1
|
Romero AH, Delgado F. 4-Aminoquinoline as a privileged scaffold for the design of leishmanicidal agents: structure-property relationships and key biological targets. Front Chem 2025; 12:1527946. [PMID: 39981131 PMCID: PMC11841433 DOI: 10.3389/fchem.2024.1527946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/26/2024] [Indexed: 02/22/2025] Open
Abstract
Leishmaniasis is one of the most important neglected tropical diseases, with more than two million new cases annually. It is endemic in several regions worldwide, representing a public health problem for more than 88 countries, in particular in the tropical and subtropical regions of developing countries. At the moment, there are neither approved vaccines nor effective drugs for the treatment of human leishmaniasis for any of its three typical clinical manifestations, and, importantly, the drugs of clinical use have several side effects, require complex administration regimens, present high cost, and are ineffective in many populations due to pathogen resistance. Moreover, beyond the pharmacological exigencies, there are other challenges concerning its parasitic nature, such as its great genetic plasticity and adaptability, enabling it to activate a battery of genes to develop resistance quickly. All these aspects demand the identification and development of new, safe, and effective chemical systems, which must not only be focused on medicinal chemistry and pharmacological aspects but also consider key aspects relative to parasite survival. In this sense, the quinolines and, in particular, 4-aminoquinoline, represent a privileged scaffold for the design of potential leishmanicidal candidates due not only to their versatility to generate highly active and selective compounds but also to their correlation with well-defined biological targets. These facts make it possible to generate safe leishmanicidal agents targeted at key aspects of parasite survival. The current review summarizes the most current examples of leishmanicidal agents based on 4-aminoquinolines focusing the analysis on two essential aspects: (i) structure-property relationship to identify the key pharmacophores and (ii) mode of action focused on key targets in parasite survival (e.g., depolarization of potential mitochondrial, accumulation into macrophage lysosome, and immunostimulation of host cells). With that information, we seek to give useful guidelines for interested researchers to face the drug discovery and development process for selective and potent leishmanicidal agents based on 4-aminoquinolines.
Collapse
Affiliation(s)
- Angel H. Romero
- Grupo de Química Orgánica Medicinal, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | | |
Collapse
|
2
|
Mollo MC, Cambiaso ML, Ferreira LLG, Kilimciler NB, Bisceglia JA, Andricopulo AD, Orelli LR. Synthesis and in vitro leishmanicidal activity of novel N-arylspermidine derivatives. Bioorg Chem 2025; 154:108083. [PMID: 39721144 DOI: 10.1016/j.bioorg.2024.108083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
This work describes the synthesis and biological evaluation of hitherto unknown N-arylspermidine derivatives 3. Compounds 3 were efficiently prepared from cyclic amidines through a novel synthetic approach comprising alkylation with ω-halonitriles followed by reduction. The cyclic N-arylamidine directs the alkylation to the unsubstituted nitrogen and also provides the N-benzyl group present in the triamine after simultaneous reduction of the resulting quaternary salt 2 and the cyano group. The N-aryl spermidines were tested in Leishmania infantum promastigotes and also in the more challenging form intracellular amastigotes. The compounds toxicity was also assessed in two cell lines, THP-1 and HepG2. In silico physicochemical and ADME predictions were also carried out. Eight out of ten compounds displayed EC50 around 5 µM against L. infantum intracellular amastigotes. Among them, derivatives 3c, 3d, and 3h showed potency in the low micromolar range with SI > 5 and suitable predicted physicochemical ADME properties. The antileishmanial activity of the compounds would rely on the N-arylspermidine moiety, as assessed by evaluation of related substructures which were inactive. This first series of compounds, among which two derivatives (3b,h) displayed EC50 values comparable to Miltefosine, represent a good starting point for further studies and multiparametric optimization to obtain more potent and selective candidates for the treatment of this neglected tropical disease.
Collapse
Affiliation(s)
- María C Mollo
- Universidad de Buenos Aires, CONICET, Cátedra de Química Orgánica II, Departamento de Ciencias Químicas, Facultad de Farmacia y Bioquímica, Junín 956, 1113 Buenos Aires, Argentina; Laboratory of Medicinal and Computational Chemistry, Physics Institute of São Carlos, University of São Paulo, Av. Joao Dagnone 1100, 13563-120 São Carlos, SP, Brazil
| | - Mariana L Cambiaso
- Universidad de Buenos Aires, CONICET, Cátedra de Química Orgánica II, Departamento de Ciencias Químicas, Facultad de Farmacia y Bioquímica, Junín 956, 1113 Buenos Aires, Argentina
| | - Leonardo L G Ferreira
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of São Carlos, University of São Paulo, Av. Joao Dagnone 1100, 13563-120 São Carlos, SP, Brazil
| | - Natalia B Kilimciler
- Universidad de Buenos Aires, CONICET, Cátedra de Química Orgánica II, Departamento de Ciencias Químicas, Facultad de Farmacia y Bioquímica, Junín 956, 1113 Buenos Aires, Argentina
| | - Juan A Bisceglia
- Universidad de Buenos Aires, CONICET, Cátedra de Química Orgánica II, Departamento de Ciencias Químicas, Facultad de Farmacia y Bioquímica, Junín 956, 1113 Buenos Aires, Argentina
| | - Adriano D Andricopulo
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of São Carlos, University of São Paulo, Av. Joao Dagnone 1100, 13563-120 São Carlos, SP, Brazil.
| | - Liliana R Orelli
- Universidad de Buenos Aires, CONICET, Cátedra de Química Orgánica II, Departamento de Ciencias Químicas, Facultad de Farmacia y Bioquímica, Junín 956, 1113 Buenos Aires, Argentina.
| |
Collapse
|
3
|
Corfu AI, Santarem N, Luelmo S, Mazza G, Greco A, Altomare A, Ferrario G, Nasta G, Keminer O, Aldini G, Tamborini L, Basilico N, Parapini S, Gul S, Cordeiro-da-Silva A, Conti P, Borsari C. Discovery of 1,3,4-Oxadiazole Derivatives as Broad-Spectrum Antiparasitic Agents. ACS Infect Dis 2024; 10:2222-2238. [PMID: 38717116 DOI: 10.1021/acsinfecdis.4c00181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Vector-borne parasitic diseases (VBPDs) pose a significant threat to public health on a global scale. Collectively, Human African Trypanosomiasis (HAT), Leishmaniasis, and Malaria threaten millions of people, particularly in developing countries. Climate change might alter the transmission and spread of VBPDs, leading to a global burden of these diseases. Thus, novel agents are urgently needed to expand therapeutic options and limit the spread of drug-resistant parasites. Herein, we report the development of broad-spectrum antiparasitic agents by screening a known library of antileishmanial and antimalarial compounds toward Trypanosoma brucei (T. brucei) and identifying a 1,3,4-oxadiazole derivative (19) as anti-T. brucei hit with predicted blood-brain barrier permeability. Subsequently, extensive structure-activity-relationship studies around the lipophilic tail of 19 led to a potent antitrypanosomal and antimalarial compound (27), with moderate potency also toward Leishmania infantum (L. infantum) and Leishmania tropica. In addition, we discovered a pan-active antiparasitic molecule (24), showing low-micromolar IC50s toward T. brucei and Leishmania spp. promastigotes and amastigotes, and nanomolar IC50 against Plasmodium falciparum, together with high selectivity for the parasites over mammalian cells (THP-1). Early ADME-toxicity assays were used to assess the safety profile of the compounds. Overall, we characterized 24 and 27, bearing the 1,3,4-oxadiazole privileged scaffold, as broad-spectrum low-toxicity agents for the treatment of VBPDs. An alkyne-substituted chemical probe (30) was synthesized and will be utilized in proteomics experiments aimed at deconvoluting the mechanism of action in the T. brucei parasite.
Collapse
Affiliation(s)
- Alexandra Ioana Corfu
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Nuno Santarem
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sara Luelmo
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Gaia Mazza
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Alessandro Greco
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Alessandra Altomare
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Giulio Ferrario
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Giulia Nasta
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Oliver Keminer
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Lucia Tamborini
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Nicoletta Basilico
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Silvia Parapini
- Department of Biomedical Sciences for Health, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Anabela Cordeiro-da-Silva
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Paola Conti
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Chiara Borsari
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| |
Collapse
|
4
|
Pandey AR, Singh SP, Ramalingam K, Yadav K, Bisen AC, Bhatta RS, Srivastava M, Tripathi R, Goyal N, Sashidhara KV. Antileishmanial evaluation of triazole-butenolide conjugates: design, synthesis, in vitro screening, SAR and in silico ADME predictions. RSC Med Chem 2023; 14:1131-1142. [PMID: 37360388 PMCID: PMC10285739 DOI: 10.1039/d2md00464j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/25/2023] [Indexed: 06/28/2023] Open
Abstract
In the quest to discover novel scaffolds with leishmanicidal effects, a series of 23 compounds containing the most promising 1,2,3-triazole and highly potent butenolide in one framework were synthesized. The synthesized conjugates were screened against Leishmania donovani parasite; five of them showed moderate antileishmanial activity against promastigotes (IC50 30.6 to 35.5 μM) and eight of them exhibited significant activity against amastigotes (IC50 ≤12 μM). Compound 10u was found to be the most active (IC50 8.4 ± 0.12 μM) with the highest safety index (20.47). The series was further evaluated against Plasmodium falciparum (3D7 strain) and seven compounds were found to be moderately active. Among them, again 10u emerged as the most active compound (IC50 3.65 μM). In antifilarial assays against adult female Brugia malayi, five compounds showed grade II inhibition (50-74%). Structure-activity relationship (SAR) analysis suggested a substituted phenyl ring, triazole and butenolide as essential structural features for bioactivity. Moreover, the results of in silico ADME parameter and pharmacokinetic studies indicated that the synthesized triazole-butenolide conjugates abide by the required criteria for the development of orally active drugs, and thus this scaffold can be used as a pharmacologically active framework that should be considered for the development of potential antileishmanial hits.
Collapse
Affiliation(s)
- Alka Raj Pandey
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 U.P India +91 522 2771942/2771970 +91 522 2772450, ext. 4684
- Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 U.P India
| | - Suriya Pratap Singh
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 U.P India +91 522 2771942/2771970 +91 522 2772450, ext. 4684
| | - Karthik Ramalingam
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 U.P India
| | - Kanchan Yadav
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Lucknow 226031 U.P India
| | - Amol Chhatrapati Bisen
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute Lucknow 226031 U.P India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 U.P India
| | - Rabi Sankar Bhatta
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute Lucknow 226031 U.P India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 U.P India
| | - Mrigank Srivastava
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Lucknow 226031 U.P India
| | - Renu Tripathi
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Lucknow 226031 U.P India
| | - Neena Goyal
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 U.P India
| | - Koneni V Sashidhara
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 U.P India +91 522 2771942/2771970 +91 522 2772450, ext. 4684
- Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 U.P India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 U.P India
| |
Collapse
|
5
|
Ribeiro Antinarelli LM, Glanzmann N, Mendonça DVC, Lage DP, Oliveira-da-Silva JA, Tavares GSV, Carvalho AMRS, Freitas CS, Martins VT, Duarte MC, Menezes-Souza D, da Silva AD, Coelho EAF, Soares Coimbra E. Parasitological and immunological evaluation of a quinoline derivative salt incorporated into a polymeric micelle formulation against Leishmania infantum infection. Parasitol Res 2022; 121:2129-2140. [PMID: 35614147 PMCID: PMC9132674 DOI: 10.1007/s00436-022-07544-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/06/2022] [Indexed: 11/28/2022]
Abstract
Leishmaniasis is a parasitic disease caused by Leishmania protozoa, which presents a large spectrum of clinical manifestations. In the present study, a quinoline derivative salt named N-(2-((7-chloroquinolin-4-yl)amino)ethyl)-N-(prop-2-yn-1-yl)prop-2-yn-1-aminium chloride or QDS3 was in vitro and in vivo tested against L. infantum by means of its incorporation in Poloxamer 407-based polymeric micelles (QDS3/M). The in vitro antileishmanial activity of QDS3 and QDS3/M was investigated in L. infantum promastigotes, axenic amastigotes and infected macrophages. BALB/c mice were infected with L. infantum, and parasitological parameters were evaluated 1 and 15 days post-treatment by determining the parasite load by a limiting dilution assay, besides a quantitative PCR (qPCR) method. Immunological response was assessed based on production of cellular cytokines, as well as by quantification of nitrite levels and specific antibodies. In vitro results showed that QDS3 free or in micelles presented effective antileishmanial action against both parasite stages, being more effective in amastigotes. In vivo data showed that treatment using QDS3 or QDS3/M reduced the parasite load in the livers, spleens, draining lymph nodes (dLN) and bone marrows of the treated animals, 1 and 15 days after treatment, when compared to values found in the control groups. Additionally, treated mice developed a polarized Th1-type immune response, with higher levels of IL-12, IFN-γ, GM-CSF and nitrite, besides high production of specific IgG2a antibodies, when compared to the controls. Parasitological and immunological data obtained using the micellar composition were better than the others. In conclusion, QDS3, mainly when applied in a delivery adjuvant system, could be considered for future studies as therapeutic candidate against VL.
Collapse
Affiliation(s)
- Luciana M Ribeiro Antinarelli
- Departamento de Parasitologia, Microbiologia E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, 36036-900, Brazil
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Nícolas Glanzmann
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, 36036-900, Brazil
| | - Débora V C Mendonça
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Daniela P Lage
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - João A Oliveira-da-Silva
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Grasiele S V Tavares
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Ana Maria R S Carvalho
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Camila S Freitas
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Vívian T Martins
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Mariana C Duarte
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
- Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Daniel Menezes-Souza
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
- Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Adilson David da Silva
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, 36036-900, Brazil
| | - Eduardo Antônio Ferraz Coelho
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia E Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
- Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Elaine Soares Coimbra
- Departamento de Parasitologia, Microbiologia E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, 36036-900, Brazil.
| |
Collapse
|
6
|
Perea-Martínez A, García-Hernández R, Manzano JI, Gamarro F. Transcriptomic Analysis in Human Macrophages Infected with Therapeutic Failure Clinical Isolates of Leishmania infantum. ACS Infect Dis 2022; 8:800-810. [PMID: 35352952 PMCID: PMC9003231 DOI: 10.1021/acsinfecdis.1c00513] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Leishmaniasis is one of the neglected tropical diseases with a worldwide distribution, affecting humans and animals. In the absence of an effective vaccine, current treatment is through the use of chemotherapy; however, existing treatments have frequent appearance of drug resistance and therapeutic failure (TF). The identification of factors that contribute to TF in leishmaniasis will provide the basis for a future therapeutic strategy more efficient for the control of this disease. In this article, we have evaluated the transcriptomic changes in the host cells THP-1 after infection with clinical Leishmania infantum isolates from leishmaniasis patients with TF. Our results show that distinct L. infantum isolates differentially modulate host cell response, inducing phenotypic changes that probably may account for parasite survival and TF of patients. Analysis of differential expression genes (DEGs), with a statistical significance threshold of a fold change ≥ 2 and a false discovery rate value ≤ 0.05, revealed a different number of DEGs according to the Leishmanialine. Globally, there was a similar number of genes up- and downregulated in all the infected host THP-1 cells, with exception of Hi-L2221, which showed a higher number of downregulated DEGs. We observed a total of 58 DEGs commonly modulated in all infected host cells, including upregulated (log2FC ≥ 1) and downregulated (log2FC ≤ -1) genes. Based on the results obtained from the analysis of RNA-seq, volcano plot, and GO enrichment analysis, we identified the most significant transcripts of relevance for their possible contribution to the TF observed in patients with leishmaniasis.
Collapse
Affiliation(s)
- Ana Perea-Martínez
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Avda del Conocimiento 17, 18016 Armilla, Granada, Spain
| | - Raquel García-Hernández
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Avda del Conocimiento 17, 18016 Armilla, Granada, Spain
| | - José Ignacio Manzano
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Avda del Conocimiento 17, 18016 Armilla, Granada, Spain
| | - Francisco Gamarro
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Avda del Conocimiento 17, 18016 Armilla, Granada, Spain
| |
Collapse
|
7
|
Total Synthesis of the Natural Chalcone Lophirone E, Synthetic Studies toward Benzofuran and Indole-Based Analogues, and Investigation of Anti-Leishmanial Activity. Molecules 2022; 27:molecules27020463. [PMID: 35056779 PMCID: PMC8778746 DOI: 10.3390/molecules27020463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/02/2022] [Accepted: 01/06/2022] [Indexed: 11/24/2022] Open
Abstract
The potential of natural and synthetic chalcones as therapeutic leads against different pathological conditions has been investigated for several years, and this class of compounds emerged as a privileged chemotype due to its interesting anti-inflammatory, antimicrobial, antiviral, and anticancer properties. The objective of our study was to contribute to the investigation of this class of natural products as anti-leishmanial agents. We aimed at investigating the structure–activity relationships of the natural chalcone lophirone E, characterized by the presence of benzofuran B-ring, and analogues on anti-leishmania activity. Here we describe an effective synthetic strategy for the preparation of the natural chalcone lophirone E and its application to the synthesis of a small set of chalcones bearing different substitution patterns at both the A and heterocyclic B rings. The resulting compounds were investigated for their activity against Leishmania infantum promastigotes disclosing derivatives 1 and 28a,b as those endowed with the most interesting activities (IC50 = 15.3, 27.2, 15.9 μM, respectively). The synthetic approaches here described and the early SAR investigations highlighted the potential of this class of compounds as antiparasitic hits, making this study worthy of further investigation.
Collapse
|
8
|
Galbiati A, Zana A, Coser C, Tamborini L, Basilico N, Parapini S, Taramelli D, Conti P. Development of Potent 3-Br-isoxazoline-Based Antimalarial and Antileishmanial Compounds. ACS Med Chem Lett 2021; 12:1726-1732. [PMID: 34795860 DOI: 10.1021/acsmedchemlett.1c00354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/07/2021] [Indexed: 11/30/2022] Open
Abstract
Starting from the structure of previously reported 3-Br-isoxazoline-based covalent inhibitors of P. falciparum glyceraldehyde 3-phosphate dehydrogenase, and with the intent to improve their metabolic stability and antimalarial activity, we designed and synthesized a series of simplified analogues that are characterized by the insertion of the oxadiazole ring as a bioisosteric replacement for the metabolically labile ester/amide function. We then further replaced the oxadiazole ring with a series of five-membered heterocycles and finally combined the most promising structural features. All the new derivatives were tested in vitro for antimalarial as well as antileishmanial activity. We identified two very promising new lead compounds, endowed with submicromolar antileishmanial activity and nanomolar antiplasmodial activity, respectively, and a very high selectivity index with respect to mammalian cells.
Collapse
Affiliation(s)
- Andrea Galbiati
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, Milano 20133, Italy
- Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Università degli Studi di Milano, Via Pascal 36, Milano 20133, Italy
| | - Aureliano Zana
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, Milano 20133, Italy
| | - Consuelo Coser
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, Milano 20133, Italy
| | - Lucia Tamborini
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, Milano 20133, Italy
- Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Università degli Studi di Milano, Via Pascal 36, Milano 20133, Italy
| | - Nicoletta Basilico
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Via Pascal 36, Milano 20133, Italy
- Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Università degli Studi di Milano, Via Pascal 36, Milano 20133, Italy
| | - Silvia Parapini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Pascal 36, Milano 20133, Italy
- Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Università degli Studi di Milano, Via Pascal 36, Milano 20133, Italy
| | - Donatella Taramelli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Pascal 36, Milano 20133, Italy
- Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Università degli Studi di Milano, Via Pascal 36, Milano 20133, Italy
| | - Paola Conti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, Milano 20133, Italy
- Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Università degli Studi di Milano, Via Pascal 36, Milano 20133, Italy
| |
Collapse
|
9
|
Glanzmann N, Antinarelli LMR, da Costa Nunes IK, Pereira HMG, Coelho EAF, Coimbra ES, da Silva AD. Synthesis and biological activity of novel 4-aminoquinoline/1,2,3-triazole hybrids against Leishmania amazonensis. Biomed Pharmacother 2021; 141:111857. [PMID: 34323702 DOI: 10.1016/j.biopha.2021.111857] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 01/07/2023] Open
Abstract
Quinoline and 1,2,3-triazoles are well-known nitrogen-based heterocycles presenting diverse pharmacological properties, although their antileishmanial activity is still poorly exploited. As an effort to contribute with studies involving these interesting chemical groups, in the present study, a series of compounds derived from 4-aminoquinoline and 1,2,3-triazole were synthetized and biological studies using L. amazonensis species were performed. The results pointed that the derivative 4, a hybrid of 4-aminoquinoline/1,2,3-triazole exhibited the best antileishmanial action, with inhibitory concentration (IC50) values of ~1 µM against intramacrophage amastigotes of L. amazonensis , and being 16-fold more active to parasites than to the host cell. The mechanism of action of derivative 4 suggest a multi-target action on Leishmania parasites, since the treatment of L. amazonensis promastigotes caused mitochondrial membrane depolarization, accumulation of ROS products, plasma membrane permeabilization, increase in neutral lipids, exposure of phosphatidylserine to the cell surface, changes in the cell cycle and DNA fragmentation. The results suggest that the antileishmanial effect of this compound is primarily altering critical biochemical processes for the correct functioning of organelles and macromolecules of parasites, with consequent cell death by processes related to apoptosis-like and necrosis. No up-regulation of reactive oxygen and nitrogen intermediates was promoted by derivative 4 on L. amazonensis -infected macrophages, suggesting a mechanism of action independent from the activation of the host cell. In conclusion, data suggest that derivative 4 presents selective antileishmanial effect, which is associated with multi-target action, and can be considered for future studies for the treatment against disease.
Collapse
Affiliation(s)
- Nícolas Glanzmann
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais 36.036-900, Brazil
| | - Luciana Maria Ribeiro Antinarelli
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais 36.036-900, Brazil; Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 30.130-100, Brazil
| | - Isabelle Karine da Costa Nunes
- Laboratório de Apoio ao Desenvolvimento Tecnológico, Polo de Química, Universidade Federal do Rio de Janeiro, Cidade Universitária Ilha do Fundão, Rio de Janeiro 21.941-598, Brazil
| | - Henrique Marcelo Gualberto Pereira
- Laboratório de Apoio ao Desenvolvimento Tecnológico, Polo de Química, Universidade Federal do Rio de Janeiro, Cidade Universitária Ilha do Fundão, Rio de Janeiro 21.941-598, Brazil
| | - Eduardo Antonio Ferraz Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 30.130-100, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Elaine Soares Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais 36.036-900, Brazil
| | - Adilson David da Silva
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais 36.036-900, Brazil.
| |
Collapse
|
10
|
Saresella M, Basilico N, Marventano I, Perego F, La Rosa F, Piancone F, Taramelli D, Banks H, Clerici M. Leishmania infantum infection reduces the amyloid β 42-stimulated NLRP3 inflammasome activation. Brain Behav Immun 2020; 88:597-605. [PMID: 32335194 DOI: 10.1016/j.bbi.2020.04.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/09/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
Activation of the NLRP3 inflammasome has been shown to play a major role in the neuroinflammation that accompanies Alzheimer's disease (AD); interventions that down regulate the NLRP3 inflammasome could thus be beneficial in AD. Parasite infections were recently shown to be associated with improved cognitive functions in Apolipoprotein E4 (ApoE4)-expressing members of an Amazonian tribe. We verified in an in vitro model whether Leishmania infantum infection could reduce NLRP3. Results obtained in an initial experimental model in which PBMC were LPS primed and nigericin-stimulated showed that L. infantum infection significantly reduced ASC-speck formation (i.e. intracellular inflammasome proteins assembly), as well as the production of activated caspase 5 and IL-1β, but increased that of activated caspase 1 and IL-18. Moreover, L. infantum infection induced the generation of an anti-inflammatory milieu by suppressing the production of TNFα and increasing that of IL-10. These results were replicated when cells that had been LPS-primed were stimulated with Aβ42 and infected with L. infantum. Results herein indicate that Leishmania infection favors an anti-inflammatory milieu, which includes the down-regulation of NLRP3 inflammasome activation, possibly to facilitate its survival inside host cells. A side effect of Leishmaniasis would be the hampering of neuroinflammation; this could play a protective role against AD development.
Collapse
Affiliation(s)
| | - Nicoletta Basilico
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy.
| | | | - Federica Perego
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy.
| | | | | | - Donatella Taramelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20100 Milan, Italy.
| | - Helen Banks
- Centre for Research on Health and Social Care Management (Cergas), SDA Bocconi School of Management, Milan 20100, Italy.
| | - Mario Clerici
- IRCCS Fondazione don Carlo Gnocchi, 20148 Milan, Italy; Department of Physiopathology and Transplants, University of Milan, 20122 Milan, Italy.
| |
Collapse
|
11
|
Methyl gallate: Selective antileishmanial activity correlates with host-cell directed effects. Chem Biol Interact 2020; 320:109026. [DOI: 10.1016/j.cbi.2020.109026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/11/2020] [Accepted: 02/25/2020] [Indexed: 11/22/2022]
|
12
|
He J, Zheng S. NF-κB Phosphorylation Inhibition Prevents Articular Cartilage Degradation in Osteoarthritis Rats via 2-Aminoquinoline. Med Sci Monit 2020; 26:e920346. [PMID: 31978040 PMCID: PMC6998790 DOI: 10.12659/msm.920346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background Osteoarthritis is a chronic degenerative disease of the joints that is common in older people worldwide. The characteristic features of osteoarthritis include cartilage degradation, synovitis, and remodelling of subchondral bone. The present study investigated the effect of 2-aminoquinoline on knee articular cartilage degradation in an osteoarthritis rat model. Material/Methods The rat model of osteoarthritis was established in Wistar rats by intra-articular injection of monosodium iodoacetate. The rats were randomly divided into 6 groups of 10 rats each: a normal control group, an untreated group, and 4 (5, 10, 15 and 20 mg/kg) treatment groups. The rats in treatment groups received 5, 10, 15, or 20 mg/kg doses of 2-aminoquinoline on day 2 of monosodium iodoacetate injection. Results The 2-aminoquinoline treatment of monosodium iodoacetate-injected rats markedly decreased weight-bearing asymmetry, inhibited edema formation, and improved paw withdrawal thresholds. The expression of inflammatory cytokines was markedly higher in the osteoarthritis rats. Treatment with 2-aminoquinoline led to a significant reduction in inflammatory cytokine expression in osteoarthritis rats in a dose-dependent manner. In osteoarthritis rats, the expressions of prostaglandin E2 (PGE2), matrix metalloproteinase-13 (MMP-13), and substance P were also higher in comparison to the control group. The 2-aminoquinoline treatment supressed PGE2, MMP-13, and substance P levels in osteoarthritis rats. Moreover, the expression of phosphorylated nuclear factor kappaB (p-NF-κB) was markedly higher in the untreated rats. However, activation of NF-κB was downregulated in the osteoarthritis rats by treatment with 2-aminoquinoline. Conclusions The present study demonstrated that 2-aminoquinoline prevents articular cartilage damage in osteoarthritis rats through inhibition of inflammatory factors and downregulation of NF-κB activation, suggesting that 2-aminoquinoline would be effective in treatment of osteoarthritis.
Collapse
Affiliation(s)
- Jinlong He
- Department of Integrated Traditional Chinese Medicine (TCM) and Western Medicine Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Shicheng Zheng
- Department of Integrated Traditional Chinese Medicine (TCM) and Western Medicine Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|