared using the BF3•OEt2-catalyzed imino Diels-Alder reaction between ketimine-isatin derivatives and trans-isoeugenol. Ten spiro-oxiindole derivatives were selected and evaluated at different stages of the life cycle of Leishmania braziliensis parasites, responsible for cutaneous leishmaniasis in South America. Among them, the 8'-ethyl-4'-(4-hydroxy-3-methoxyphenyl)-3'-methyl-3',4'-dihydro-1'H-spiro[indoline-3,2'-quinolin]-2-one called JS87 was able to inhibit the growth of promastigotes without affecting the mammalian cells viability, and to decrease the number of intracellular amastigotes of L. braziliensis. This spiro compound was found to act through the alteration of parasite internal regulation by disrupting the regulatory volume decrease (RVD), and to affect the sterol biosynthetic pathway at level of squalene epoxidase (SE) enzyme. These results revealed that the spiro annulation between quinoline and oxindole scaffolds enhances the anti-leishmanial activity, and could assist in the development of potent quinoline-oxindole hybrids against Leishmania braziliensis, the main etiological agent of cutaneous leishmaniasis in South America.
Collapse
|
14
|
Dorlo TPC, Kip AE, Younis BM, Ellis SJ, Alves F, Beijnen JH, Njenga S, Kirigi G, Hailu A, Olobo J, Musa AM, Balasegaram M, Wasunna M, Karlsson MO, Khalil EAG. Visceral leishmaniasis relapse hazard is linked to reduced miltefosine exposure in patients from Eastern Africa: a population pharmacokinetic/pharmacodynamic study. J Antimicrob Chemother 2018; 72:3131-3140. [PMID: 28961737 PMCID: PMC5890687 DOI: 10.1093/jac/dkx283] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/11/2017] [Indexed: 01/02/2023] Open
Abstract
Background Low efficacy of miltefosine in the treatment of visceral leishmaniasis was recently observed in Eastern Africa. Objectives To describe the pharmacokinetics and establish a pharmacokinetic/pharmacodynamic relationship for miltefosine in Eastern African patients with visceral leishmaniasis, using a time-to-event approach to model relapse of disease. Methods Miltefosine plasma concentrations from 95 patients (48 monotherapy versus 47 combination therapy) were included in the population pharmacokinetic model using non-linear mixed effects modelling. Subsequently a time-to-event model was developed to model the time of clinical relapse. Various summary pharmacokinetic parameters (various AUCs, Time > EC50, Time > EC90), normalized within each treatment arm to allow simultaneous analysis, were evaluated as relapse hazard-changing covariates. Results A two-compartment population model with first-order absorption fitted the miltefosine pharmacokinetic data adequately. Relative bioavailability was reduced (−74%, relative standard error 4.7%) during the first week of treatment of the monotherapy arm but only the first day of the shorter combination regimen. Time to the relapse of infection could be described using a constant baseline hazard (baseline 1.8 relapses/year, relative standard error 72.7%). Miltefosine Time > EC90 improved the model significantly when added in a maximum effect function on the baseline hazard (half maximal effect with Time > EC90 6.97 days for monotherapy). Conclusions Miltefosine drug exposure was found to be decreased in Eastern African patients with visceral leishmaniasis, due to a (transient) initial lower bioavailability. Relapse hazard was inversely linked to miltefosine exposure. Significantly lower miltefosine exposure was observed in children compared with adults, further urging the need for implementation of dose adaptations for children.
Collapse
Affiliation(s)
- Thomas P C Dorlo
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.,Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital/Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anke E Kip
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital/Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Brima M Younis
- Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | - Sally J Ellis
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Fabiana Alves
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital/Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Simon Njenga
- Kenya Medical Research Institute, Nairobi, Kenya
| | | | - Asrat Hailu
- Addis Ababa University, Addis Ababa, Ethiopia
| | | | - Ahmed M Musa
- Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | | | | | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
15
|
Armitage EG, Alqaisi AQI, Godzien J, Peña I, Mbekeani AJ, Alonso-Herranz V, López-Gonzálvez Á, Martín J, Gabarro R, Denny PW, Barrett MP, Barbas C. Complex Interplay between Sphingolipid and Sterol Metabolism Revealed by Perturbations to the Leishmania Metabolome Caused by Miltefosine. Antimicrob Agents Chemother 2018; 62:e02095-17. [PMID: 29463533 PMCID: PMC5923112 DOI: 10.1128/aac.02095-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/21/2018] [Indexed: 12/24/2022] Open
Abstract
With the World Health Organization reporting over 30,000 deaths and 200,000 to 400,000 new cases annually, visceral leishmaniasis is a serious disease affecting some of the world's poorest people. As drug resistance continues to rise, there is a huge unmet need to improve treatment. Miltefosine remains one of the main treatments for leishmaniasis, yet its mode of action (MoA) is still unknown. Understanding the MoA of this drug and parasite response to treatment could help pave the way for new and more successful treatments for leishmaniasis. A novel method has been devised to study the metabolome and lipidome of Leishmania donovani axenic amastigotes treated with miltefosine. Miltefosine caused a dramatic decrease in many membrane phospholipids (PLs), in addition to amino acid pools, while sphingolipids (SLs) and sterols increased. Leishmania major promastigotes devoid of SL biosynthesis through loss of the serine palmitoyl transferase gene (ΔLCB2) were 3-fold less sensitive to miltefosine than wild-type (WT) parasites. Changes in the metabolome and lipidome of miltefosine-treated L. major mirrored those of L. donovani A lack of SLs in the ΔLCB2 mutant was matched by substantial alterations in sterol content. Together, these data indicate that SLs and ergosterol are important for miltefosine sensitivity and, perhaps, MoA.
Collapse
Affiliation(s)
- Emily G Armitage
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Montepríncipe, Boadilla del Monte, Madrid, Spain
- GSK I+D Diseases of the Developing World (DDW), Parque Tecnológico de Madrid, Tres Cantos, Madrid, Spain
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences & Glasgow Polyomics, University of Glasgow, Glasgow, United Kingdom
| | - Amjed Q I Alqaisi
- Department of Biosciences, Durham University, Lower Mountjoy, Durham, United Kingdom
- University of Baghdad, College of Science, Biology Department, Baghdad, Iraq
| | - Joanna Godzien
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Imanol Peña
- GSK I+D Diseases of the Developing World (DDW), Parque Tecnológico de Madrid, Tres Cantos, Madrid, Spain
| | - Alison J Mbekeani
- Department of Biosciences, Durham University, Lower Mountjoy, Durham, United Kingdom
| | - Vanesa Alonso-Herranz
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Ángeles López-Gonzálvez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Julio Martín
- GSK I+D Diseases of the Developing World (DDW), Parque Tecnológico de Madrid, Tres Cantos, Madrid, Spain
| | - Raquel Gabarro
- GSK I+D Diseases of the Developing World (DDW), Parque Tecnológico de Madrid, Tres Cantos, Madrid, Spain
| | - Paul W Denny
- Department of Biosciences, Durham University, Lower Mountjoy, Durham, United Kingdom
| | - Michael P Barrett
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences & Glasgow Polyomics, University of Glasgow, Glasgow, United Kingdom
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Montepríncipe, Boadilla del Monte, Madrid, Spain
| |
Collapse
|
16
|
Zulueta Díaz YDLM, Fanani ML. Crossregulation between the insertion of Hexadecylphosphocholine (miltefosine) into lipid membranes and their rheology and lateral structure. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017. [DOI: 10.1016/j.bbamem.2017.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
17
|
Villa-Pulgarín JA, Gajate C, Botet J, Jimenez A, Justies N, Varela-M RE, Cuesta-Marbán Á, Müller I, Modolell M, Revuelta JL, Mollinedo F. Mitochondria and lipid raft-located FOF1-ATP synthase as major therapeutic targets in the antileishmanial and anticancer activities of ether lipid edelfosine. PLoS Negl Trop Dis 2017; 11:e0005805. [PMID: 28829771 PMCID: PMC5568728 DOI: 10.1371/journal.pntd.0005805] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/13/2017] [Indexed: 11/18/2022] Open
Abstract
Background Leishmaniasis is the world’s second deadliest parasitic disease after malaria, and current treatment of the different forms of this disease is far from satisfactory. Alkylphospholipid analogs (APLs) are a family of anticancer drugs that show antileishmanial activity, including the first oral drug (miltefosine) for leishmaniasis and drugs in preclinical/clinical oncology trials, but their precise mechanism of action remains to be elucidated. Methodology/Principal findings Here we show that the tumor cell apoptosis-inducer edelfosine was the most effective APL, as compared to miltefosine, perifosine and erucylphosphocholine, in killing Leishmania spp. promastigotes and amastigotes as well as tumor cells, as assessed by DNA breakdown determined by flow cytometry. In studies using animal models, we found that orally-administered edelfosine showed a potent in vivo antileishmanial activity and diminished macrophage pro-inflammatory responses. Edelfosine was also able to kill Leishmania axenic amastigotes. Edelfosine was taken up by host macrophages and killed intracellular Leishmania amastigotes in infected macrophages. Edelfosine accumulated in tumor cell mitochondria and Leishmania kinetoplast-mitochondrion, and led to mitochondrial transmembrane potential disruption, and to the successive breakdown of parasite mitochondrial and nuclear DNA. Ectopic expression of Bcl-XL inhibited edelfosine-induced cell death in both Leishmania parasites and tumor cells. We found that the cytotoxic activity of edelfosine against Leishmania parasites and tumor cells was associated with a dramatic recruitment of FOF1-ATP synthase into lipid rafts following edelfosine treatment in both parasites and cancer cells. Raft disruption and specific FOF1-ATP synthase inhibition hindered edelfosine-induced cell death in both Leishmania parasites and tumor cells. Genetic deletion of FOF1-ATP synthase led to edelfosine drug resistance in Saccharomyces cerevisiae yeast. Conclusions/Significance The present study shows that the antileishmanial and anticancer actions of edelfosine share some common signaling processes, with mitochondria and raft-located FOF1-ATP synthase being critical in the killing process, thus identifying novel druggable targets for the treatment of leishmaniasis. Leishmaniasis is a major health problem worldwide, and can result in loss of human life or a lifelong stigma because of bodily scars. According to World Health Organization, leishmaniasis is considered as an emerging and uncontrolled disease, and its current treatment is far from ideal, with only a few drugs available that could lead to drug resistance or cause serious side-effects. Here, we have found that mitochondria and raft-located FOF1-ATPase synthase are efficient druggable targets, through which an ether lipid named edelfosine exerts its antileishmanial action. Edelfosine effectively kills Leishmania spp. promastigotes and amastigotes. Our experimental animal models demonstrate that oral administration of edelfosine exerts a potent antileishmanial activity, while inhibits macrophage pro-inflammatory responses. Our results show that both Leishmania and tumor cells share mitochondria and raft-located FOF1-ATPase synthase as major druggable targets in leishmaniasis and cancer therapy. These data, showing a potent antileishmanial activity of edelfosine and unveiling its mechanism of action, together with the inhibition of the inflammatory responses elicited by macrophages, suggest that the ether lipid edelfosine is a promising oral drug for leishmaniasis, and highlight mitochondria and lipid raft-located FOF1-ATP synthase as major therapeutic targets for the treatment of this disease.
Collapse
Affiliation(s)
- Janny A Villa-Pulgarín
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Consuelo Gajate
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Laboratory of Cell Death and Cancer Therapy, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Javier Botet
- Metabolic Engineering Group, Departamento de Microbiología y Genética, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - Alberto Jimenez
- Metabolic Engineering Group, Departamento de Microbiología y Genética, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - Nicole Justies
- Department of Cellular Immunology, Max-Planck-Institut für Immunbiologie und Epigenetik, Freiburg, Germany
| | - Rubén E Varela-M
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Álvaro Cuesta-Marbán
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Ingrid Müller
- Department of Medicine, Section of Immunology, St. Mary's Campus, Imperial College London, London, United Kingdom
| | - Manuel Modolell
- Department of Cellular Immunology, Max-Planck-Institut für Immunbiologie und Epigenetik, Freiburg, Germany
| | - José L Revuelta
- Metabolic Engineering Group, Departamento de Microbiología y Genética, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - Faustino Mollinedo
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Laboratory of Cell Death and Cancer Therapy, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
18
|
Ríos-Marco P, Marco C, Gálvez X, Jiménez-López JM, Carrasco MP. Alkylphospholipids: An update on molecular mechanisms and clinical relevance. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1657-1667. [PMID: 28238819 DOI: 10.1016/j.bbamem.2017.02.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 11/16/2022]
Abstract
Alkylphospholipids (APLs) represent a new class of drugs which do not interact directly with DNA but act on the cell membrane where they accumulate and interfere with lipid metabolism and signalling pathways. This review summarizes the mode of action at the molecular level of these compounds. In this sense, a diversity of mechanisms has been suggested to explain the actions of clinically-relevant APLs, in particular, in cancer treatment. One consistently reported finding is that APLs reduce the biosynthesis of phosphatidylcholine (PC) by inhibiting the rate-limiting enzyme CTP:phosphocholine cytidylyltransferase (CT). APLs also alter intracellular cholesterol traffic and metabolism in human tumour-cell lines, leading to an accumulation of cholesterol inside the cell. An increase in cholesterol biosynthesis associated with a decrease in the synthesis of choline-containing phospholipids and cholesterol esterification leads to a change in the free-cholesterol:PC ratio in cells exposed to APLs. Akt phosphorylation status after APL exposure shows that this critical regulator for cell survival is modulated by changes in cholesterol levels induced in the plasma membrane by these lipid analogues. Furthermore, APLs produce cell ultrastructural alterations with an abundant autophagic vesicles and autolysosomes in treated cells, indicating an interference of autophagy process after APL exposure. Thus, antitumoural APLs interfere with the proliferation of tumour cells via a complex mechanism involving phospholipid and cholesterol metabolism, interfere with lipid-dependent survival-signalling pathways and autophagy. Although APLs also exert antiparasitic, antibacterial, and antifungal effects, in this review we provide a summary of the antileishmanial activity of these lipid analogues. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
- Pablo Ríos-Marco
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain
| | - Carmen Marco
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain
| | - Xiomara Gálvez
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain
| | - José M Jiménez-López
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain.
| | - María P Carrasco
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain.
| |
Collapse
|
19
|
Romero AH, Medina R, Alcala A, García-Marchan Y, Núñez-Duran J, Leañez J, Mijoba A, Ciangherotti C, Serrano-Martín X, López SE. Design, synthesis, structure-activity relationship and mechanism of action studies of a series of 4-chloro-1-phthalazinyl hydrazones as a potent agent against Leishmania braziliensis. Eur J Med Chem 2017; 127:606-620. [DOI: 10.1016/j.ejmech.2017.01.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 11/29/2022]
|
20
|
Experimental resistance to drug combinations in Leishmania donovani: metabolic and phenotypic adaptations. Antimicrob Agents Chemother 2015; 59:2242-55. [PMID: 25645828 DOI: 10.1128/aac.04231-14] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Together with vector control, chemotherapy is an essential tool for the control of visceral leishmaniasis (VL), but its efficacy is jeopardized by growing resistance and treatment failure against first-line drugs. To delay the emergence of resistance, the use of drug combinations of existing antileishmanial agents has been tested systematically in clinical trials for the treatment of visceral leishmaniasis (VL). In vitro, Leishmania donovani promastigotes are able to develop experimental resistance to several combinations of different antileishmanial drugs after 10 weeks of drug pressure. Using an untargeted liquid chromatography-mass spectrometry (LC-MS) metabolomics approach, we identified metabolic changes in lines that were experimentally resistant to drug combinations and their respective single-resistant lines. This highlighted both collective metabolic changes (found in all combination therapy-resistant [CTR] lines) and specific ones (found in certain CTR lines). We demonstrated that single-resistant and CTR parasite cell lines show distinct metabolic adaptations, which all converge on the same defensive mechanisms that were experimentally validated: protection against drug-induced and external oxidative stress and changes in membrane fluidity. The membrane fluidity changes were accompanied by changes in drug uptake only in the lines that were resistant against drug combinations with antimonials, and surprisingly, drug accumulation was higher in these lines. Together, these results highlight the importance and the central role of protection against oxidative stress in the different resistant lines. Ultimately, these phenotypic changes might interfere with the mode of action of all drugs that are currently used for the treatment of VL and should be taken into account in drug development.
Collapse
|
21
|
Dinesh N, Kaur PK, Swamy KK, Singh S. Mianserin, an antidepressant kills Leishmania donovani by depleting ergosterol levels. Exp Parasitol 2014; 144:84-90. [PMID: 24950381 DOI: 10.1016/j.exppara.2014.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 05/30/2014] [Accepted: 06/02/2014] [Indexed: 01/25/2023]
Abstract
In the present study, we have investigated the antileishmanial potential of mianserin, an antidepressant. Mianserin was found to inhibit both the promastigote and amastigote forms of the parasite in a dose dependant manner. The IC50 values for promastigotes and amastigotes were 21 μM and 46 μM respectively. Interestingly, mianserin failed to inhibit THP-1 differentiated macrophages up to 100 μM concentration thus, exhibiting parasite selectivity. When mianserin was incubated with recombinant Leishmania donovani 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR) enzyme, it exhibited an IC50 value of 19.8 μM. Inhibition kinetics revealed competitive mode of enzyme inhibition as the Km increased with no change in Vmax. Further structural investigation of enzyme-inhibitor interaction revealed quenching of HMGR tryptophan intrinsic fluorescence with a K(sv) value of 3.025±0.37 M(-1) and an apparent binding constant of 0.0954 mM. We further estimated ergosterol levels which is a major component of Leishmania cell membrane. It is synthesized by HMGR enzyme, the first rate limiting enzyme of the sterol biosynthetic pathway. Analysis of ergosterol levels by HPLC revealed ∼2.5-fold depletion in mianserin treated promastigotes with respect to untreated parasites. This data was further validated by exogenous supplementation of mianserin treated cells with ergosterol and cholesterol. Reversal of growth inhibition was observed only upon ergosterol addition though it was refractory to cholesterol supplementation. Overall, our results demonstrate the possibility of repositioning of an antidepressant for the treatment of Visceral Leishmaniasis.
Collapse
Affiliation(s)
- Neeradi Dinesh
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali 160062, Punjab, India
| | - Preet Kamal Kaur
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali 160062, Punjab, India
| | - Kayala Kambagiri Swamy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali 160062, Punjab, India
| | - Sushma Singh
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
22
|
de Morais-Teixeira E, Gallupo MK, Rodrigues LF, Romanha AJ, Rabello A. In vitro interaction between paromomycin sulphate and four drugs with leishmanicidal activity against three New World Leishmania species. J Antimicrob Chemother 2013; 69:150-4. [DOI: 10.1093/jac/dkt318] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
23
|
Bompart D, Núñez-Durán J, Rodríguez D, Kouznetsov VV, Meléndez Gómez CM, Sojo F, Arvelo F, Visbal G, Alvarez A, Serrano-Martín X, García-Marchán Y. Anti-leishmanial evaluation of C2-aryl quinolines: mechanistic insight on bioenergetics and sterol biosynthetic pathway of Leishmania braziliensis. Bioorg Med Chem 2013; 21:4426-31. [PMID: 23719286 DOI: 10.1016/j.bmc.2013.04.063] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 04/11/2013] [Accepted: 04/18/2013] [Indexed: 10/26/2022]
Abstract
A series of diverse simple C2-aryl quinolines was synthesized de novo via a straightforward synthesis based on the acid-catalyzed multicomponent imino Diels-Alder reactions. Seven selected quinolines were evaluated at different stages of Leishmania braziliensis parasite. Among them, the 6-ethyl-2-phenylquinoline 5f was able to inhibit the growth of promastigotes of this parasite without affecting the mammalian cells viability and decreasing the number of intracellular L. braziliensis amastigotes on BMDM macrophages. The mechanism of action studied for the selected compound consisted in: (1) alteration of parasite bioenergetics, by disrupting mitochondrial electrochemical potential and alkalinization of acidocalcisomes, and (2) inhibition of ergosterol biosynthetic pathway in promastigote forms. These results validate the efficiency of quinoline molecules as leishmanicide compounds.
Collapse
Affiliation(s)
- Daznia Bompart
- Laboratorio de Señalización Celular y Bioquímica de Parásitos, Área de Salud, Instituto de Estudios Avanzados (IDEA), Caracas, Bolivarian Republic of Venezuela
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Attenuation of Leishmania infantum chagasi metacyclic promastigotes by sterol depletion. Infect Immun 2013; 81:2507-17. [PMID: 23630964 DOI: 10.1128/iai.00214-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The infectious metacyclic promastigotes of Leishmania protozoa establish infection in a mammalian host after they are deposited into the dermis by a sand fly vector. Several Leishmania virulence factors promote infection, including the glycosylphosphatidylinositol membrane-anchored major surface protease (MSP). Metacyclic Leishmania infantum chagasi promastigotes were treated with methyl-beta-cyclodextrin (MβCD), a sterol-chelating reagent, causing a 3-fold reduction in total cellular sterols as well as enhancing MSP release without affecting parasite viability in vitro. MβCD-treated promastigotes were more susceptible to complement-mediated lysis than untreated controls and reduced the parasite load 3-fold when inoculated into BALB/c mice. Paradoxically, MβCD-treated promastigotes caused a higher initial in vitro infection rate in human or murine macrophages than untreated controls, although their intracellular multiplication was hindered upon infection establishment. There was a corresponding larger amount of covalently bound C3b than iC3b on the parasite surfaces of MβCD-treated promastigotes exposed to healthy human serum in vitro, as well as loss of MSP, a protease that enhances C3b cleavage to iC3b. Mass spectrometry showed that MβCD promotes the release of proteins into the extracellular medium, including both MSP and MSP-like protein (MLP), from virulent metacyclic promastigotes. These data support the hypothesis that plasma membrane sterols are important for the virulence of Leishmania protozoa at least in part through retention of membrane virulence proteins.
Collapse
|
25
|
Goldston AM, Powell RR, Temesvari LA. Sink or swim: lipid rafts in parasite pathogenesis. Trends Parasitol 2012; 28:417-26. [PMID: 22906512 DOI: 10.1016/j.pt.2012.07.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 12/26/2022]
Abstract
Lipid rafts, sterol- and sphingolipid-rich membrane microdomains, have been extensively studied in mammalian cells. Recently, lipid rafts have been shown to control virulence in a variety of parasites including Entamoeba histolytica, Giardia intestinalis, Leishmania spp., Plasmodium spp., Toxoplasma gondii, and Trypanosoma spp. Parasite rafts regulate adhesion to host and invasion, and parasite adhesion molecules often localize to rafts. Parasite rafts also control vesicle trafficking, motility, and cell signaling. Parasites disrupt host cell rafts; the dysregulation of host membrane function facilitates the establishment of infection and evasion of the host immune system. Discerning the mechanism by which lipid rafts regulate parasite pathogenesis is essential to our understanding of virulence. Such insight may guide the development of new drugs for disease management.
Collapse
Affiliation(s)
- Amanda M Goldston
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
| | | | | |
Collapse
|
26
|
De Cicco NNT, Pereira MG, Corrêa JR, Andrade-Neto VV, Saraiva FB, Chagas-Lima AC, Gondim KC, Torres-Santos EC, Folly E, Saraiva EM, Cunha-E-Silva NL, Soares MJ, Atella GC. LDL uptake by Leishmania amazonensis: involvement of membrane lipid microdomains. Exp Parasitol 2012; 130:330-40. [PMID: 22381219 DOI: 10.1016/j.exppara.2012.02.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 02/10/2012] [Accepted: 02/13/2012] [Indexed: 02/02/2023]
Abstract
Leishmania amazonensis lacks a de novo mechanism for cholesterol synthesis and therefore must scavenge this lipid from the host environment. In this study we show that the L. amazonensis takes up and metabolizes human LDL(1) particles in both a time and dose-dependent manner. This mechanism implies the presence of a true LDL receptor because the uptake is blocked by both low temperature and by the excess of non-labelled LDL. This receptor is probably associated with specific microdomains in the membrane of the parasite, such as rafts, because this process is blocked by methyl-β-cyclodextrin (MCBD). Cholesteryl ester fluorescently-labeled LDL (BODIPY-cholesteryl-LDL) was used to follow the intracellular distribution of this lipid. After uptake it was localized in large compartments along the parasite body. The accumulation of LDL was analyzed by flow cytometry using FITC-labeled LDL particles. Together these data show for the first time that L. amazonensis is able to compensate for its lack of lipid synthesis through the use of a lipid importing machinery largely based on the uptake of LDL particles from the host. Understanding the details of the molecular events involved in this mechanism may lead to the identification of novel targets to block Leishmania infection in human hosts.
Collapse
Affiliation(s)
- Nuccia N T De Cicco
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21.941-902, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Silva AM, Cordeiro-da-Silva A, Coombs GH. Metabolic variation during development in culture of Leishmania donovani promastigotes. PLoS Negl Trop Dis 2011; 5:e1451. [PMID: 22206037 PMCID: PMC3243725 DOI: 10.1371/journal.pntd.0001451] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 11/10/2011] [Indexed: 11/28/2022] Open
Abstract
The genome sequencing of several Leishmania species has provided immense amounts of data and allowed the prediction of the metabolic pathways potentially operating. Subsequent genetic and proteomic studies have identified stage-specific proteins and putative virulence factors but many aspects of the metabolic adaptations of Leishmania remain to be elucidated. In this study, we have used an untargeted metabolomics approach to analyze changes in the metabolite profile as promastigotes of L. donovani develop during in vitro cultures from logarithmic to stationary phase. The results show that the metabolomes of promastigotes on days 3–6 of culture differ significantly from each other, consistent with there being distinct developmental changes. Most notable were the structural changes in glycerophospholipids and increase in the abundance of sphingolipids and glycerolipids as cells progress from logarithmic to stationary phase. Leishmania infections are considered neglected tropical diseases as the parasites affect millions of people worldwide but there are limited research efforts aimed at obtaining vaccines and new drugs. Leishmania has a digenetic life cycle alternating between promastigote forms, which develop in the sand-fly, the vector of the disease, and an amastigote form, which grows in mammals after being bitten by an infected sand-fly. In vitro studies with the promastigote forms are routinely used to gain insights about the parasite's cell biology. Little is known about how the different promastigotes forms are metabolically adapted to their particular micro-environment in the host or how they are pre-adapted metabolically for infecting a mammal, thus we have undertaken a study of the metabolite profile of L. donovani promastigotes in order to gain an understanding of the changes that occur during promastigote development. The analysis has revealed that the changes in promastigotes' metabolome between days 3 and 6 take place in a progressive manner; however major differences were observed when comparing the promastigotes on days 3 and 6. An increase in lipid abundance as promastigote development occurred was notable and is likely to reflect remodelling of the parasite's surface in readiness for infecting a mammal.
Collapse
Affiliation(s)
- Ana Marta Silva
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Laboratório de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto, Porto, Portugal
| | - Anabela Cordeiro-da-Silva
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Laboratório de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto, Porto, Portugal
| | - Graham H. Coombs
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
- * E-mail:
| |
Collapse
|
28
|
Membrane lipidomics for the discovery of new antiparasitic drug targets. Trends Parasitol 2011; 27:496-504. [DOI: 10.1016/j.pt.2011.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 07/21/2011] [Accepted: 07/26/2011] [Indexed: 01/04/2023]
|
29
|
Manzano JI, Carvalho L, García-Hernández R, Poveda JA, Ferragut JA, Castanys S, Gamarro F. Uptake of the antileishmania drug tafenoquine follows a sterol-dependent diffusion process in Leishmania. J Antimicrob Chemother 2011; 66:2562-5. [PMID: 21846675 DOI: 10.1093/jac/dkr345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The present study was designed to elucidate the mechanism of tafenoquine uptake in Leishmania and its sterol dependence. METHODS Because tafenoquine is a fluorescent compound, spectrofluorimetric analysis allowed us to monitor its uptake by Leishmania promastigotes and intracellular amastigotes, and to evaluate the effect of temperature, energy and H+ gradient on drug entry. The influence of sterols on tafenoquine uptake in Leishmania parasites was determined in experiments using sterol-depleting agents such as methyl-β-cyclodextrin or cholesterol oxidase. RESULTS Tafenoquine exhibited fast entry kinetics into Leishmania in an energy-independent, but pH- and temperature-dependent, non-saturable process. Furthermore, sterol depletion decreased tafenoquine uptake. CONCLUSIONS These findings suggest that Leishmania takes up tafenoquine by a diffusion process and that decreases in membrane sterol content may induce a decrease in drug uptake.
Collapse
Affiliation(s)
- José Ignacio Manzano
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC (IPBLN-CSIC), Parque Tecnológico de Ciencias de la Salud, Avda. del Conocimiento s/n, 18100 Armilla, Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
30
|
Van Assche T, Deschacht M, da Luz RAI, Maes L, Cos P. Leishmania-macrophage interactions: insights into the redox biology. Free Radic Biol Med 2011; 51:337-51. [PMID: 21620959 DOI: 10.1016/j.freeradbiomed.2011.05.011] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 04/24/2011] [Accepted: 05/07/2011] [Indexed: 11/19/2022]
Abstract
Leishmaniasis is a neglected tropical disease that affects about 350 million individuals worldwide. The protozoan parasite has a relatively simple life cycle with two principal stages: the flagellated mobile promastigote living in the gut of the sandfly vector and the intracellular amastigote within phagolysosomal vesicles of the vertebrate host macrophage. This review presents a state-of-the-art overview of the redox biology at the parasite-macrophage interface. Although Leishmania species are susceptible in vitro to exogenous superoxide radical, hydrogen peroxide, nitric oxide, and peroxynitrite, they manage to survive the endogenous oxidative burst during phagocytosis and the subsequent elevated nitric oxide production in the macrophage. The parasite adopts various defense mechanisms to cope with oxidative stress: the lipophosphoglycan membrane decreases superoxide radical production by inhibiting NADPH oxidase assembly and the parasite also protects itself by expressing antioxidant enzymes and proteins. Some of these enzymes could be considered potential drug targets because they are not expressed in mammals. In respect to antileishmanial therapy, the effects of current drugs on parasite-macrophage redox biology and its involvement in the development of drug resistance and treatment failure are presented.
Collapse
Affiliation(s)
- Tim Van Assche
- Laboratory of Microbiology Parasitology, and Hygiene, University of Antwerp, B-2020 Antwerp, Belgium
| | | | | | | | | |
Collapse
|
31
|
Sundar S, Sinha PK, Rai M, Verma DK, Nawin K, Alam S, Chakravarty J, Vaillant M, Verma N, Pandey K, Kumari P, Lal CS, Arora R, Sharma B, Ellis S, Strub-Wourgaft N, Balasegaram M, Olliaro P, Das P, Modabber F. Comparison of short-course multidrug treatment with standard therapy for visceral leishmaniasis in India: an open-label, non-inferiority, randomised controlled trial. Lancet 2011; 377:477-86. [PMID: 21255828 DOI: 10.1016/s0140-6736(10)62050-8] [Citation(s) in RCA: 251] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Improved treatment approaches are needed for visceral leishmaniasis. We assessed the efficacy and safety of three potential short-course combination treatments compared with the standard monotherapy in India. METHODS Standard treatment (1 mg/kg amphotericin B infusion on alternate days for 30 days, total dose 15 mg/kg) was compared with three drug combinations (single injection of 5 mg/kg liposomal amphotericin B and 7-day 50 mg oral miltefosine or single 10-day 11 mg/kg intramuscular paromomycin; or 10 days each of miltefosine and paromomycin) in an open-label, parallel-group, non-inferiority, randomised controlled trial in two hospital sites in Bihar, India. Patients aged 5-60 years with parasitologically confirmed visceral leishmaniasis were randomly assigned one of the four treatments by the trial statistician by use of a computer-generated list. Clinical assessments were done at the end of treatment (15 days on combination treatment; 31 days for standard treatment) and after 45 days and 6 months. The primary endpoint was definitive cure (defined as no sign or symptom of visceral leishmaniasis and parasitologically cured to the last follow-up). Analyses were done both by intention to treat and per protocol. This trial is registered with ClinicalTrials.gov, number NCT00696969. FINDINGS Between June, 2008, and July, 2009, 634 patients were assigned amphotericin B (n=157), liposomal amphotericin B with miltefosine (n=160) or paromomycin (n=158), or miltefosine and paromomycin (n=159). 618 patients were in the per-protocol population. There were two relapses in each group. The numbers with definitive cure at 6 months for the intention-to-treat population were 146 (cure rate 93·0%; CI 87·5-96·3) for amphotericin B, 156 (97·5%; 93·3-99·2) for liposomal amphotericin B and miltefosine, 154 (97·5%; 93·24-99·2) for liposomal amphotericin B and paromomycin, and 157 (98·7%; 95·1-99·8) for miltefosine and paromomycin. All combinations were non-inferior to the standard treatment, in both the intention-to-treat and per-protocol populations. Patients in the combination groups had fewer adverse events than did those assigned standard treatment. INTERPRETATION Combination treatments for visceral leishmaniasis are efficacious and safe, and decrease the duration of therapy, thereby encouraging adherence and reducing emergence of drug-resistant parasites. FUNDING Drugs for Neglected Diseases initiative and the Indian Council of Medical Research.
Collapse
Affiliation(s)
- Shyam Sundar
- Kala-Azar Medical Research Center, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
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: 5.0] [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
|
33
|
Coimbra ES, Libong D, Cojean S, Saint-Pierre-Chazalet M, Solgadi A, Le Moyec L, Duenas-Romero AM, Chaminade P, Loiseau PM. Mechanism of interaction of sitamaquine with Leishmania donovani. J Antimicrob Chemother 2010; 65:2548-55. [PMID: 20956354 DOI: 10.1093/jac/dkq371] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES This study focuses on the mechanism of interaction of sitamaquine with Leishmania donovani membranes, and its accumulation within the parasites. METHODS A biomimetic model of the outer layer of a Leishmania plasma membrane was used to examine the interactions of sitamaquine with lipids. The plasma membranes of L. donovani promastigotes were depleted of sterol using cholesterol oxidase, in order to assess the importance of sterols in drug-membrane interactions. Sterols were quantified and sitamaquine susceptibility was assessed using the MTT test. Kinetics of sitamaquine accumulation and efflux were measured under different conditions. RESULTS Sitamaquine interacts first with phospholipid anionic polar head groups and then with phospholipid acyl chains to insert within biological membranes and accumulates rapidly in the Leishmania cytosol according to a sterol-independent process. The rapid sitamaquine efflux observed was related to an energy-dependent mechanism since the intracellular amount of sitamaquine was enhanced three times in the absence of glucose and the efflux was inhibited in energy-depleted conditions. (1)H NMR analysis of motile lipid showed that sitamaquine did not affect lipid trafficking in Leishmania. CONCLUSIONS We propose that sitamaquine rapidly accumulates in Leishmania by diffusion along an electrical gradient and is concentrated in the cytosol by an energy- and sterol-independent process. The affinity of sitamaquine for membranes was transitory and an energy-dependent efflux was demonstrated, suggesting the presence of an as yet uncharacterized transporter.
Collapse
Affiliation(s)
- E S Coimbra
- Université Paris-Sud, UMR 8076, Chimiothérapie Antiparasitaire, Faculté de Pharmacie, Chatenay-Malabry, France
| | | | | | | | | | | | | | | | | |
Collapse
|