1
|
Lemos ASDO, Granato JDT, Antinarelli LMR, Machado PDA, Campos LM, Bastos JPRC, Midlej VDV, Silva Neto AFD, Fabri RL, Coimbra ES. Lantana camara L. induces a multi-targeted cell death process in Leishmania amazonensis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118766. [PMID: 39222759 DOI: 10.1016/j.jep.2024.118766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/13/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
ETNOPHARMACOLOGICAL RELEVANCE Lantana camara L. is a species known for its broad spectrum of bioactivities and is commonly used in folk therapy to address inflammatory, dermatological, gastrointestinal, intestinal worms and protozoan diseases. It boasts a diverse array of secondary metabolites such as terpenes, flavonoids, and saponins. However, despite its rich chemical profile, there remains a scarcity of studies investigating its antileishmanial properties. AIM OF THE STUDY This research aims to explore the antileishmanial potential of L. camara, focusing also on its mechanism of action against Leishmania amazonensis. MATERIAL AND METHODS The ethanolic extract of L. camara leaves (LCE) was obtained through static maceration, and its phytoconstituents were identified using UFLC-QTOF-MS. The colorimetric MTT method was conducted to determine the effect of LCE on promastigotes of L. amazonensis and murine macrophages. The anti-amastigote activity was evaluated by counting intracellular parasites in macrophages after Giemsa staining. Additionally, investigations into the mechanisms underlying its action were conducted using cellular and biochemical approaches. RESULTS LCE exhibited significant activity against both promastigotes and intracellular amastigotes of L. amazonensis, with IC50 values of 12.20 μg/mL ± 0.12 and 7.09 μg/mL ± 1.24, respectively. These IC50 values indicate very promising antileishmanial activity, comparable to those found for the positive control miltefosine (5.10 μg/mL ± 1.79 and 8.96 μg/mL ± 0.50, respectively). Notably, LCE exhibited negligible cytotoxicity on macrophages (IC50 = 223.40 μg/mL ± 47.02), demonstrating selectivity towards host cells (SI = 31.50). The antileishmanial activity of LCE involved a multi-targeted cell death process, characterized by morphological and ultrastructural alterations observed through SEM and TEM analyses, as well as oxidative effects evidenced by the inhibition of trypanothione reductase, elevation of ROS and lipid levels, and mitochondrial dysfunction evaluated using DTNB, H2DCFDA, Nile red, and JC-1 assays. Additionally, extraction of ergosterol and double labeling with annexin V and PI revealed modifications to the organization and permeability of the treated parasite's plasma membrane. LCE was found to consist predominantly of terpenes, with lantadenes A, B, and C being among the eleven compounds identified through UFLC-QTOF-MS analysis. CONCLUSIONS The extract of L. camara presents a diverse array of chemical constituents, prominently featuring high terpene content, which may underlie its antileishmanial properties through a combination of apoptotic and non-apoptotic mechanisms of cell death induced by LCE. This study underscores the therapeutic potential of L. camara as a candidate for antileishmanial treatment, pending further validation.
Collapse
Affiliation(s)
- Ari Sérgio de Oliveira Lemos
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | - Juliana da Trindade Granato
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | | | - Patrícia de Almeida Machado
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | - Lara Melo Campos
- Bioactive Natural Products Laboratory, Department of Biochemistry, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, CEP 36036-900, Brazil
| | - João Pedro Reis Costa Bastos
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | - Victor do Valle Midlej
- Laboratory of Cellular and Ultrastructure, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, 21040-900, Brazil
| | - Adolfo Firmino da Silva Neto
- Department of Veterinary Medicine, Faculty of Medicine, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | - Rodrigo Luiz Fabri
- Laboratory of Bioactive Natural Products, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, CEP 36036-900, Brazil
| | - Elaine Soares Coimbra
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil.
| |
Collapse
|
2
|
Granato JDT, Silva ETD, Lemos ASDO, Machado PDA, Midlej VDV, Antinarelli LMR, Silva Neto AFD, Souza MVN, Coimbra ES. 4-Quinolinylhydrazone analogues kill Leishmania (Leishmania) amazonensis by inducing apoptosis and mitochondria-dependent pathway cell death. Chem Biol Drug Des 2024; 103:e14535. [PMID: 38772877 DOI: 10.1111/cbdd.14535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/03/2024] [Accepted: 04/23/2024] [Indexed: 05/23/2024]
Abstract
Despite efforts, available alternatives for the treatment of leishmaniasis are still scarce. In this work we tested a class of 15 quinolinylhydrazone analogues and presented data that support the use of the most active compound in cutaneous leishmaniasis caused by Leishmania amazonensis. In general, the compounds showed activity at low concentrations for both parasitic forms (5.33-37.04 μM to promastigotes, and 14.31-61.98 μM to amastigotes). In addition, the best compound (MHZ15) is highly selective for the parasite. Biochemical studies indicate that the treatment of promastigotes with MHZ15 leads the loss of mitochondrial potential and increase in ROS levels as the primary effects, which triggers accumulation of lipid droplets, loss of plasma membrane integrity and apoptosis hallmarks, including DNA fragmentation and phosphatidylserine exposure. These effects were similar in the intracellular form of the parasite. However, in this parasitic form there is no change in plasma membrane integrity in the observed treatment time, which can be attributed to metabolic differences and the resilience of the amastigote. Also, ultrastructural changes such as vacuolization suggesting autophagy were observed. The in vivo effectiveness of MHZ15 in the experimental model of cutaneous leishmaniasis was carried out in mice of the BALB/c strain infected with L. amazonensis. The treatment by intralesional route showed that MHZ15 acted with great efficiency with significantly reduction in the parasite load in the injured paws and draining lymph nodes, without clinical signs of distress or compromise of animal welfare. In vivo toxicity was also evaluated and null alterations in the levels of hepatic enzymes aspartate aminotransferase, and alanine aminotransferase was observed. The data presented herein demonstrates that MHZ15 exhibits a range of favorable characteristics conducive to the development of an antileishmanial agent.
Collapse
Affiliation(s)
- Juliana da Trindade Granato
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Emerson Teixeira da Silva
- Fundação Oswaldo Cruz (Fiocruz), Instituto de Tecnologia em Fármacos Farmanguinhos, Rio de Janeiro, Brazil
| | - Ari Sérgio de Oliveira Lemos
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Patrícia de Almeida Machado
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Victor do Valle Midlej
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Luciana Maria Ribeiro Antinarelli
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Adolfo Firmino da Silva Neto
- Departamento de Medicina Veterinária, Faculdade de Medicina, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Marcus Vinícius Nora Souza
- Fundação Oswaldo Cruz (Fiocruz), Instituto de Tecnologia em Fármacos Farmanguinhos, Rio de Janeiro, Brazil
| | - Elaine Soares Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| |
Collapse
|
3
|
Jorge J, Del Pino Santos KF, Timóteo F, Vasconcelos RRP, Ayala Cáceres OI, Granja IJA, de Souza DM, Frizon TEA, Di Vaccari Botteselle G, Braga AL, Saba S, Rashid HU, Rafique J. Recent Advances on the Antimicrobial Activities of Schiff Bases and their Metal Complexes: An Updated Overview. Curr Med Chem 2024; 31:2330-2344. [PMID: 36823995 DOI: 10.2174/0929867330666230224092830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/12/2022] [Accepted: 12/29/2022] [Indexed: 02/25/2023]
Abstract
Schiff bases represent a valuable class of organic compounds, synthesized via condensation of primary amines with ketones or aldehydes. They are renowned for possessing innumerable applications in agricultural chemistry, organic synthesis, chemical and biological sensing, coating, polymer and resin industries, catalysis, coordination chemistry, and drug designing. Schiff bases contain imine or azomethine (-C=N-) functional groups which are important pharmacophores for the design and synthesis of lead bioactive compounds. In medicinal chemistry, Schiff bases have attracted immense attention due to their diverse biological activities. This review aims to encompass the recent developments on the antimicrobial activities of Schiff bases. The article summarizes the antibacterial, antifungal, antiviral, antimalarial, and antileishmanial activities of Schiff bases reported since 2011.
Collapse
Affiliation(s)
- Juliana Jorge
- Instituto de Química, Universidade Federal do Mato Grosso do Sul, Campo Grande, 79074-460, MS, Brazil
| | | | - Fernanda Timóteo
- Instituto de Química, Universidade Federal do Mato Grosso do Sul, Campo Grande, 79074-460, MS, Brazil
| | | | | | | | - David Monteiro de Souza
- Instituto de Química, Universidade Federal do Mato Grosso do Sul, Campo Grande, 79074-460, MS, Brazil
| | - Tiago Elias Allievi Frizon
- Department of Energy and Sustainability, Universidade Federal de Santa Catarina - UFSC, Campus Araranguá, Araranguá, 88905-120, SC, Brazil
| | | | - Antonio Luiz Braga
- Departamento de Química, Universidade Federal de Santa Catarina, 88040-970, Florianópolis, SC, Brazil
| | - Sumbal Saba
- Instituto de Química, Universidade Federal de Goiás - UFG, Goiânia, 74690-900, GO, Brazil
| | - Haroon Ur Rashid
- Instituto de Química, Universidade Federal do Mato Grosso do Sul, Campo Grande, 79074-460, MS, Brazil
- Departamento de Química, Universidade Federal de Santa Catarina, 88040-970, Florianópolis, SC, Brazil
| | - Jamal Rafique
- Instituto de Química, Universidade Federal do Mato Grosso do Sul, Campo Grande, 79074-460, MS, Brazil
- Instituto de Química, Universidade Federal de Goiás - UFG, Goiânia, 74690-900, GO, Brazil
| |
Collapse
|
4
|
Lemos ASDO, Campos LM, Granato JDT, Goliatt PVZC, Dib PRB, Hottz ED, Glanzmann N, Campos LC, Bizarro HDS, Chedier LM, Coimbra ES, Fabri RL. Mitracarpus frigidus reduces lipid metabolism and PGE2 levels in inflammatory cells. J Pharm Pharmacol 2023; 75:1388-1393. [PMID: 37487573 DOI: 10.1093/jpp/rgad069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023]
Abstract
OBJECTIVES To evaluate the ability of the aqueous extract of Mitracarpus frigidus (MFAq) to inhibit lipid body formation and inflammatory mediator production in macrophages stimulated with lipopolysaccharide (LPS) and interferon gamma (IFN-γ). METHODS MFAq was chemically characterized by ultrafast liquid chromatography/quadruple time-of-flight tandem mass spectrometry. The macrophages obtained from mice were incubated with MFAq. Cell viability and membrane integrity were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and propidium iodide assays, respectively. Moreover, NO, reactive oxygen species (ROS), transforming growth factor beta (TGF-β), prostaglandin E2 (PGE2) levels and lipid bodies (LBs) were examined in macrophages that were stimulated with LPS and IFN-γ and treated with MFAq. Finally, molecular docking analysis was conducted to investigate the interaction of MFAq with the cyclooxygenase 2 (COX-2) enzyme. KEY FINDINGS Chlorogenic acid, clarinoside, harounoside, rutin, kaempferol-3O-rutinoside and 2-azaanthraquinone were identified in MFAq. MFAq significantly inhibited NO, ROS and LBs, and did not affect the membrane integrity of macrophages. MFAq-treated cells showed significantly lower levels of TGF-β and PGE2. Molecular docking demonstrated that the compounds found in MFAq are able to inhibit COX-2 by binding to important residues in the catalytic site. CONCLUSIONS MFAq interferes with lipid metabolism in stimulated macrophages, leading to the reduction of important inflammatory mediators. Furthermore, MFAq can directly inhibit the COX-2 enzyme or inhibit its expression owing to its ability to reduce NO production.
Collapse
Affiliation(s)
- Ari Sérgio de O Lemos
- Bioactive Natural Products Laboratory, Department of Biochemistry, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Lara M Campos
- Bioactive Natural Products Laboratory, Department of Biochemistry, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Juliana da T Granato
- Department of Parasitology, Microbiology, and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Priscila V Z C Goliatt
- Department of Computer Science, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Paula R B Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eugenio D Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Nícolas Glanzmann
- Department of Chemistry, Institute of Exact Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Laíris C Campos
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Heloisa D S Bizarro
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Luciana M Chedier
- Plant Chemistry Laboratory, Department of Botany, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Elaine S Coimbra
- Department of Parasitology, Microbiology, and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Rodrigo L Fabri
- Bioactive Natural Products Laboratory, Department of Biochemistry, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
5
|
Baldissera FG, Fazolo T, da Silva MB, de Santana Filho PC, da Silva VD, Rivillo Perez DM, Klitzke JS, de Oliveira Soares EG, Rodrigues Júnior LC, Peres A, Dallegrave E, Navegantes-Lima KC, Monteiro MC, Schrekker HS, Torres Romão PR. Imidazolium salts as an alternative for anti-Leishmania drugs: Oxidative and immunomodulatory activities. Front Immunol 2023; 13:1096312. [PMID: 36733394 PMCID: PMC9886892 DOI: 10.3389/fimmu.2022.1096312] [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: 11/14/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023] Open
Abstract
In this study we explored the previously established leishmanicidal activity of a complementary set of 24 imidazolium salts (IS), 1-hexadecylimidazole (C16Im) and 1-hexadecylpyridinium chloride (C16PyrCl) against Leishmania (Leishmania) amazonensis and Leishmania (Leishmania) infantum chagasi. Promastigotes of L. amazonensis and L. infantum chagasi were incubated with 0.1 to 100 μM of the compounds and eight of them demonstrated leishmanicidal activity after 48 h - C10MImMeS (IC50 L. amazonensis = 11.6), C16MImPF6(IC50 L. amazonensis = 6.9), C16MImBr (IC50 L. amazonensis = 6), C16M2ImCl (IC50 L. amazonensis = 4.1), C16M4ImCl (IC50 L. amazonensis = 1.8), (C10)2MImCl (IC50 L. amazonensis = 1.9), C16Im (IC50 L. amazonensis = 14.6), and C16PyrCl (IC50 L. amazonensis = 4).The effect of IS on reactive oxygen species production, mitochondrial membrane potential, membrane integrity and morphological alterations of promastigotes was determined, as well as on L. amazonensis-infected macrophages. Their cytotoxicity against macrophages and human erythrocytes was also evaluated. The IS C10MImMeS, C16MImPF6, C16MImBr, C16M2ImCl, C16M4ImCl and (C10)2MImCl, and the compounds C16Im and C16PyrCl killed and inhibited the growth of promastigote forms of L. amazonensis and L. infantum chagasi in a concentration-dependent manner, contributing to a better understanding of the structure-activity relationship of IS against Leishmania. These IS induced ROS production, mitochondrial dysfunction, membrane disruption and morphological alterations in infective forms of L. amazonensis and killed intracellular amastigote forms in very low concentrations (IC50 amastigotes ≤ 0.3), being potential drug candidates against L. amazonensis.
Collapse
Affiliation(s)
- Fernanda Giesel Baldissera
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil,Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Tiago Fazolo
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil,Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Matheus Brasil da Silva
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Paulo Cesar de Santana Filho
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Vinícius Demétrio da Silva
- Laboratory of Technological Processes and Catalysis, Institute of Chemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - David Max Rivillo Perez
- Laboratory of Technological Processes and Catalysis, Institute of Chemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Joice Sandra Klitzke
- Laboratory of Technological Processes and Catalysis, Institute of Chemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Eduardo Giovanni de Oliveira Soares
- Laboratory of Technological Processes and Catalysis, Institute of Chemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Luiz Carlos Rodrigues Júnior
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil,Graduate Program in Pharmaceutical Science, Graduate Program in Neuroscience and Cellular Biology, Faculty of Pharmacy, Universidade Federal do Pará, Belém, PA, Brazil
| | - Alessandra Peres
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil,Graduate Program in Pharmaceutical Science, Graduate Program in Neuroscience and Cellular Biology, Faculty of Pharmacy, Universidade Federal do Pará, Belém, PA, Brazil
| | - Eliane Dallegrave
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Kely Campos Navegantes-Lima
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Marta Chagas Monteiro
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil,*Correspondence: Henri Stephan Schrekker, ; Marta Chagas Monteiro,
| | - Henri Stephan Schrekker
- Laboratory of Technological Processes and Catalysis, Institute of Chemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil,*Correspondence: Henri Stephan Schrekker, ; Marta Chagas Monteiro,
| | - Pedro Roosevelt Torres Romão
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil,Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil,Graduate Program in Pharmaceutical Science, Graduate Program in Neuroscience and Cellular Biology, Faculty of Pharmacy, Universidade Federal do Pará, Belém, PA, Brazil
| |
Collapse
|
6
|
Chaiswing L, Xu F, Zhao Y, Thorson J, Wang C, He D, Lu J, Ellingson SR, Zhong W, Meyer K, Luo W, St. Clair W, Clair DS. The RelB-BLNK Axis Determines Cellular Response to a Novel Redox-Active Agent Betamethasone during Radiation Therapy in Prostate Cancer. Int J Mol Sci 2022; 23:ijms23126409. [PMID: 35742868 PMCID: PMC9223669 DOI: 10.3390/ijms23126409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022] Open
Abstract
Aberrant levels of reactive oxygen species (ROS) are potential mechanisms that contribute to both cancer therapy efficacy and the side effects of cancer treatment. Upregulation of the non-canonical redox-sensitive NF-kB family member, RelB, confers radioresistance in prostate cancer (PCa). We screened FDA-approved compounds and identified betamethasone (BET) as a drug that increases hydrogen peroxide levels in vitro and protects non-PCa tissues/cells while also enhancing radiation killing of PCa tissues/cells, both in vitro and in vivo. Significantly, BET increases ROS levels and exerts different effects on RelB expression in normal cells and PCa cells. BET induces protein expression of RelB and RelB target genes, including the primary antioxidant enzyme, manganese superoxide dismutase (MnSOD), in normal cells, while it suppresses protein expression of RelB and MnSOD in LNCaP cells and PC3 cells. RNA sequencing analysis identifies B-cell linker protein (BLNK) as a novel RelB complementary partner that BET differentially regulates in normal cells and PCa cells. RelB and BLNK are upregulated and correlate with the aggressiveness of PCa in human samples. The RelB-BLNK axis translocates to the nuclear compartment to activate MnSOD protein expression. BET promotes the RelB-BLNK axis in normal cells but suppresses the RelB-BLNK axis in PCa cells. Targeted disruptions of RelB-BLNK expressions mitigate the radioprotective effect of BET on normal cells and the radiosensitizing effect of BET on PCa cells. Our study identified a novel RelB complementary partner and reveals a complex redox-mediated mechanism showing that the RelB-BLNK axis, at least in part, triggers differential responses to the redox-active agent BET by stimulating adaptive responses in normal cells but pushing PCa cells into oxidative stress overload.
Collapse
Affiliation(s)
- Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
- Correspondence: (L.C.); (D.S.C.)
| | - Fangfang Xu
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
| | - Yanming Zhao
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
| | - Jon Thorson
- Center for Pharmaceutical Research and Innovation, Lexington, KY 40536, USA;
- College of Pharmacy, Pharmaceutical Sciences Department, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Daheng He
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Jinpeng Lu
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Sally R. Ellingson
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Weixiong Zhong
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (W.Z.); (K.M.)
| | - Kristy Meyer
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (W.Z.); (K.M.)
| | - Wei Luo
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA; (W.L.); (W.S.C.)
| | - William St. Clair
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA; (W.L.); (W.S.C.)
| | - Daret St. Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
- Correspondence: (L.C.); (D.S.C.)
| |
Collapse
|
7
|
Almeida FS, Sousa GLS, Rocha JC, Ribeiro FF, de Oliveira MR, de Lima Grisi TCS, Araújo DAM, de C Nobre MS, Castro RN, Amaral IPG, Keesen TSL, de Moura RO. In vitro anti-Leishmania activity and molecular docking of spiro-acridine compounds as potential multitarget agents against Leishmania infantum. Bioorg Med Chem Lett 2021; 49:128289. [PMID: 34311084 DOI: 10.1016/j.bmcl.2021.128289] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 11/28/2022]
Abstract
Leishmaniasis is an infectious disease with several limitations regarding treatment schemes. This work reports the anti-Leishmania activity of spiroacridine compounds against the promastigote (IC50 = 1.1 to 6.0 µg / mL) and amastigote forms of the best compounds (EC50 = 4.9 and 0.9 µg / mL) inLeishmania (L.) infantumand proposes an in-silico study with possible selective therapeutic targets for L. infantum. The substituted dimethyl-amine compound (AMTAC 11) showed the best leishmanicidal activity in vitro, and was found to interact with TryRandLdTopoI. comparisons with standard inhibitors were performed, and its main interactions were elucidated. Based on the biological assessment and the structure-activity relationship study, the spiroacridine compounds appear to be promisinganti-leishmaniachemotherapeutic agents to be explored.
Collapse
Affiliation(s)
- Fernanda S Almeida
- Programa de Doutorado em Biotecnologia, Rede Nordeste de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil; Laboratório de Imunologia das Doenças Infeciosas, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Gleyton L S Sousa
- Programa de Doutorado em Química, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ 23897-000, Brazil
| | - Juliana C Rocha
- Laboratório de Imunologia das Doenças Infeciosas, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Frederico F Ribeiro
- Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, João Pessoa, PB 58059-900, Brazil
| | - Márcia Rosa de Oliveira
- Departamento de Biologia Molecular, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, Joao Pessoa, Paraíba CEP 58059-900, Brazil
| | | | - Demetrius A M Araújo
- Departamento de Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, PB 58059-900, Brazil
| | - Michelangela S de C Nobre
- Programa de Doutorado em Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, PE 50670-901, Brazil
| | - Rosane N Castro
- Programa de Doutorado em Química, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ 23897-000, Brazil
| | - Ian P G Amaral
- Departamento de Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, PB 58059-900, Brazil
| | - Tatjana S L Keesen
- Departamento de Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, PB 58059-900, Brazil; Laboratório de Imunologia das Doenças Infeciosas, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Ricardo Olímpio de Moura
- Centro de Ciências Biológicas e da Saúde, Universidade Estadual da Paraíba, Campina Grande, PB 58429-500, Brazil.
| |
Collapse
|
8
|
Kaushik D, Granato JT, Macedo GC, Dib PRB, Piplani S, Fung J, da Silva AD, Coimbra ES, Petrovsky N, Salunke DB. Toll-like receptor-7/8 agonist kill Leishmania amazonensis by acting as pro-oxidant and pro-inflammatory agent. J Pharm Pharmacol 2021; 73:1180-1190. [PMID: 33940589 PMCID: PMC8359742 DOI: 10.1093/jpp/rgab063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 03/29/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Evaluation of the anti-Leishmanial activity of imidazoquinoline-based TLR7/8 agonists. METHODS TLR7/8-active imidazoquinolines (2 and 3) were synthesized and assessed for activity against Leishmania amazonensis-intracellular amastigotes using mouse peritoneal macrophages. The production of reactive oxygen species (ROS), nitric oxide (NO) and cytokines was determined in infected and non-infected macrophages. KEY FINDINGS The imidazoquinolines, 2 and 3, were primarily agonists of TLR7 with compound 3 also showing modest TLR8 activity. Docking studies showed them to occupy the same binding pocket on TLR7 and 8 as the known agonists, imiquimod and resiquimod. Compounds 2 and 3 inhibited the growth of L. amazonensis-intracellular amastigotes with the most potent compound (3, IC50 = 5.93 µM) having an IC50 value close to miltefosine (IC50 = 4.05 µM), a known anti-Leishmanial drug. Compound 3 induced macrophages to produce ROS, NO and inflammatory cytokines that likely explain the anti-Leishmanial effects. CONCLUSIONS This study shows that activating TLR7 using compounds 2 or 3 induces anti-Leishmanial activity associated with induction of free radicals and inflammatory cytokines able to kill the parasites. While 2 and 3 had a very narrow cytotoxicity window for macrophages, this identifies the possibility to further develop this chemical scaffold to less cytotoxic TLR7/8 agonist for potential use as anti-Leishmanial drug.
Collapse
Affiliation(s)
- Deepender Kaushik
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, India
| | - Juliana T Granato
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Gilson C Macedo
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Paula R B Dib
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Sakshi Piplani
- Vaxine Pty Ltd., Warradale, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Johnson Fung
- Vaxine Pty Ltd., Warradale, South Australia, Australia
| | - Adilson D da Silva
- Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Elaine S Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Warradale, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, India
- National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials (NICOVIA), Panjab University, Chandigarh, India
| |
Collapse
|
9
|
Freitas CS, Lage DP, Oliveira-da-Silva JA, Costa RR, Mendonça DVC, Martins VT, Reis TAR, Antinarelli LMR, Machado AS, Tavares GSV, Ramos FF, Brito RCF, Ludolf F, Chávez-Fumagalli MA, Roatt BM, Ramos GS, Munkert J, Ottoni FM, Campana PRV, Duarte MC, Gonçalves DU, Coimbra ES, Braga FC, Pádua RM, Coelho EAF. In vitro and in vivo antileishmanial activity of β-acetyl-digitoxin, a cardenolide of Digitalis lanata potentially useful to treat visceral leishmaniasis. ACTA ACUST UNITED AC 2021; 28:38. [PMID: 33851916 PMCID: PMC8045677 DOI: 10.1051/parasite/2021036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 04/01/2021] [Indexed: 12/11/2022]
Abstract
Current treatments of visceral leishmaniasis face limitations due to drug side effects and/or high cost, along with the emergence of parasite resistance. Novel and low-cost antileishmanial agents are therefore required. We report herein the antileishmanial activity of β-acetyl-digitoxin (b-AD), a cardenolide isolated from Digitalis lanata leaves, assayed in vitro and in vivo against Leishmania infantum. Results showed direct action of b-AD against parasites, as well as efficacy for the treatment of Leishmania-infected macrophages. In vivo experiments using b-AD-containing Pluronic® F127 polymeric micelles (b-AD/Mic) to treat L. infantum-infected mice showed that this composition reduced the parasite load in distinct organs in more significant levels. It also induced the development of anti-parasite Th1-type immunity, attested by high levels of IFN-γ, IL-12, TNF-α, GM-CSF, nitrite and specific IgG2a antibodies, in addition to low IL-4 and IL-10 contents, along with higher IFN-γ-producing CD4+ and CD8+ T-cell frequency. Furthermore, low toxicity was found in the organs of the treated animals. Comparing the therapeutic effect between the treatments, b-AD/Mic was the most effective in protecting animals against infection, when compared to the other groups including miltefosine used as a drug control. Data found 15 days after treatment were similar to those obtained one day post-therapy. In conclusion, the results obtained suggest that b-AD/Mic is a promising antileishmanial agent and deserves further studies to investigate its potential to treat visceral leishmaniasis.
Collapse
Affiliation(s)
- 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, 30130-100 Minas Gerais, 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, 30130-100 Minas Gerais, 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, 30130-100 Minas Gerais, Brazil
| | - Rafaella R Costa
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, 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, 30130-100 Minas Gerais, 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, 30130-100 Minas Gerais, Brazil
| | - Thiago A R Reis
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Luciana M R Antinarelli
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, 36036-900 Minas Gerais, Brazil
| | - Amanda S Machado
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, 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, 30130-100 Minas Gerais, Brazil
| | - Fernanda F Ramos
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Rory C F Brito
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Fernanda Ludolf
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | | | - Bruno M Roatt
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Gabriela S Ramos
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Jennifer Munkert
- Departament Biologie, LS Pharmazeutische Biologie, Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Flaviano M Ottoni
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Priscilla R V Campana
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, 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, 30130-100 Minas Gerais, Brazil - Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Denise U Gonçalves
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Elaine S Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, 36036-900 Minas Gerais, Brazil
| | - Fernão C Braga
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Rodrigo M Pádua
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil - Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| |
Collapse
|
10
|
Boniface PK, Sano CM, Elizabeth FI. Unveiling the Targets Involved in the Quest of Antileishmanial Leads Using In silico Methods. Curr Drug Targets 2021; 21:681-712. [PMID: 32003668 DOI: 10.2174/1389450121666200128112948] [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] [Received: 10/08/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Leishmaniasis is a neglected tropical disease associated with several clinical manifestations, including cutaneous, mucocutaneous, and visceral forms. As currently available drugs have some limitations (toxicity, resistance, among others), the target-based identification has been an important approach to develop new leads against leishmaniasis. The present study aims to identify targets involved in the pharmacological action of potent antileishmanial compounds. METHODS The literature information regarding molecular interactions of antileishmanial compounds studied over the past half-decade is discussed. The information was obtained from databases such as Wiley, SciFinder, Science Direct, National Library of Medicine, American Chemical Society, Scientific Electronic Library Online, Scopus, Springer, Google Scholar, Web of Science, etc. Results: Numerous in vitro antileishmanial compounds showed affinity and selective interactions with enzymes such as arginase, pteridine reductase 1, trypanothione reductase, pyruvate kinase, among others, which are crucial for the survival and virulence of the Leishmania parasite. CONCLUSION The in-silico activity of small molecules (enzymes, proteins, among others) might be used as pharmacological tools to develop candidate compounds for the treatment of leishmaniasis. As some pharmacologically active compounds may act on more than one target, additional studies of the mechanism (s) of action of potent antileishmanial compounds might help to better understand their pharmacological action. Also, the optimization of promising antileishmanial compounds might improve their biological activity.
Collapse
Affiliation(s)
- Pone K Boniface
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Cinthya M Sano
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Ferreira I Elizabeth
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
11
|
Alves MA, de Queiroz AC, Leite AB, Martins FT, Doriguetto AC, Barreiro EJ, Alexandre-Moreira MS, Lima LM. Carbamoyl- N-aryl-imine-urea: a new framework to obtain a putative leishmanicidal drug-candidate. RSC Adv 2020; 10:12384-12394. [PMID: 35497630 PMCID: PMC9050848 DOI: 10.1039/d0ra00287a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/08/2020] [Indexed: 11/21/2022] Open
Abstract
Leishmaniasis is a neglected parasitic disease, and current treatment includes limitations of toxicity, variable efficacy, high costs and inconvenient doses and treatment schedules. Therefore, new leishmanicidal drugs are still an unquestionable medical need. In this paper we described the design conception of a new framework, the carbamoyl-N-aryl-imine-urea, to obtain putative leishmanicidal drug-candidates. Compounds 9a-e and 10a-e were designed and synthesized and their leishmanicidal activity was studied in comparison to pentamidine, miltefosine and meglumine antimoniate. The conformational profile of the new carbamoyl-N-aryl-imine-urea framework was investigated by X-ray diffraction studies, using compound 9a as a model. The plasma stability of this putative peptide mimetic subunit was studied for compound 10e (LASSBio-1736). Among the congeneric series, LASSBio-1736 was identified as a new antileishmanial drug-candidate, displaying plasma stability, cytotoxicity against amastigote forms of L. amazonensis and L. braziliensis, and leishmanicidal activity in a cutaneous leishmaniasis murine model, without preliminary evidence of hepatic or renal toxicity.
Collapse
Affiliation(s)
- Marina A Alves
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio, ®), Universidade Federal do Rio de Janeiro (UFRJ), CCS PO Box 68023, Cidade Universitária 21941-902 Rio de Janeiro RJ Brazil http://www.inct-inofar.ccs.ufrj.br http://www.lassbio.icb.ufrj.br.,Programa de Pós-graduação em Química, Instituto de Química, UFRJ 21941-909 Rio de Janeiro RJ Brazil
| | - Aline C de Queiroz
- Laboratório de Farmacologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas (UFAL) 57072-900 Maceió AL Brazil +55 82 3214 1528
| | - Anderson Brandão Leite
- Laboratório de Farmacologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas (UFAL) 57072-900 Maceió AL Brazil +55 82 3214 1528
| | - Felipe T Martins
- Instituto de Química, Universidade Federal de Goiás (UFG) Campus Samambaia, CP 131 Goiânia GO 74001-970 Brazil
| | - Antonio C Doriguetto
- Instituto de Química, Universidade Federal de Goiás (UFG) Campus Samambaia, CP 131 Goiânia GO 74001-970 Brazil
| | - Eliezer J Barreiro
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio, ®), Universidade Federal do Rio de Janeiro (UFRJ), CCS PO Box 68023, Cidade Universitária 21941-902 Rio de Janeiro RJ Brazil http://www.inct-inofar.ccs.ufrj.br http://www.lassbio.icb.ufrj.br.,Programa de Pós-graduação em Química, Instituto de Química, UFRJ 21941-909 Rio de Janeiro RJ Brazil
| | - Magna S Alexandre-Moreira
- Laboratório de Farmacologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas (UFAL) 57072-900 Maceió AL Brazil +55 82 3214 1528
| | - Lídia M Lima
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio, ®), Universidade Federal do Rio de Janeiro (UFRJ), CCS PO Box 68023, Cidade Universitária 21941-902 Rio de Janeiro RJ Brazil http://www.inct-inofar.ccs.ufrj.br http://www.lassbio.icb.ufrj.br.,Programa de Pós-graduação em Química, Instituto de Química, UFRJ 21941-909 Rio de Janeiro RJ Brazil
| |
Collapse
|
12
|
Synthesis, biological activity, and mechanism of action of new 2-pyrimidinyl hydrazone and N-acylhydrazone derivatives, a potent and new classes of antileishmanial agents. Eur J Med Chem 2019; 184:111742. [DOI: 10.1016/j.ejmech.2019.111742] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/12/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
|
13
|
Semicarbazone derivatives as promising therapeutic alternatives in leishmaniasis. Exp Parasitol 2019; 201:57-66. [PMID: 31004571 DOI: 10.1016/j.exppara.2019.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 03/09/2019] [Accepted: 04/12/2019] [Indexed: 01/28/2023]
Abstract
In the present study, we investigated the in vitro and in vivo leishmanicidal activity of synthetic compounds, containing a semicarbazone scaffold as a peptide mimetic framework. The leishmanicidal effect against amastigotes of Leishmania amazonensis was also evaluated at concentration of 100 μM-0.01 nM. The derivatives 2e, 2f, 2g and 1g, beyond the standards miltefosine and pentamidine, significantly diminished the number of L. amazonensis amastigotes in macrophages. These derivatives were also active against amastigotes of L. braziliensis. As 2g presented potent leishmanicidal activity against the amastigotes of L. amazonensis in macrophages, we also investigated the in vivo leishmanicidal activity of this compound against L. amazonensis. Approximately 105L. amazonensis promastigotes were subcutaneously inoculated into the dermis of the right ear of BALB/c mice, which were subsequently treated with 2g (p.o. or i.p.), miltefosine (p.o.) or glucantime (i.p.) at 30 μmol/kg/day x 28 days. Thus, a similar reduction in the lesion size was observed after the administration of 2g through oral (63.7 ± 10.1%) and intraperitoneal (61.8 ± 3.7%) routes. A larger effect was observed after treatment with miltefosine (97.7 ± 0.4%), and glucantime did not exhibit activity at the dose administered. With respect to the ear parasite load, 2g diminished the number of parasites by p.o. (30.5 ± 5.1%) and i.p. (33.3 ± 4.3%) administration. In addition, 2g induced in vitro apoptosis, autophagy and cell cycle alterations on L. amazonensis promastigotes. In summary, the derivative 2g might represent a lead candidate for antileishmanial drugs, as this compound displayed pronounced leishmanicidal activity.
Collapse
|
14
|
SB-83, a 2-Amino-thiophene derivative orally bioavailable candidate for the leishmaniasis treatment. Biomed Pharmacother 2018; 108:1670-1678. [DOI: 10.1016/j.biopha.2018.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/19/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022] Open
|
15
|
Ferreira LLG, Andricopulo AD. Chemoinformatics Strategies for Leishmaniasis Drug Discovery. Front Pharmacol 2018; 9:1278. [PMID: 30443215 PMCID: PMC6221941 DOI: 10.3389/fphar.2018.01278] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/18/2018] [Indexed: 12/15/2022] Open
Abstract
Leishmaniasis is a fatal neglected tropical disease (NTD) that is caused by more than 20 species of Leishmania parasites. The disease kills approximately 20,000 people each year and more than 1 billion are susceptible to infection. Although counting on a few compounds, the therapeutic arsenal faces some drawbacks such as drug resistance, toxicity issues, high treatment costs, and accessibility problems, which highlight the need for novel treatment options. Worldwide efforts have been made to that aim and, as well as in other therapeutic areas, chemoinformatics have contributed significantly to leishmaniasis drug discovery. Breakthrough advances in the comprehension of the parasites’ molecular biology have enabled the design of high-affinity ligands for a number of macromolecular targets. In addition, the use of chemoinformatics has allowed highly accurate predictions of biological activity and physicochemical and pharmacokinetics properties of novel antileishmanial compounds. This review puts into perspective the current context of leishmaniasis drug discovery and focuses on the use of chemoinformatics to develop better therapies for this life-threatening condition.
Collapse
Affiliation(s)
- Leonardo L G Ferreira
- Laboratory of Medicinal and Computational Chemistry, Center for Research and Innovation in Biodiversity and Drug Discovery, São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Adriano D Andricopulo
- Laboratory of Medicinal and Computational Chemistry, Center for Research and Innovation in Biodiversity and Drug Discovery, São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| |
Collapse
|