1
|
Saini I, Joshi J, Kaur S. Unleashing the role of potential adjuvants in leishmaniasis. Int J Pharm 2025; 669:125077. [PMID: 39675537 DOI: 10.1016/j.ijpharm.2024.125077] [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: 07/09/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Leishmaniasis is amongst one of the most neglected tropical disease, caused by an intracellular protozoan of genus Leishmania. Currently, the most promising strategy to combat leishmaniasis, relies on chemotherapy but the toxicity and increasing resistance of the standard drugs, presses the demand for new alternatives. Immunization is arguably the best strategy for cure because an individual once infected becomes immune to the disease. Yet, there is no efficient vaccine capable of providing enduring immunity against the parasite. Achieving the goal of developing highly efficacious and durable vaccine is limited due to lack of an appropriate adjuvant. Adjuvants are recognized as 'immune potentiators' which redirect or amplify the immune response. A number of adjuvants like alum, MPL-A, CpG ODN, GLA-SE, imiquimod, saponins etc. have been used in combination with various classes of Leishmania antigens. However, only few have reached clinical trials. Thus, the choice of an adjuvant is critically dependent on many factors such as the route of administration, the nature of antigen, formulation, the type of required immune response, their mode of action and the immunization schedule. This review provides an updated status on the types of adjuvants used in leishmaniasis so far and their mechanism of action if known.
Collapse
Affiliation(s)
- Isha Saini
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, India
| | - Jyoti Joshi
- Goswami Ganesh Dutta Sanatan Dharma College, Sector-32C, Chandigarh, India
| | - Sukhbir Kaur
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, India.
| |
Collapse
|
2
|
Zhang H, Yan R, Liu Y, Yu M, He Z, Xiao J, Li K, Liu G, Ning Q, Li Y. Progress in antileishmanial drugs: Mechanisms, challenges, and prospects. PLoS Negl Trop Dis 2025; 19:e0012735. [PMID: 39752369 PMCID: PMC11698350 DOI: 10.1371/journal.pntd.0012735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Leishmaniasis, a neglected tropical disease caused by Leishmania parasites, continues to pose global health challenges. Current treatments face issues like resistance, safety, efficacy, and cost. This review covers the discovery, mechanisms of action, clinical applications, and limitations of key antileishmanial agents: pentavalent antimonials, amphotericin B, miltefosine, paromomycin, and pentamidine. Despite toxicity and resistance (antimonials), hospitalization needs and side effects (amphotericin B), regional efficacy variability (miltefosine), inconsistent outcomes (paromomycin), and severe side effects (pentamidine), these drugs are vital. Novel strategies to overcome the deficiencies of current therapies are highlighted, including combination regimens, advanced drug delivery systems, and immunomodulatory approaches. Comprehensive and cooperative efforts are crucial to fully realize the potential of advancements in antileishmanial pharmacotherapy and to reduce the unacceptable worldwide burden imposed by this neglected disease.
Collapse
Affiliation(s)
- Haoran Zhang
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Ruixi Yan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yahui Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Mengtao Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyi He
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Junfeng Xiao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Kaijie Li
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Gang Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qin Ning
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
3
|
Singh VK, Tiwari R, Rajneesh, Kumar A, Chauhan SB, Sudarshan M, Mehrotra S, Gautam V, Sundar S, Kumar R. Advancing Treatment for Leishmaniasis: From Overcoming Challenges to Embracing Therapeutic Innovations. ACS Infect Dis 2024. [PMID: 39737830 DOI: 10.1021/acsinfecdis.4c00693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2025]
Abstract
Protozoan parasite infections, particularly leishmaniasis, present significant public health challenges in tropical and subtropical regions, affecting socio-economic status and growth. Despite advancements in immunology, effective vaccines remain vague, leaving drug treatments as the primary intervention. However, existing medications face limitations, such as toxicity and the rise of drug-resistant parasites. This presents an urgent need to identify new therapeutic targets for leishmaniasis treatment. Understanding the complex life cycle of Leishmania and its survival in host macrophages can provide insights into potential targets for intervention. Current treatments, including antimonials, amphotericin B, and miltefosine, are constrained by side effects, costs, resistance, and reduced efficacy. Exploring novel therapeutic targets within the parasite's physiology, such as key metabolic enzymes or essential surface proteins, may lead to the development of more effective and less toxic drugs. Additionally, innovative strategies like drug repurposing, combination therapies, and nanotechnology-based delivery systems could enhance efficacy and combat resistance, thus improving anti-leishmanial therapies.
Collapse
Affiliation(s)
- Vishal Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences Banaras Hindu University, Varanasi-221005, U.P., India
| | - Rahul Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences Banaras Hindu University, Varanasi-221005, U.P., India
| | - Rajneesh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences Banaras Hindu University, Varanasi-221005, U.P., India
| | - Awnish Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences Banaras Hindu University, Varanasi-221005, U.P., India
| | - Shashi Bhushan Chauhan
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences Banaras Hindu University, Varanasi-221005, U.P., India
| | - Medhavi Sudarshan
- Department of Zoology, Jagat Narayan College, Patliputra University, Patna-801105, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab-143005, India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences Banaras Hindu University, Varanasi-221005, U.P., India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221005, U.P. India
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences Banaras Hindu University, Varanasi-221005, U.P., India
| |
Collapse
|
4
|
Ayala A, Llanes A, Lleonart R, Restrepo CM. Advances in Leishmania Vaccines: Current Development and Future Prospects. Pathogens 2024; 13:812. [PMID: 39339003 PMCID: PMC11435054 DOI: 10.3390/pathogens13090812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Leishmaniasis is a neglected tropical disease caused by parasites of the genus Leishmania. As approved human vaccines are not available, treatment and prevention rely heavily on toxic chemotherapeutic agents, which face increasing resistance problems. The development of effective vaccines against human leishmaniasis is of utmost importance for the control of the disease. Strategies that have been considered for this purpose range from whole-killed and attenuated parasites to recombinant proteins and DNA vaccines. The ideal vaccine must be safe and effective, ensuring lasting immunity through a robust IL-12-driven Th1 adaptive immune response. Despite some success and years of effort, human vaccine trials have encountered difficulties in conferring durable protection against Leishmania, a problem that may be attributed to the parasite's antigenic diversity and the intricate nature of the host's immune response. The aim of this review is to provide a thorough overview of recent advances in Leishmania vaccine development, ranging from initial trials to recent achievements, such as the ChAd63-KH DNA vaccine, which underscores the potential for effective control of leishmaniasis through continued research in this field.
Collapse
Affiliation(s)
- Andreina Ayala
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama City 0843-01103, Panama
| | - Alejandro Llanes
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama City 0843-01103, Panama
- Sistema Nacional de Investigación (SNI), Panama City 0801, Panama
| | - Ricardo Lleonart
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama City 0843-01103, Panama
- Sistema Nacional de Investigación (SNI), Panama City 0801, Panama
| | - Carlos M Restrepo
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama City 0843-01103, Panama
- Sistema Nacional de Investigación (SNI), Panama City 0801, Panama
| |
Collapse
|
5
|
Andrés-Rodríguez J, González-Montero MC, García-Fernández N, Calvo-Álvarez E, Pérez-Pertejo MY, Reguera-Torres RM, Balaña-Fouce R, García-Estrada C. Free Radical Production Induced by Nitroimidazole Compounds Lead to Cell Death in Leishmania infantum Amastigotes. Molecules 2024; 29:4041. [PMID: 39274889 PMCID: PMC11396368 DOI: 10.3390/molecules29174041] [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: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Leishmania infantum is the vector-borne trypanosomatid parasite causing visceral leishmaniasis in the Mediterranean basin. This neglected tropical disease is treated with a limited number of obsolete drugs that are not exempt from adverse effects and whose overuse has promoted the emergence of resistant pathogens. In the search for novel antitrypanosomatid molecules that help overcome these drawbacks, drug repurposing has emerged as a good strategy. Nitroaromatic compounds have been found in drug discovery campaigns as promising antileishmanial molecules. Fexinidazole (recently introduced for the treatment of stages 1 and 2 of African trypanosomiasis), and pretomanid, which share the nitroimidazole nitroaromatic structure, have provided antileishmanial activity in different studies. In this work, we have tested the in vitro efficacy of these two nitroimidazoles to validate our 384-well high-throughput screening (HTS) platform consisting of L. infantum parasites emitting the near-infrared fluorescent protein (iRFP) as a biomarker of cell viability. These molecules showed good efficacy in both axenic and intramacrophage amastigotes and were poorly cytotoxic in RAW 264.7 and HepG2 cultures. Fexinidazole and pretomanid induced the production of ROS in axenic amastigotes but were not able to inhibit trypanothione reductase (TryR), thus suggesting that these compounds may target thiol metabolism through a different mechanism of action.
Collapse
Affiliation(s)
- Julia Andrés-Rodríguez
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - María-Cristina González-Montero
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Nerea García-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Estefanía Calvo-Álvarez
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - María-Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rosa-María Reguera-Torres
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
6
|
Registre C, Silva LM, Registre F, Soares RDDOA, Rubio KTS, Carneiro SP, Dos Santos ODH. Targeting Leishmania Promastigotes and Amastigotes Forms through Amino Acids and Peptides: A Promising Therapeutic Strategy. ACS Infect Dis 2024; 10:2467-2484. [PMID: 38950147 DOI: 10.1021/acsinfecdis.4c00089] [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: 07/03/2024]
Abstract
Millions of people worldwide are affected by leishmaniasis, caused by the Leishmania parasite. Effective treatment is challenging due to the biological complexity of the parasite, drug toxicity, and increasing resistance to conventional drugs. To combat this disease, the development of specific strategies to target and selectively eliminate the parasite is crucial. This Review highlights the importance of amino acids in the developmental stages of Leishmania as a factor determining whether the infection progresses or is suppressed. It also explores the use of peptides as alternatives in parasite control and the development of novel targeted treatments. While these strategies show promise for more effective and targeted treatment, further studies to address the remaining challenges are imperative.
Collapse
Affiliation(s)
- Charmante Registre
- Phytotechnology Laboratory, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Minas Gerais 35400000, Brazil
| | - Luciana Miranda Silva
- Phytotechnology Laboratory, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Minas Gerais 35400000, Brazil
| | - Farah Registre
- School of Medicine, Goiás Federal University, Goiânia, Goiás 74605-050, Brazil
| | - Rodrigo Dian de Oliveira Aguiar Soares
- Immunopathology Laboratory, Center for Research in Biological Sciences/NUPEB, Federal University of Ouro Preto, Ouro Preto, Minas Gerais 35400000, Brazil
| | - Karina Taciana Santos Rubio
- Toxicology Laboratory, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Minas Gerais 35400000, Brazil
| | - Simone Pinto Carneiro
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| | | |
Collapse
|
7
|
Singh A, Beg MA, Jamal S, Khan A, Rahman A, Selvapandiyan A, Shafi S, Hoda N. Robust leishmanicidal upshot of some new diphenyl triazine-based molecules. RSC Adv 2024; 14:22587-22597. [PMID: 39021460 PMCID: PMC11253633 DOI: 10.1039/d4ra01904k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Amongst the neglected tropical diseases, leishmaniasis alone causes 30 000 deaths annually due to the protozoan parasite genus Leishmania. Existing therapies have serious drawbacks in safety, drug resistance, field-adapted application and cost. Therefore, new safer and shorter treatments are an urgent need of the time. Herein, we report the synthesis of fifteen novel diphenyl triazine and diphenyl triazine pyrimidine derivatives and their antileishmanial properties against Leishmania donovani, that causes fatal visceral leishmaniasis. Most of the synthesized analogues exhibited more than 90% inhibition against the promastigote stage of the parasite. Moreover, compounds T4 and T7 showed potent activity against extracellular promastigote (IC50 = 1.074 μM and IC50 = 1.158 μM) as compared to miltefosine (IC50 = 1.477 μM) and is nontoxic towards the host THP-1 macrophage cell line. Interestingly, compound T4 exhibited significant activity against amastigotes (7.186 μM) and induced the macrophages to prevent the survival of the parasite. Our results indicate that T4 represents a new structural lead for this serious and neglected disease.
Collapse
Affiliation(s)
- Anju Singh
- Department of Chemistry, Drug Design and Synthesis Lab., Jamia Millia Islamia Jamia Nagar New Delhi 110025 India +0091-11-26985507 +0091-9910200655
| | - Mirza Adil Beg
- Department of Molecular Medicine, Jamia Hamdard New Delhi 110062 India
| | - Samra Jamal
- Department of Biotechnology, Jamia Hamdard New Delhi 110062 India
| | - Arif Khan
- Department of Chemistry, SCLS, Jamia Hamdard New Delhi 110062 India
| | - Abdur Rahman
- Department of Chemistry, Drug Design and Synthesis Lab., Jamia Millia Islamia Jamia Nagar New Delhi 110025 India +0091-11-26985507 +0091-9910200655
| | | | - Syed Shafi
- Department of Chemistry, SCLS, Jamia Hamdard New Delhi 110062 India
| | - Nasimul Hoda
- Department of Chemistry, Drug Design and Synthesis Lab., Jamia Millia Islamia Jamia Nagar New Delhi 110025 India +0091-11-26985507 +0091-9910200655
| |
Collapse
|
8
|
de Souza VMR, Maciel NB, Machado YAA, de Sousa JMS, Rodrigues RRL, dos Santos ALS, Gonçalves da Silva MG, Martins da Silva IG, Barros-Cordeiro KB, Báo SN, Tavares JF, Rodrigues KADF. Anti- Leishmania amazonensis Activity of Morolic Acid, a Pentacyclic Triterpene with Effects on Innate Immune Response during Macrophage Infection. Microorganisms 2024; 12:1392. [PMID: 39065160 PMCID: PMC11279160 DOI: 10.3390/microorganisms12071392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Leishmaniasis is a group of infectious diseases transmitted to humans during vector bites and caused by protozoans of the genus Leishmania. Conventional therapies face challenges due to their serious side effects, prompting research into new anti-leishmania agents. In this context, we investigated the effectiveness of morolic acid, a pentacyclic triterpene, on L. amazonensis promastigotes and amastigotes. The present study employed the MTT assay, cytokine analysis using optEIATM kits, an H2DCFDA test, and nitric oxide dosage involving nitrite production and Griess reagent. Morolic acid inhibited promastigote and axenic amastigote growth forms at IC50 values of 1.13 µM and 2.74 µM, respectively. For cytotoxicity to macrophages and VERO cells, morolic acid obtained respective CC50 values of 68.61 µM and 82.94 µM. The compound causes damage to the parasite membrane, leading to cellular leakage. In the infection assay, there was a decrease in parasite load, resulting in a CI50 of 2.56 µM. This effect was associated with immunomodulatory activity, altering macrophage structural and cellular parasite elimination mechanisms. Morolic acid proved to be an effective and selective natural compound, making it a strong candidate for future in vivo studies in cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Vanessa Maria Rodrigues de Souza
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University Delta of Parnaiba, Parnaíba 64202-020, PI, Brazil; (V.M.R.d.S.); (N.B.M.); (Y.A.A.M.); (J.M.S.d.S.); (R.R.L.R.); (A.L.S.d.S.); (M.G.G.d.S.)
| | - Nicolle Barreira Maciel
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University Delta of Parnaiba, Parnaíba 64202-020, PI, Brazil; (V.M.R.d.S.); (N.B.M.); (Y.A.A.M.); (J.M.S.d.S.); (R.R.L.R.); (A.L.S.d.S.); (M.G.G.d.S.)
| | - Yasmim Alves Aires Machado
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University Delta of Parnaiba, Parnaíba 64202-020, PI, Brazil; (V.M.R.d.S.); (N.B.M.); (Y.A.A.M.); (J.M.S.d.S.); (R.R.L.R.); (A.L.S.d.S.); (M.G.G.d.S.)
| | - Julyanne Maria Saraiva de Sousa
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University Delta of Parnaiba, Parnaíba 64202-020, PI, Brazil; (V.M.R.d.S.); (N.B.M.); (Y.A.A.M.); (J.M.S.d.S.); (R.R.L.R.); (A.L.S.d.S.); (M.G.G.d.S.)
| | - Raiza Raianne Luz Rodrigues
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University Delta of Parnaiba, Parnaíba 64202-020, PI, Brazil; (V.M.R.d.S.); (N.B.M.); (Y.A.A.M.); (J.M.S.d.S.); (R.R.L.R.); (A.L.S.d.S.); (M.G.G.d.S.)
| | - Airton Lucas Sousa dos Santos
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University Delta of Parnaiba, Parnaíba 64202-020, PI, Brazil; (V.M.R.d.S.); (N.B.M.); (Y.A.A.M.); (J.M.S.d.S.); (R.R.L.R.); (A.L.S.d.S.); (M.G.G.d.S.)
| | - Maria Gabrielly Gonçalves da Silva
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University Delta of Parnaiba, Parnaíba 64202-020, PI, Brazil; (V.M.R.d.S.); (N.B.M.); (Y.A.A.M.); (J.M.S.d.S.); (R.R.L.R.); (A.L.S.d.S.); (M.G.G.d.S.)
| | - Ingrid Gracielle Martins da Silva
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (I.G.M.d.S.); (K.B.B.-C.); (S.N.B.)
| | - Karine Brenda Barros-Cordeiro
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (I.G.M.d.S.); (K.B.B.-C.); (S.N.B.)
| | - Sônia Nair Báo
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, DF, Brazil; (I.G.M.d.S.); (K.B.B.-C.); (S.N.B.)
| | - Josean Fechine Tavares
- Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil;
| | - Klinger Antonio da Franca Rodrigues
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University Delta of Parnaiba, Parnaíba 64202-020, PI, Brazil; (V.M.R.d.S.); (N.B.M.); (Y.A.A.M.); (J.M.S.d.S.); (R.R.L.R.); (A.L.S.d.S.); (M.G.G.d.S.)
| |
Collapse
|
9
|
Tekle E, Dese K, Girma S, Adissu W, Krishnamoorthy J, Kwa T. DeepLeish: a deep learning based support system for the detection of Leishmaniasis parasite from Giemsa-stained microscope images. BMC Med Imaging 2024; 24:152. [PMID: 38890604 PMCID: PMC11186139 DOI: 10.1186/s12880-024-01333-1] [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: 12/17/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Leishmaniasis is a vector-born neglected parasitic disease belonging to the genus Leishmania. Out of the 30 Leishmania species, 21 species cause human infection that affect the skin and the internal organs. Around, 700,000 to 1,000,000 of the newly infected cases and 26,000 to 65,000 deaths are reported worldwide annually. The disease exhibits three clinical presentations, namely, the cutaneous, muco-cutaneous and visceral Leishmaniasis which affects the skin, mucosal membrane and the internal organs, respectively. The relapsing behavior of the disease limits its diagnosis and treatment efficiency. The common diagnostic approaches follow subjective, error-prone, repetitive processes. Despite, an ever pressing need for an accurate detection of Leishmaniasis, the research conducted so far is scarce. In this regard, the main aim of the current research is to develop an artificial intelligence based detection tool for the Leishmaniasis from the Geimsa-stained microscopic images using deep learning method. METHODS Stained microscopic images were acquired locally and labeled by experts. The images were augmented using different methods to prevent overfitting and improve the generalizability of the system. Fine-tuned Faster RCNN, SSD, and YOLOV5 models were used for object detection. Mean average precision (MAP), precision, and Recall were calculated to evaluate and compare the performance of the models. RESULTS The fine-tuned YOLOV5 outperformed the other models such as Faster RCNN and SSD, with the MAP scores, of 73%, 54% and 57%, respectively. CONCLUSION The currently developed YOLOV5 model can be tested in the clinics to assist the laboratorists in diagnosing Leishmaniasis from the microscopic images. Particularly, in low-resourced healthcare facilities, with fewer qualified medical professionals or hematologists, our AI support system can assist in reducing the diagnosing time, workload, and misdiagnosis. Furthermore, the dataset collected by us will be shared with other researchers who seek to improve upon the detection system of the parasite. The current model detects the parasites even in the presence of the monocyte cells, but sometimes, the accuracy decreases due to the differences in the sizes of the parasite cells alongside the blood cells. The incorporation of cascaded networks in future and the quantification of the parasite load, shall overcome the limitations of the currently developed system.
Collapse
Affiliation(s)
- Eden Tekle
- School of Biomedical Engineering, Jimma University, Jimma, Ethiopia
| | - Kokeb Dese
- School of Biomedical Engineering, Jimma University, Jimma, Ethiopia.
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV, 26505, USA.
| | - Selfu Girma
- Pathology Unit, Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Wondimagegn Adissu
- School of Medical Laboratory Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
- Clinical Trial Unit, Jimma University, Jimma, Ethiopia
| | | | - Timothy Kwa
- School of Biomedical Engineering, Jimma University, Jimma, Ethiopia.
- Medtronic MiniMed, 18000 Devonshire St. Northridge, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Sheikh SY, Hassan F, Shukla D, Bala S, Faruqui T, Akhter Y, Khan AR, Nasibullah M. A review on potential therapeutic targets for the treatment of leishmaniasis. Parasitol Int 2024; 100:102863. [PMID: 38272301 DOI: 10.1016/j.parint.2024.102863] [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: 05/30/2023] [Revised: 12/22/2023] [Accepted: 01/21/2024] [Indexed: 01/27/2024]
Abstract
Leishmania, a protozoan parasite, is responsible for the occurrence of leishmaniasis, a disease that is prevalent in tropical regions. Visceral Leishmaniasis (VL), also known as kala-azar in Asian countries, is one of the most significant forms of VL, along with Cutaneous Leishmaniasis (CL) and Mucocutaneous Leishmaniasis (ML). Management of this condition typically entails the use of chemotherapy as the sole therapeutic option. The current treatments for leishmaniasis present several drawbacks, including a multitude of side effects, prolonged treatment duration, disparate efficacy across different regions, and the emergence of resistance. To address this urgent need, it is imperative to identify alternative treatments that are both safer and more effective. The identification of appropriate pharmacological targets in conjunction with biological pathways constitutes the initial stage of drug discovery. In this review, we have addressed the key metabolic pathways that represent potential pharmacological targets as well as prominent treatment options for leishmaniasis.
Collapse
Affiliation(s)
- Sabahat Yasmeen Sheikh
- Department of Chemistry, Integral University, Dasauli, Kursi road, Lucknow 226026, India
| | - Firoj Hassan
- Department of Chemistry, Integral University, Dasauli, Kursi road, Lucknow 226026, India
| | - Deepanjali Shukla
- Department of Chemistry, Integral University, Dasauli, Kursi road, Lucknow 226026, India
| | - Shashi Bala
- Department of Chemistry, Lucknow University, Lucknow 226026, India
| | - Tabrez Faruqui
- Department of Biosciences, Integral University, Lucknow 226026, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Abdul Rahman Khan
- Department of Chemistry, Integral University, Dasauli, Kursi road, Lucknow 226026, India
| | - Malik Nasibullah
- Department of Chemistry, Integral University, Dasauli, Kursi road, Lucknow 226026, India.
| |
Collapse
|
11
|
González-Montero MC, Andrés-Rodríguez J, García-Fernández N, Pérez-Pertejo Y, Reguera RM, Balaña-Fouce R, García-Estrada C. Targeting Trypanothione Metabolism in Trypanosomatids. Molecules 2024; 29:2214. [PMID: 38792079 PMCID: PMC11124245 DOI: 10.3390/molecules29102214] [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: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Infectious diseases caused by trypanosomatids, including African trypanosomiasis (sleeping sickness), Chagas disease, and different forms of leishmaniasis, are Neglected Tropical Diseases affecting millions of people worldwide, mainly in vulnerable territories of tropical and subtropical areas. In general, current treatments against these diseases are old-fashioned, showing adverse effects and loss of efficacy due to misuse or overuse, thus leading to the emergence of resistance. For these reasons, searching for new antitrypanosomatid drugs has become an urgent necessity, and different metabolic pathways have been studied as potential drug targets against these parasites. Considering that trypanosomatids possess a unique redox pathway based on the trypanothione molecule absent in the mammalian host, the key enzymes involved in trypanothione metabolism, trypanothione reductase and trypanothione synthetase, have been studied in detail as druggable targets. In this review, we summarize some of the recent findings on the molecules inhibiting these two essential enzymes for Trypanosoma and Leishmania viability.
Collapse
Affiliation(s)
- María-Cristina González-Montero
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
| | - Julia Andrés-Rodríguez
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
| | - Nerea García-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
| | - Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
12
|
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
|
13
|
Pivotto AP, de Souza Lima LB, Michelon A, Ferreira CZP, Gandra RF, Ayala TS, Menolli RA. Topical application of ozonated sunflower oil accelerates the healing of lesions of cutaneous leishmaniasis in mice under meglumine antimoniate treatment. Med Microbiol Immunol 2024; 213:4. [PMID: 38532203 DOI: 10.1007/s00430-024-00788-x] [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: 04/10/2023] [Accepted: 01/24/2024] [Indexed: 03/28/2024]
Abstract
Besides being scarce, the drugs available for treating cutaneous leishmaniasis have many adverse effects. Ozone is an option to enhance the standard treatment due to the wound-healing activity reported in the literature. In this study, we evaluated the efficiency of ozonated sunflower oil as an adjuvant in treating cutaneous lesions caused by Leishmania amazonensis. BALB/c mice were infected with L. amazonensis, and after the lesions appeared, they were treated in four different schedules using the drug treatment with meglumine antimoniate (Glucantime®), with or without ozonated oil. After thirty days of treatment, the lesions' thickness and their parasitic burden, blood leukocytes, production of NO and cytokines from peritoneal macrophages and lymph node cells were analyzed. The group treated with ozonated oil plus meglumine antimoniate showed the best performance, improving the lesion significantly. The parasitic burden showed that ozonated oil enhanced the leishmanicidal activity of the treatment, eliminating the parasites in the lesion. Besides, a decrease in the TNF levels from peritoneal macrophages and blood leukocytes demonstrated an immunomodulatory action of ozone in the ozonated oil-treated animals compared to the untreated group. Thus, ozonated sunflower oil therapy has been shown as an adjuvant in treating Leishmania lesions since this treatment enhanced the leishmanicidal and wound healing effects of meglumine antimoniate.
Collapse
Affiliation(s)
- Ana Paula Pivotto
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, PR, Zip Code 85819-110, Brazil
| | - Lucas Bonatto de Souza Lima
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, PR, Zip Code 85819-110, Brazil
| | - Alexandra Michelon
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, PR, Zip Code 85819-110, Brazil
| | - Camilla Zottesso Pellon Ferreira
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, PR, Zip Code 85819-110, Brazil
| | - Rinaldo Ferreira Gandra
- Laboratory of Microbiology, Western Parana University Hospital, Western Parana State University, Cascavel, PR, Brazil
| | - Thaís Soprani Ayala
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, PR, Zip Code 85819-110, Brazil
| | - Rafael Andrade Menolli
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, PR, Zip Code 85819-110, Brazil.
| |
Collapse
|
14
|
Sifontes-Rodríguez S, Mollineda-Diogo N, Monzote-Fidalgo L, Escalona-Montaño AR, Escario García-Trevijano JA, Aguirre-García MM, Meneses-Marcel A. In Vitro and In Vivo Antileishmanial Activity of Thioridazine. Acta Parasitol 2024; 69:324-331. [PMID: 38070122 PMCID: PMC11001698 DOI: 10.1007/s11686-023-00746-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 11/06/2023] [Indexed: 04/11/2024]
Abstract
INTRODUCTION Leishmaniasis is a neglected disease with high prevalence and incidence in tropical and subtropical areas. Existing drugs are limited due to cost, toxicity, declining efficacy and unavailability in endemic places. Drug repurposing has established as an efficient way for the discovery of drugs for a variety of diseases. PURPOSE The objective of the present work was testing the antileishmanial activity of thioridazine, an antipsychotic agent with demonstrated effect against other intracellular pathogens. METHODS The cytotoxicity for mouse peritoneal macrophages as well as the activity against Leishmania amazonensis, Leishmania mexicana and Leishmania major promastigotes and intracellular amastigotes, as well as in a mouse model of cutaneous leishmaniasis, were assessed. RESULTS Thioridazine inhibited the in vitro proliferation of promastigotes (50% inhibitory concentration-IC50-values in the range of 0.73 µM to 3.8 µM against L. amazonensis, L. mexicana and L. major) and intracellular amastigotes (IC50 values of 1.27 µM to 4.4 µM for the same species). In contrast, in mouse peritoneal macrophages, the 50% cytotoxic concentration was 24.0 ± 1.89 µM. Thioridazine inhibited the growth of cutaneous lesions and reduced the number of parasites in the infected tissue of mice. The dose of thioridazine that inhibited lesion development by 50% compared to controls was 23.3 ± 3.1 mg/kg and in terms of parasite load, it was 11.1 ± 0.97 mg/kg. CONCLUSIONS Thioridazine was effective against the promastigote and intracellular amastigote stages of three Leishmania species and in a mouse model of cutaneous leishmaniasis, supporting the potential repurposing of this drug as an antileishmanial agent.
Collapse
Affiliation(s)
- Sergio Sifontes-Rodríguez
- División de Investigación, Facultad de Medicina, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Niurka Mollineda-Diogo
- Centro de Bioactivos Químicos, Universidad Central "Martha Abreu" de Las Villas, Santa Clara, Villa Clara, Cuba
| | | | - Alma Reyna Escalona-Montaño
- División de Investigación, Facultad de Medicina, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | - María Magdalena Aguirre-García
- División de Investigación, Facultad de Medicina, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.
| | - Alfredo Meneses-Marcel
- Centro de Bioactivos Químicos, Universidad Central "Martha Abreu" de Las Villas, Santa Clara, Villa Clara, Cuba
| |
Collapse
|
15
|
Melcón-Fernández E, Galli G, Balaña-Fouce R, García-Fernández N, Martínez-Valladares M, Reguera RM, García-Estrada C, Pérez-Pertejo Y. In Vitro and Ex Vivo Synergistic Effect of Pyrvinium Pamoate Combined with Miltefosine and Paromomycin against Leishmania. Trop Med Infect Dis 2024; 9:30. [PMID: 38393119 PMCID: PMC10891607 DOI: 10.3390/tropicalmed9020030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
One of the major drawbacks of current treatments for neglected tropical diseases is the low safety of the drugs used and the emergence of resistance. Leishmaniasis is a group of neglected diseases caused by protozoa of the trypanosomatidae family that lacks preventive vaccines and whose pharmacological treatments are scarce and unsafe. Combination therapy is a strategy that could solve the above-mentioned problems, due to the participation of several mechanisms of action and the reduction in the amount of drug necessary to obtain the therapeutic effect. In addition, this approach also increases the odds of finding an effective drug following the repurposing strategy. From the previous screening of two collections of repositioning drugs, we found that pyrvinium pamoate had a potent leishmanicidal effect. For this reason, we decided to combine it separately with two clinically used leishmanicidal drugs, miltefosine and paromomycin. These combinations were tested in axenic amastigotes of Leishmania infantum obtained from bone marrow cells and in intramacrophagic amastigotes obtained from primary cultures of splenic cells, both cell types coming from experimentally infected mice. Some of the combinations showed synergistic behavior, especially in the case of the combination of pyrvinium pamoate with paromomycin, and exhibited low cytotoxicity and good tolerability on intestinal murine organoids, which reveal the potential of these combinations for the treatment of leishmaniasis.
Collapse
Affiliation(s)
- Estela Melcón-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, 24071 Leon, Spain; (E.M.-F.); (G.G.); (R.B.-F.); (N.G.-F.); (R.M.R.); (C.G.-E.)
| | - Giulio Galli
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, 24071 Leon, Spain; (E.M.-F.); (G.G.); (R.B.-F.); (N.G.-F.); (R.M.R.); (C.G.-E.)
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, 24071 Leon, Spain; (E.M.-F.); (G.G.); (R.B.-F.); (N.G.-F.); (R.M.R.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 Leon, Spain
| | - Nerea García-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, 24071 Leon, Spain; (E.M.-F.); (G.G.); (R.B.-F.); (N.G.-F.); (R.M.R.); (C.G.-E.)
| | | | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, 24071 Leon, Spain; (E.M.-F.); (G.G.); (R.B.-F.); (N.G.-F.); (R.M.R.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 Leon, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, 24071 Leon, Spain; (E.M.-F.); (G.G.); (R.B.-F.); (N.G.-F.); (R.M.R.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 Leon, Spain
| | - Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, 24071 Leon, Spain; (E.M.-F.); (G.G.); (R.B.-F.); (N.G.-F.); (R.M.R.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 Leon, Spain
| |
Collapse
|
16
|
dos Santos JV, Medina JM, Dias Teixeira KL, Agostinho DMJ, Chorev M, Diotallevi A, Galluzzi L, Aktas BH, Gazos Lopes U. Activity of the Di-Substituted Urea-Derived Compound I-17 in Leishmania In Vitro Infections. Pathogens 2024; 13:104. [PMID: 38392842 PMCID: PMC10893125 DOI: 10.3390/pathogens13020104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/25/2024] Open
Abstract
Protein synthesis has been a very rich target for developing drugs to control prokaryotic and eukaryotic pathogens. Despite the development of new drug formulations, treating human cutaneous and visceral Leishmaniasis still needs significant improvements due to the considerable side effects and low adherence associated with the current treatment regimen. In this work, we show that the di-substituted urea-derived compounds I-17 and 3m are effective in inhibiting the promastigote growth of different Leishmania species and reducing the macrophage intracellular load of amastigotes of the Leishmania (L.) amazonensis and L. major species, in addition to exhibiting low macrophage cytotoxicity. We also show a potential immunomodulatory effect of I-17 and 3m in infected macrophages, which exhibited increased expression of inducible Nitric Oxide Synthase (NOS2) and production of Nitric Oxide (NO). Our data indicate that I-17, 3m, and their analogs may be helpful in developing new drugs for treating leishmaniasis.
Collapse
Affiliation(s)
- José Vitorino dos Santos
- Laboratory of Molecular Parasitology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.V.d.S.); (J.M.M.); (D.M.J.A.)
| | - Jorge Mansur Medina
- Laboratory of Molecular Parasitology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.V.d.S.); (J.M.M.); (D.M.J.A.)
| | | | - Daniel Marcos Julio Agostinho
- Laboratory of Molecular Parasitology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.V.d.S.); (J.M.M.); (D.M.J.A.)
| | - Michael Chorev
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Aurora Diotallevi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (A.D.)
| | - Luca Galluzzi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (A.D.)
| | - Bertal Huseyin Aktas
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Ulisses Gazos Lopes
- Laboratory of Molecular Parasitology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.V.d.S.); (J.M.M.); (D.M.J.A.)
| |
Collapse
|
17
|
Fermiano MH, das Neves AR, da Silva F, Barros MSA, Vieira CB, Stein AL, Frizon TEA, Braga AL, de Arruda CCP, Parisotto EB, Saba S, Rafique J, Riul TB. Selenium-Containing (Hetero)Aryl Hybrids as Potential Antileishmanial Drug Candidates: In Vitro Screening against L. amazonensis. Biomedicines 2024; 12:213. [PMID: 38255318 PMCID: PMC10812941 DOI: 10.3390/biomedicines12010213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Leishmaniasis remains a significant global health concern, with current treatments relying on outdated drugs associated with high toxicity, lengthy administration, elevated costs, and drug resistance. Consequently, the urgent need for safer and more effective therapeutic options in leishmaniasis treatment persists. Previous research has highlighted selenium compounds as promising candidates for innovative leishmaniasis therapy. In light of this, a library of 10 selenium-containing diverse compounds was designed and evaluated in this study. These compounds included selenium-substituted indole, coumarin, chromone, oxadiazole, imidazo[1,2-a]pyridine, Imidazo[2,1-b]thiazole, and oxazole, among others. These compounds were screened against Leishmania amazonensis promastigotes and intracellular amastigotes, and their cytotoxicity was assessed in peritoneal macrophages, NIH/3T3, and J774A.1 cells. Among the tested compounds, MRK-106 and MRK-108 displayed the highest potency against L. amazonensis promastigotes with reduced cytotoxicity. Notably, MRK-106 and MRK-108 exhibited IC50 values of 3.97 µM and 4.23 µM, respectively, and most of the tested compounds showed low cytotoxicity in host cells (CC50 > 200 µM). Also, compounds MRK-107 and MRK-113 showed activity against intracellular amastigotes (IC50 18.31 and 15.93 µM and SI 12.55 and 10.92, respectively). In conclusion, the identified selenium-containing compounds hold potential structures as antileishmanial drug candidates to be further explored in subsequent studies. These findings represent a significant step toward the development of safer and more effective therapies for leishmaniasis, addressing the pressing need for novel and improved treatments.
Collapse
Affiliation(s)
- Maria Helena Fermiano
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil (A.R.d.N.)
| | - Amarith Rodrigues das Neves
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil (A.R.d.N.)
| | - Fernanda da Silva
- Instituto de Biociências (INBIO), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil
| | | | - Camila Barbosa Vieira
- LABSO, Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Goiânia 74690-900, GO, Brazil (S.S.)
| | - André L. Stein
- Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Cuiabá 78060-900, MT, Brazil
| | - Tiago Elias Allievi Frizon
- Departamento de Energia e Sustentabilidade, Universidade Federal de Santa Catarina (UFSC), Campus Araranguá, Araranguá 88905-120, SC, Brazil
| | - Antonio Luiz Braga
- Departamento de Química, Universidade Federal de Santa Catarina (UFSC), Florianópolis 88040-970, SC, Brazil
| | - Carla Cardozo Pinto de Arruda
- Instituto de Biociências (INBIO), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil
| | - Eduardo Benedetti Parisotto
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil (A.R.d.N.)
| | - Sumbal Saba
- LABSO, Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Goiânia 74690-900, GO, Brazil (S.S.)
| | - Jamal Rafique
- Instituto de Química (INQUI), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79074-460, MS, Brazil;
- LABSO, Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Goiânia 74690-900, GO, Brazil (S.S.)
| | - Thalita Bachelli Riul
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil (A.R.d.N.)
| |
Collapse
|
18
|
Pérez-Pertejo Y, García-Estrada C, Martínez-Valladares M, Murugesan S, Reguera RM, Balaña-Fouce R. Polyamine Metabolism for Drug Intervention in Trypanosomatids. Pathogens 2024; 13:79. [PMID: 38251386 PMCID: PMC10820115 DOI: 10.3390/pathogens13010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Neglected tropical diseases transmitted by trypanosomatids include three major human scourges that globally affect the world's poorest people: African trypanosomiasis or sleeping sickness, American trypanosomiasis or Chagas disease and different types of leishmaniasis. Different metabolic pathways have been targeted to find antitrypanosomatid drugs, including polyamine metabolism. Since their discovery, the naturally occurring polyamines, putrescine, spermidine and spermine, have been considered important metabolites involved in cell growth. With a complex metabolism involving biosynthesis, catabolism and interconversion, the synthesis of putrescine and spermidine was targeted by thousands of compounds in an effort to produce cell growth blockade in tumor and infectious processes with limited success. However, the discovery of eflornithine (DFMO) as a curative drug against sleeping sickness encouraged researchers to develop new molecules against these diseases. Polyamine synthesis inhibitors have also provided insight into the peculiarities of this pathway between the host and the parasite, and also among different trypanosomatid species, thus allowing the search for new specific chemical entities aimed to treat these diseases and leading to the investigation of target-based scaffolds. The main molecular targets include the enzymes involved in polyamine biosynthesis (ornithine decarboxylase, S-adenosylmethionine decarboxylase and spermidine synthase), enzymes participating in their uptake from the environment, and the enzymes involved in the redox balance of the parasite. In this review, we summarize the research behind polyamine-based treatments, the current trends, and the main challenges in this field.
Collapse
Affiliation(s)
- Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Campus de Vegazana s/n, Universidad de León, 24071 León, Spain; (Y.P.-P.); (C.G.-E.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Campus de Vegazana s/n, Universidad de León, 24071 León, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Campus de Vegazana s/n, Universidad de León, 24071 León, Spain; (Y.P.-P.); (C.G.-E.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Campus de Vegazana s/n, Universidad de León, 24071 León, Spain
| | | | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani 333031, India;
| | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Campus de Vegazana s/n, Universidad de León, 24071 León, Spain; (Y.P.-P.); (C.G.-E.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Campus de Vegazana s/n, Universidad de León, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Campus de Vegazana s/n, Universidad de León, 24071 León, Spain; (Y.P.-P.); (C.G.-E.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Campus de Vegazana s/n, Universidad de León, 24071 León, Spain
| |
Collapse
|
19
|
de Carvalho Moreira LMC, de Sousa Silva ABA, de Araújo Medeiros K, Oshiro Júnior JA, da Silva DTC, de Lima Damasceno BPG. Effectiveness In Vivo and In Vitro of Polymeric Nanoparticles as a Drug Release System in the Treatment of Leishmaniasis. Curr Med Chem 2024; 31:286-307. [PMID: 36683370 DOI: 10.2174/0929867330666230120163543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/13/2022] [Accepted: 11/08/2022] [Indexed: 01/24/2023]
Abstract
Leishmaniasis is a neglected disease caused by the parasite of the genus Leishmania. Current treatment regimens are obsolete and cause several side effects, promoting poor patient compliance, in addition to the vast majority already having the potential for resistance. Therefore, polymeric nanoparticles emerge as one of the viable alternatives to overcome existing limitations, through passive or active vectorization. This review aims to summarize the latest studies of polymeric nanoparticles as an alternative treatment for leishmaniasis. In the first section, the main pharmacokinetic and pharmacodynamic challenges of current drugs are reported. The second section details how nanoparticles with and without functionalization are efficient in the treatment of leishmaniasis, discussing the characteristics of the polymer in the formulation. In this way, polymeric nanoparticles can improve the physicochemical properties of leishmanicidal drugs, improving solubility and stability, as well as improve the release of these drugs, directly or indirectly reaching monocytes/macrophages. 64.28% drugs were focused on the treatment of visceral leishmaniasis, and 28.57% on cutaneous leishmaniasis. The most chosen polymers in the literature are chitosan (35.71%) and PLGA (35.71%), the others represented 14.30% drugs, with all able to manage the drug release and increase the in vitro and/or in vivo efficacy of the original molecule. However, there are several barriers for these nanoformulations to cross laboratory research and is necessary more in-depth studies about the metabolites and degradation pathways of the polymers used in the formulations and plasma proteomics studies.
Collapse
Affiliation(s)
- Lívia Maria Coelho de Carvalho Moreira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| | | | - Kaline de Araújo Medeiros
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| | - João Augusto Oshiro Júnior
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| | - Dayanne Tomaz Casimiro da Silva
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| | - Bolívar Ponciano Goulart de Lima Damasceno
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
- Laboratório de Desenvolvimento e Caracterização de Produtos Farmacêuticos, Universidade Estadual da Paraíba, Campina Grande, PB, Brasil
| |
Collapse
|
20
|
Melcón-Fernandez E, Martín-Encinas E, Palacios F, Galli G, Reguera RM, Martínez-Valladares M, Balaña-Fouce R, Alonso C, Pérez-Pertejo Y. Antileishmanial Effect of 1,5- and 1,8-Substituted Fused Naphthyridines. Molecules 2023; 29:74. [PMID: 38202656 PMCID: PMC10780244 DOI: 10.3390/molecules29010074] [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: 11/04/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
In the absence of a vaccine, there is a need to find new drugs for the treatment of neglected tropical diseases, such as leishmaniasis, that can overcome the many drawbacks of those currently used. These disadvantages include cost, the need to maintain a cold chain, the route of administration, the associated adverse effects and the generation of resistance. In this work we have evaluated the antileishmanial effect of 1,5- and 1,8-substituted fused naphthyridines through in vitro and ex vivo assays, using genetically modified axenic and intramacrophagic Leishmania infantum amastigotes. The toxicity of these compounds has been tested in the mammalian host cell using murine splenic macrophages, as well as in murine intestinal organoids (miniguts) in order to assess their potential for oral administration. The 1,8- derivatives showed greater leishmanicidal activity and the presence of a nitrogen atom in the fused ring to the naphthyridine was important to increase the activity of both types of molecules. The aromatization of the pyridine ring also had marked differences in the activity of the compounds.
Collapse
Affiliation(s)
- Estela Melcón-Fernandez
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain (G.G.)
| | - Endika Martín-Encinas
- Departamento de Química Orgánica I, Facultad de Farmacia, Lascaray Research Center, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
| | - Francisco Palacios
- Departamento de Química Orgánica I, Facultad de Farmacia, Lascaray Research Center, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
| | - Gulio Galli
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain (G.G.)
| | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain (G.G.)
| | - María Martínez-Valladares
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain (G.G.)
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain (G.G.)
| | - Concepción Alonso
- Departamento de Química Orgánica I, Facultad de Farmacia, Lascaray Research Center, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
| | - Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain (G.G.)
| |
Collapse
|
21
|
Kaye PM, Matlashewski G, Mohan S, Le Rutte E, Mondal D, Khamesipour A, Malvolti S. Vaccine value profile for leishmaniasis. Vaccine 2023; 41 Suppl 2:S153-S175. [PMID: 37951693 DOI: 10.1016/j.vaccine.2023.01.057] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/22/2022] [Accepted: 01/24/2023] [Indexed: 11/14/2023]
Abstract
Leishmania infections are global, occurring in 98 countries and all World Health Organization (WHO) regions with 600 million to 1 billion people at risk of infection. Visceral leishmaniasis is associated with almost 20,000 reported deaths annually, with children under 5 years of age being at the greatest risk of mortality. Amongst WHO-recognised Neglected Tropical Diseases (NTDs), leishmaniasis is one of the most important in terms of mortality and morbidity. With an increasing global burden of disease and a growing threat from climate change, urbanisation and drug resistance, there remains an imperative to develop leishmaniasis vaccines. New tools to understand correlates of protection and to assess vaccine efficacy are being developed to ease the transition into larger scale efficacy trials or provide alternate routes to licensure. Early indications suggest a diverse portfolio of manufacturers exists in endemic countries with an appetite to develop leishmaniasis vaccines. This Vaccine Value Profile (VVP) provides a high-level, comprehensive assessment of the currently available data to inform the potential public health, economic, and societal value of leishmaniasis vaccines. The leishmaniasis VVP was developed by a working group of subject matter experts from academia, public health groups, policy organizations, and non-profit organizations. All contributors have extensive expertise on various elements of the leishmaniasis VVP and have collectively described the state of knowledge and identified the current gaps. The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK.
| | - Greg Matlashewski
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada.
| | - Sakshi Mohan
- Center for Health Economics (CHE), University of York, York, UK.
| | - Epke Le Rutte
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | - Dinesh Mondal
- Laboratory Sciences and Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh.
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
22
|
Kumar R, Singh R, das Chagas Almeida A, da Trindade Granato J, de Oliveira Lemos AS, Kumar K, Patil MT, da Silva AD, Rode AB, Coimbra ES, Salunke DB. Imidazo[1,2- a]pyrimidine as a New Antileishmanial Pharmacophore against Leishmania amazonensis Promastigotes and Amastigotes. ACS OMEGA 2023; 8:40613-40621. [PMID: 37929127 PMCID: PMC10621021 DOI: 10.1021/acsomega.3c05441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023]
Abstract
Leishmania poses a substantial threat to the human population all over the globe because of its visceral and cutaneous spread engendered by all 20 species. Unfortunately, the available drugs against leishmania are already hobbled with toxicity, prolonged treatment, and increasing instances of acquirement of resistance. Under these grave circumstances, the development of new drugs has become imperative to keep these harmful microbes at bay. To this end, a Groebke-Blackburn-Bienaymé multicomponent reaction-based library of different imidazo-fused heterocycles has been synthesized and screened against Leishmania amazonensis promastigotes and amastigotes. Among the library compounds, the imidazo-pyrimidine 24 has been found to be the most effective (inhibitory concentration of 50% (IC50) < 10 μM), with selective antileishmanial activity on amastigote forms, a stage of the parasite related to human disease. The compound 24 has exhibited an IC50 value of 6.63 μM, being ∼two times more active than miltefosine, a reference drug. Furthermore, this compound is >10 times more destructive to the intracellular parasites than host cells. The observed in vitro antileishmanial activity along with suitable in silico physicochemical and absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties of compound 24 reinforce the imidazo-pyrimidine scaffold as a new antileishmanial pharmacophore and encourage further murine experimental leishmaniasis studies.
Collapse
Affiliation(s)
- Ravinder Kumar
- Department
of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160 014, India
| | - Rahul Singh
- Department
of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160 014, India
| | - Ayla das Chagas Almeida
- Department
of Parasitology, Microbiology and Immunology, Institute of Biological
Sciences, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | - Juliana da Trindade Granato
- Department
of Parasitology, Microbiology and Immunology, Institute of Biological
Sciences, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | - Ari Sérgio de Oliveira Lemos
- Department
of Parasitology, Microbiology and Immunology, Institute of Biological
Sciences, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | - Kushvinder Kumar
- Department
of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160 014, India
| | - Madhuri T. Patil
- Mehr
Chand Mahajan DAV College for Women, Sector 36, Chandigarh 160036, India
| | - Adilson D. da Silva
- Department
of Chemistry, Institute of Exacts Sciences, Federal University of Juiz de Fora, 36036-900, Juiz de Fora, Brazil
| | - Ambadas B. Rode
- Regional
Centre for Biotechnology, NCR Biotech Science
Cluster, third Milestone, Faridabad-Gurgaon Expressway, Faridabad - 121 001, India
| | - Elaine S. Coimbra
- Department
of Parasitology, Microbiology and Immunology, Institute of Biological
Sciences, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | - Deepak B. Salunke
- Department
of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160 014, India
- National
Interdisciplinary Centre of Vaccine, Immunotherapeutic and Antimicrobials, Panjab University, Chandigarh 160 014, India
| |
Collapse
|
23
|
Sousa JPAD, Sousa JMSD, Rodrigues RRL, Nunes TADL, Machado YAA, Araujo ACD, da Silva IGM, Barros-Cordeiro KB, Báo SN, Alves MMDM, Mendonça-Junior FJB, Rodrigues KADF. Antileishmanial activity of 2-amino-thiophene derivative SB-200. Int Immunopharmacol 2023; 123:110750. [PMID: 37536181 DOI: 10.1016/j.intimp.2023.110750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/28/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023]
Abstract
Leishmaniasis, presenting the highest number of cases worldwide is one of the most serious Neglected Tropical Diseases (NTDs). Clinical manifestations are intrinsically related to the host's immune response making immunomodulatory substances the target of numerous studies on antileishmanial activity. The currently available drugs used for treatment present various problems including high toxicity, low efficacy, and associated drug resistance. The search for therapeutic alternatives is urgent, and in this context, thiophene derivatives appear to be a promising therapeutic alternative (many have shown promising anti-leishmanial activity). The objective of this study was to investigate the antileishmanial activity of the 2-amino-thiophenic derivative SB-200. The thiophenic derivative was effective in inhibiting the growth of Leishmania braziliensis, Leishmania major, and Leishmania infantum promastigotes, obtaining respective IC50 values of 4.25 μM, 4.65 μM, and 3.96 μM. For L. infantum, it was demonstrated that the antipromastigote effect of SB-200 is associated with cell membrane integrity losses, and with morphological changes observed during scanning and transmission electron microscopy. Cytotoxicity was performed for J774.A1 macrophages and VERO cells, to obtain a CC50 of 42.52 μM and a SI of 10.74 for macrophages and a CC50 of 39.2 μM and an SI of 9.89 for VERO cells. The anti-amastigote activity of SB-200 revealed an IC50 of 2.85 μM and an SI of 14.97 against macrophages and SI of 13.8 for VERO cells. The anti-amastigote activity of SB-200 is associated with in vitro immunomodulation. For acute toxicity, SB-200 against Zophobas morio larvae permitted 100% survival. We conclude that the 2-amino-thiophenic derivative SB-200 is a promising candidate for in vivo anti-leishmania drug tests to evaluate its activity, efficacy, and safety.
Collapse
Affiliation(s)
- João Paulo Araujo de Sousa
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Julyanne Maria Saraiva de Sousa
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Raiza Raianne Luz Rodrigues
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Thais Amanda de Lima Nunes
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Yasmim Alves Aires Machado
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Alexandre Carvalho de Araujo
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Ingrid Gracielle Martins da Silva
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Karine Brenda Barros-Cordeiro
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Sônia Nair Báo
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Michel Muálem de Moraes Alves
- Laboratory of Antileishmania Activity, Medicinal Plants Research Center, Federal University of Piauí, Teresina 64049-550, Brazil
| | | | | |
Collapse
|
24
|
Santos GDA, Sousa JM, de Aguiar AHBM, Torres KCS, Coelho AJS, Ferreira AL, Lima MIS. Systematic Review of Treatment Failure and Clinical Relapses in Leishmaniasis from a Multifactorial Perspective: Clinical Aspects, Factors Associated with the Parasite and Host. Trop Med Infect Dis 2023; 8:430. [PMID: 37755891 PMCID: PMC10534360 DOI: 10.3390/tropicalmed8090430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023] Open
Abstract
Leishmaniasis is a disease caused by protozoa of the genus Leishmania. Treatment options are limited, and there are frequent cases of treatment failure and clinical relapse. To understand these phenomena better, a systematic review was conducted, considering studies published between 1990 and 2021 in Portuguese, English, and Spanish. The review included 64 articles divided into three categories. Case reports (26 articles) focused on treatment failure and clinical relapse in cutaneous leishmaniasis patients (47.6%), primarily affecting males (74%) and children (67%), regardless of the clinical manifestation. Experimental studies on the parasite (19 articles), particularly with L. major (25%), indicated that alterations in DNA and genic expression (44.82%) played a significant role in treatment failure and clinical relapse. Population data on the human host (19 articles) identified immunological characteristics as the most associated factor (36%) with treatment failure and clinical relapse. Each clinical manifestation of the disease presented specificities in these phenomena, suggesting a multifactorial nature. Additionally, the parasites were found to adapt to the drugs used in treatment. In summary, the systematic review revealed that treatment failure and clinical relapse in leishmaniasis are complex processes influenced by various factors, including host immunology and parasite adaptation.
Collapse
Affiliation(s)
- Gustavo de Almeida Santos
- Postgraduate Program in Health and Environment, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil;
- Department of Biology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil; (J.M.S.); (A.H.B.M.d.A.); (K.C.S.T.); (A.J.S.C.); (A.L.F.)
| | - Juliana Mendes Sousa
- Department of Biology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil; (J.M.S.); (A.H.B.M.d.A.); (K.C.S.T.); (A.J.S.C.); (A.L.F.)
| | - Antônio Henrique Braga Martins de Aguiar
- Department of Biology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil; (J.M.S.); (A.H.B.M.d.A.); (K.C.S.T.); (A.J.S.C.); (A.L.F.)
| | - Karina Cristina Silva Torres
- Department of Biology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil; (J.M.S.); (A.H.B.M.d.A.); (K.C.S.T.); (A.J.S.C.); (A.L.F.)
- Postgraduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil
| | - Ana Jessica Sousa Coelho
- Department of Biology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil; (J.M.S.); (A.H.B.M.d.A.); (K.C.S.T.); (A.J.S.C.); (A.L.F.)
- Postgraduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil
| | - André Leite Ferreira
- Department of Biology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil; (J.M.S.); (A.H.B.M.d.A.); (K.C.S.T.); (A.J.S.C.); (A.L.F.)
| | - Mayara Ingrid Sousa Lima
- Postgraduate Program in Health and Environment, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil;
- Department of Biology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil; (J.M.S.); (A.H.B.M.d.A.); (K.C.S.T.); (A.J.S.C.); (A.L.F.)
- Postgraduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil
| |
Collapse
|
25
|
dos Santos FS, de Freitas RP, de Freitas CS, Mendonça DVC, Lage DP, Tavares GDSV, Machado AS, Martins VT, Costa AV, de Queiroz VT, de Oliveira MB, de Oliveira FM, Antinarelli LMR, Coimbra ES, Pilau EJ, da Silva GP, Coelho EAF, Teixeira RR. Synthesis of 1,2,3-Triazole-Containing Methoxylated Cinnamides and Their Antileishmanial Activity against the Leishmania braziliensis Species. Pharmaceuticals (Basel) 2023; 16:1113. [PMID: 37631028 PMCID: PMC10459042 DOI: 10.3390/ph16081113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Leishmaniasis is a group of infectious diseases caused by protozoan parasites that belong to the genus Leishmania. Currently, there is no human vaccine, and the available treatments are associated with toxicity, high cost, and the emergence of resistant strains. These factors highlight the need to identify new antileishmanial candidates. In this study, we synthesized twenty-four methoxylated cinnamides containing 1,2,3-triazole fragments and evaluated their antileishmanial activity against the Leishmania braziliensis species, which is the main etiological agent responsible for American Tegumentary Leishmaniasis (ATL). The cinnamides were synthetically prepared using nucleophilic acyl substitution and copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC) reactions. The compounds were characterized using infrared, nuclear magnetic resonance, and high-resolution mass spectrometry techniques. We performed preliminary studies to evaluate the biological activity of these compounds against L. braziliensis promastigotes and axenic amastigotes. Compound 28, N-((1-(7-(diethylamino)-2-oxo-2H-chromen-3-yl)-1H-1,2,3-triazole-4-yl) methyl)-3,4-dimethoxy cinnamide, demonstrated relevant antileishmanial activity with low toxicity in murine cells. The selectivity index values for this compound were superior compared with data obtained using amphotericin B. Furthermore, this cinnamide derivative reduced the infection percentage and number of recovered amastigotes in L. braziliensis-infected macrophages. It also induced an increase in reactive oxygen species production, depolarization of the mitochondrial potential, and disruption of the parasite membrane. Taken together, these findings suggest that this synthetic compound holds potential as an antileishmanial candidate and should be considered for future studies in the treatment of ATL.
Collapse
Affiliation(s)
- Fabíola Suelen dos Santos
- Laboratório de Síntese Orgânica (LABSINTO), Departamento de Química, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; (F.S.d.S.); (R.P.d.F.)
| | - Rossimiriam Pereira de Freitas
- Laboratório de Síntese Orgânica (LABSINTO), Departamento de Química, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; (F.S.d.S.); (R.P.d.F.)
| | - Camila Simões de Freitas
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; (C.S.d.F.); (D.V.C.M.); (D.P.L.); (G.d.S.V.T.); (A.S.M.); (V.T.M.); (E.A.F.C.)
| | - Débora Vasconcelos Costa Mendonça
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; (C.S.d.F.); (D.V.C.M.); (D.P.L.); (G.d.S.V.T.); (A.S.M.); (V.T.M.); (E.A.F.C.)
| | - Daniela Pagliara Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; (C.S.d.F.); (D.V.C.M.); (D.P.L.); (G.d.S.V.T.); (A.S.M.); (V.T.M.); (E.A.F.C.)
| | - Grasiele de Sousa Vieira Tavares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; (C.S.d.F.); (D.V.C.M.); (D.P.L.); (G.d.S.V.T.); (A.S.M.); (V.T.M.); (E.A.F.C.)
| | - Amanda Sanchez Machado
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; (C.S.d.F.); (D.V.C.M.); (D.P.L.); (G.d.S.V.T.); (A.S.M.); (V.T.M.); (E.A.F.C.)
| | - Vivian Tamieti Martins
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil; (C.S.d.F.); (D.V.C.M.); (D.P.L.); (G.d.S.V.T.); (A.S.M.); (V.T.M.); (E.A.F.C.)
| | - Adilson Vidal Costa
- Grupo de Estudo Aplicado em Produtos Naturais e Síntese Orgânica (GEAPS), Departamento de Química e Física, Universidade Federal do Espírito Santo, Alegre 29500-000, Espírito Santo, Brazil; (A.V.C.); (V.T.d.Q.); (M.B.d.O.)
| | - Vagner Tebaldi de Queiroz
- Grupo de Estudo Aplicado em Produtos Naturais e Síntese Orgânica (GEAPS), Departamento de Química e Física, Universidade Federal do Espírito Santo, Alegre 29500-000, Espírito Santo, Brazil; (A.V.C.); (V.T.d.Q.); (M.B.d.O.)
| | - Mariana Belizario de Oliveira
- Grupo de Estudo Aplicado em Produtos Naturais e Síntese Orgânica (GEAPS), Departamento de Química e Física, Universidade Federal do Espírito Santo, Alegre 29500-000, Espírito Santo, Brazil; (A.V.C.); (V.T.d.Q.); (M.B.d.O.)
| | | | - Luciana Maria Ribeiro 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; (L.M.R.A.); (E.S.C.)
| | - Elaine Soares 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; (L.M.R.A.); (E.S.C.)
| | - Eduardo Jorge Pilau
- Centro de Ciências Exatas, Departamento de Química, Universidade Estadual de Maringá, Maringá 87020-900, Paraná, Brazil; (E.J.P.); (G.P.d.S.)
| | - Geovane Perez da Silva
- Centro de Ciências Exatas, Departamento de Química, Universidade Estadual de Maringá, Maringá 87020-900, Paraná, Brazil; (E.J.P.); (G.P.d.S.)
| | - 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 31270-901, Minas Gerais, Brazil; (C.S.d.F.); (D.V.C.M.); (D.P.L.); (G.d.S.V.T.); (A.S.M.); (V.T.M.); (E.A.F.C.)
| | - Róbson Ricardo Teixeira
- Grupo de Síntese e Pesquisa de Compostos Bioativos (GSPCB), Departamento de Química, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| |
Collapse
|
26
|
Garcia AR, Amorim MMB, Amaral ACF, da Cruz JD, Vermelho AB, Nico D, Rodrigues IA. Anti- Leishmania amazonensis Activity, Cytotoxic Features, and Chemical Profile of Allium sativum (Garlic) Essential Oil. Trop Med Infect Dis 2023; 8:375. [PMID: 37505671 PMCID: PMC10384145 DOI: 10.3390/tropicalmed8070375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023] Open
Abstract
Human tegumentary leishmaniasis (HTL) is a serious tropical disease caused by Leishmania amazonensis. Developing new leishmanicidal agents can help overcome current treatment challenges, such as drug resistance and toxicity. Essential oils are a source of lipophilic substances with diverse therapeutic properties. This study aimed to determine the anti-L. amazonensis activity, cytotoxicity, and chemical profile of Allium sativum essential oil (ASEO). The effect of ASEO on parasite and mammalian cells viability was evaluated using resazurin and MTT assays, respectively. The oil's effect against intracellular amastigotes was also determined. Transmission electron microscopy was used to assess the ultrastructural changes induced by ASEO. In addition, the chemical constituents of ASEO were identified by gas chromatography-mass spectrometry (GC-MS). The cytotoxic potential was evaluated in vitro and in silico. The oil displayed IC50 of 1.76, 3.46, and 3.77 µg/mL against promastigotes, axenic, and intracellular amastigotes, respectively. Photomicrographs of treated parasites showed plasma membrane disruption, increased lipid bodies, and autophagic-like structures. ASEO chemical profiling revealed 1,2,4,6-tetrathiepane (24.84%) and diallyl disulfide (16.75%) as major components. Computational pharmacokinetics and toxicological analysis of ASEO's major components demonstrated good oral bioavailability and better toxicological endpoints than the reference drugs. Altogether, the results suggest that ASEO could be an alternative drug candidate against HTL.
Collapse
Affiliation(s)
- Andreza R Garcia
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Mariana M B Amorim
- Instituto Municipal de Vigilância Sanitária, Vigilância de Zoonoses e de Inspeção Agropecuária, Rio de Janeiro 22290-240, Brazil
| | - Ana Claudia F Amaral
- Departamento de Produtos Naturais, Farmanguinhos Fiocruz, Manguinhos, Rio de Janeiro 21041-250, Brazil
| | - Jefferson D da Cruz
- Departamento de Produtos Naturais, Farmanguinhos Fiocruz, Manguinhos, Rio de Janeiro 21041-250, Brazil
| | - Alane B Vermelho
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Dirlei Nico
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Igor A Rodrigues
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Departamento de Produtos Naturais e Alimentos, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
27
|
Ghasemian Yadegari J, Khudair Khalaf A, Ezzatkhah F, Shakibaie M, Mohammadi HR, Mahmoudvand H. Antileishmanial, cellular mechanisms, and cytotoxic effects of green synthesized zinc nanoparticles alone and in combined with glucantime against Leishmania major infection. Biomed Pharmacother 2023; 164:114984. [PMID: 37321058 DOI: 10.1016/j.biopha.2023.114984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND We decided to investigate the antileishmanial, cellular mechanisms, and cytotoxic effects of green synthesized Zinc nanoparticles (ZnNPs) alone and combined with glucantime against Leishmania major infection. METHODS The effect of green synthesized ZnNP on L. major amastigote was studied through macrophage cells. The mRNA expression level of iNOS and IFN-γ followed by the exposure of J774-A1 macrophage cells to ZnNPs was assessed by Real-time PCR. The Caspase-3-like activity of promastigotes exposed to ZnNPs was studied. Effects of ZnNPs alone and combined with glucantime (MA) were studied on cutaneous leishmaniasis in BALB/c mice. RESULTS ZnNPs displayed the spherical shape with sizes ranging from 30 to 80 nm. The obtained IC50 values for ZnNPs, MA, and ZnNPs + MA were 43.2, 26.3, and 12.6 µg/mL, respectively; indicating the synergistic effects of ZnNPs in combination with MA. CL lesions had completely improved in the mice received with ZnNPs in combination with MA. The mRNA expression level of iNOS, TNF-α, and IFN-γ was dose-dependently (p < 0.01) upregulated; whereas it was downregulated in IL-10. ZnNPs markedly stimulated the caspase-3 activation with no significant toxicity on normal cells. CONCLUSION Based on these in vitro and in vivo results, green synthesized ZnNPs, mainly along with MA, showed that has the potential to be introduced as a new drug for CL therapy. Triggering of NO production, and inhibition of infectivity rate are revealed as mechanisms of action ZnNPs on L. major. But, supplementary investigations are necessary to clear the efficacy and safety of these agents.
Collapse
Affiliation(s)
- Javad Ghasemian Yadegari
- Department of Pharmacognosy, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Fatemeh Ezzatkhah
- Department of Laboratory Sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Mojtaba Shakibaie
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Reza Mohammadi
- Department of Pharmacognosy, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hossein Mahmoudvand
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Molecular and Cellular Laboratory, School of Allied Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
28
|
Nascimento IJDS, Cavalcanti MDAT, de Moura RO. Exploring N-myristoyltransferase as a promising drug target against parasitic neglected tropical diseases. Eur J Med Chem 2023; 258:115550. [PMID: 37336067 DOI: 10.1016/j.ejmech.2023.115550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Neglected tropical diseases (NTDs) constitute a group of approximately 20 infectious diseases that mainly affect the impoverished population without basic sanitation in tropical countries. These diseases are responsible for many deaths worldwide, costing billions of dollars in public health investment to treat and control these infections. Among them are the diseases caused by protozoa of the Trypanosomatid family, which constitute Trypanosoma cruzi (Chagas disease), Trypanosoma brucei (sleeping sickness), and Leishmaniasis. In addition, there is a classification of other diseases, called the big three, AIDS, tuberculosis, and malaria, which are endemic in countries with tropical conditions. Despite the high mortality rates, there is still a gap in the treatment. The drugs have a high incidence of side effects and protozoan resistance, justifying the investment in developing new alternatives. In fact, the Target-Based Drug Design (TBDD) approach is responsible for identifying several promising compounds, and among the targets explored through this approach, N-myristoyltransferase (NMT) stands out. It is an enzyme related to the co-translational myristoylation of N-terminal glycine in various peptides. The myristoylation process is a co-translation that occurs after removing the initiator methionine. This process regulates the assembly of protein complexes and stability, which justifies its potential as a drug target. In order to propose NMT as a potential target for parasitic diseases, this review will address the entire structure and function of this enzyme and the primary studies demonstrating its promising potential against Leishmaniasis, T. cruzi, T. brucei, and malaria. We hope our information can help researchers worldwide search for potential drugs against these diseases that have been threatening the health of the world's population.
Collapse
Affiliation(s)
- Igor José Dos Santos Nascimento
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil; Cesmac University Center, Pharmacy Departament, Maceió, Brazil; Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil.
| | - Misael de Azevedo Teotônio Cavalcanti
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil; Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| | - Ricardo Olimpio de Moura
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil; Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| |
Collapse
|
29
|
Barazorda-Ccahuana HL, Goyzueta-Mamani LD, Candia Puma MA, Simões de Freitas C, de Sousa Vieria Tavares G, Pagliara Lage D, Ferraz Coelho EA, Chávez-Fumagalli MA. Computer-aided drug design approaches applied to screen natural product's structural analogs targeting arginase in Leishmania spp. F1000Res 2023; 12:93. [PMID: 37424744 PMCID: PMC10323282 DOI: 10.12688/f1000research.129943.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction: Leishmaniasis is a disease with high mortality rates and approximately 1.5 million new cases each year. Despite the new approaches and advances to fight the disease, there are no effective therapies. Methods: Hence, this study aims to screen for natural products' structural analogs as new drug candidates against leishmaniasis. We applied Computer-aided drug design (CADD) approaches, such as virtual screening, molecular docking, molecular dynamics simulation, molecular mechanics-generalized Born surface area (MM-GBSA) binding free estimation, and free energy perturbation (FEP) aiming to select structural analogs from natural products that have shown anti-leishmanial and anti-arginase activities and that could bind selectively against the Leishmania arginase enzyme. Results: The compounds 2H-1-benzopyran, 3,4-dihydro-2-(2-methylphenyl)-(9CI), echioidinin, and malvidin showed good results against arginase targets from three parasite species and negative results for potential toxicities. The echioidinin and malvidin ligands generated interactions in the active center at pH 2.0 conditions by MM-GBSA and FEP methods. Conclusions: This work suggests the potential anti-leishmanial activity of the compounds and thus can be further in vitro and in vivo experimentally validated.
Collapse
Affiliation(s)
- Haruna Luz Barazorda-Ccahuana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Arequipa, Peru
| | - Luis Daniel Goyzueta-Mamani
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Arequipa, Peru
- Sustainable Innovative Biomaterials Department, Le Qara Research Center, Arequipa, Peru
| | - Mayron Antonio Candia Puma
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Arequipa, Peru
- Universidad Católica de Santa María, Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Arequipa, Peru
| | - Camila Simões de Freitas
- Universidade Federal de Minas Gerais, Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Belo Horizonte, Minas Gerais, Brazil
| | - Grasiele de Sousa Vieria Tavares
- Universidade Federal de Minas Gerais, Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela Pagliara Lage
- Universidade Federal de Minas Gerais, Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo Antonio Ferraz Coelho
- Universidade Federal de Minas Gerais, Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Belo Horizonte, Minas Gerais, Brazil
- Universidade Federal de Minas Gerais, Departamento de Patologia Clínica, COLTEC, Belo Horizonte, Minas Gerais, Brazil
| | - Miguel Angel Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Arequipa, Peru
| |
Collapse
|
30
|
AlGabbani Q. Nanotechnology: A promising strategy for the control of parasitic infections. Exp Parasitol 2023:108548. [PMID: 37196702 DOI: 10.1016/j.exppara.2023.108548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/17/2023] [Accepted: 05/13/2023] [Indexed: 05/19/2023]
Abstract
Annually 3.5 billion people are affected by the parasitic infections that results around 200,000 deaths per annum. Major diseases occur due to the neglected tropical parasites. Variety of methods have been used to treat the parasitic infections but now these methods have become ineffective due to the development of resistance in the parasites and some other side effects of traditional treatment methods. Previous methods include use of chemotherapeutic agents and ethnobotanicals for the treatment of parasites. Parasites have developed resistance against the chemotherapeutic agents. A major problem related to Ethnobotanicals is the unequal availability of drug at the target site which is responsible for the low efficacy of drug. Nanotechnology technology involves the manipulation of matter on a nanoscale level and has the potential to enhance the efficacy and safety of existing drugs, develop new treatments, and improve diagnostic methods for parasitic infections. Nanoparticles can be designed to selectively target parasites while minimizing toxicity to the host, and they can also be used to improve drug delivery and increase drug stability. Some important nanotechnology-based tools for parasitic control include nanoparticle-based drug delivery, nanoparticle diagnostics, nanoparticle vaccines, nanoparticle insecticides. Nanotechnology has the potential to revolutionize the field of parasitic control by providing new methods for detection, prevention and treatment of parasitic infections. This review discusses the current state of nanotechnology-based approaches for controlling parasitic infections and highlights their potential to revolutionize the field of parasitology.
Collapse
Affiliation(s)
- Qwait AlGabbani
- Department of Biology, College of Sciences and Humanities, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| |
Collapse
|
31
|
Berneburg I, Stumpf M, Velten AS, Rahlfs S, Przyborski J, Becker K, Fritz-Wolf K. Structure of Leishmania donovani 6-Phosphogluconate Dehydrogenase and Inhibition by Phosphine Gold(I) Complexes: A Potential Approach to Leishmaniasis Treatment. Int J Mol Sci 2023; 24:ijms24108615. [PMID: 37239962 DOI: 10.3390/ijms24108615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
As unicellular parasites are highly dependent on NADPH as a source for reducing equivalents, the main NADPH-producing enzymes glucose 6-phosphate dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase (6PGD) of the pentose phosphate pathway are considered promising antitrypanosomatid drug targets. Here we present the biochemical characterization and crystal structure of Leishmania donovani 6PGD (Ld6PGD) in complex with NADP(H). Most interestingly, a previously unknown conformation of NADPH is visible in this structure. In addition, we identified auranofin and other gold(I)-containing compounds as efficient Ld6PGD inhibitors, although it has so far been assumed that trypanothione reductase is the sole target of auranofin in Kinetoplastida. Interestingly, 6PGD from Plasmodium falciparum is also inhibited at lower micromolar concentrations, whereas human 6PGD is not. Mode-of-inhibition studies indicate that auranofin competes with 6PG for its binding site followed by a rapid irreversible inhibition. By analogy with other enzymes, this suggests that the gold moiety is responsible for the observed inhibition. Taken together, we identified gold(I)-containing compounds as an interesting class of inhibitors against 6PGDs from Leishmania and possibly from other protozoan parasites. Together with the three-dimensional crystal structure, this provides a valid basis for further drug discovery approaches.
Collapse
Affiliation(s)
- Isabell Berneburg
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, 35392 Giessen, Germany
| | - Michaela Stumpf
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, 35392 Giessen, Germany
| | - Ann-Sophie Velten
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, 35392 Giessen, Germany
| | - Stefan Rahlfs
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, 35392 Giessen, Germany
| | - Jude Przyborski
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, 35392 Giessen, Germany
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, 35392 Giessen, Germany
| | - Karin Fritz-Wolf
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, 35392 Giessen, Germany
- Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| |
Collapse
|
32
|
Lima SKSD, Jesus JA, Raminelli C, Laurenti MD, Passero LFD. High Selectivity of 8-Hydroxyquinoline on Leishmania (Leishmania) and Leishmania (Viannia) Species Correlates with a Potent Therapeutic Activity In Vivo. Pharmaceuticals (Basel) 2023; 16:ph16050707. [PMID: 37242490 DOI: 10.3390/ph16050707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Leishmaniasis is a neglected disease caused by protozoa of the genus Leishmania, which causes different clinical manifestations. Drugs currently used in the treatment such as pentavalent antimonial and amphotericin B cause severe side effects in patients, and parasite resistance has been reported. Thus, it is necessary and urgent to characterize new and effective alternative drugs to replace the current chemotherapy of leishmaniasis. In this regard, it has been experimentally demonstrated that quinoline derivatives present significative pharmacological and parasitic properties. Thus, the aim of this work was to demonstrate the leishmanicidal activity of 8-hydroxyquinoline (8-HQ) in vitro and in vivo. The leishmanicidal activity (in vitro) of 8-HQ was assayed on promastigote and intracellular amastigote forms of L. (L.) amazonensis, L. (L.) infantum chagasi, L. (V.) guyanensis L. (V.) naiffi, L. (V.) lainsoni, and L. (V.) shawi. Additionally, the levels of nitric oxide and hydrogen peroxide were analyzed. The therapeutic potential of 8-HQ was analyzed in BALB/c mice infected with a strain of L. (L.) amazonensis that causes anergic cutaneous diffuse leishmaniasis. In vitro data showed that at 24 and 72 h, 8-HQ eliminated promastigote and intracellular amastigote forms of all studied species and this effect may be potentialized by nitric oxide. Furthermore, 8-HQ was more selective than miltefosine. Infected animals treated with 8-HQ by the intralesional route dramatically reduced the number of tissue parasites in the skin, and it was associated with an increase in IFN-γ and decrease in IL-4, which correlated with a reduction in inflammatory reaction in the skin. These results strongly support the idea that 8-HQ is an alternative molecule that can be employed in the treatment of leishmaniasis, given its selectivity and multispectral action in parasites from the Leishmania genus.
Collapse
Affiliation(s)
- Sarah Kymberly Santos de Lima
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, São Paulo 01246-903, Brazil
| | - Jéssica Adriana Jesus
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil
| | - Cristiano Raminelli
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema 09920-000, Brazil
| | - Márcia Dalastra Laurenti
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, São Paulo 01246-903, Brazil
| | - Luiz Felipe Domingues Passero
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil
- Institute for Advanced Studies of Ocean, São Paulo State University (UNESP), Rua João Francisco Bensdorp, 1178, São Vicente 11350-011, Brazil
| |
Collapse
|
33
|
Glidden CK, Murran AR, Silva RA, Castellanos AA, Han BA, Mordecai EA. Phylogenetic and biogeographical traits predict unrecognized hosts of zoonotic leishmaniasis. PLoS Negl Trop Dis 2023; 17:e0010879. [PMID: 37256857 PMCID: PMC10231829 DOI: 10.1371/journal.pntd.0010879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 05/01/2023] [Indexed: 06/02/2023] Open
Abstract
The spatio-temporal distribution of leishmaniasis, a parasitic vector-borne zoonotic disease, is significantly impacted by land-use change and climate warming in the Americas. However, predicting and containing outbreaks is challenging as the zoonotic Leishmania system is highly complex: leishmaniasis (visceral, cutaneous and muco-cutaneous) in humans is caused by up to 14 different Leishmania species, and the parasite is transmitted by dozens of sandfly species and is known to infect almost twice as many wildlife species. Despite the already broad known host range, new hosts are discovered almost annually and Leishmania transmission to humans occurs in absence of a known host. As such, the full range of Leishmania hosts is undetermined, inhibiting the use of ecological interventions to limit pathogen spread and the ability to accurately predict the impact of global change on disease risk. Here, we employed a machine learning approach to generate trait profiles of known zoonotic Leishmania wildlife hosts (mammals that are naturally exposed and susceptible to infection) and used trait-profiles of known hosts to identify potentially unrecognized hosts. We found that biogeography, phylogenetic distance, and study effort best predicted Leishmania host status. Traits associated with global change, such as agricultural land-cover, urban land-cover, and climate, were among the top predictors of host status. Most notably, our analysis suggested that zoonotic Leishmania hosts are significantly undersampled, as our model predicted just as many unrecognized hosts as unknown hosts. Overall, our analysis facilitates targeted surveillance strategies and improved understanding of the impact of environmental change on local transmission cycles.
Collapse
Affiliation(s)
- Caroline K. Glidden
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Aisling Roya Murran
- Department of Biology, Stanford University, Stanford, California, United States of America
| | | | | | - Barbara A. Han
- Cary Institute of Ecosystem Studies, Millbrook, New York, United States of America
| | - Erin A. Mordecai
- Department of Biology, Stanford University, Stanford, California, United States of America
| |
Collapse
|
34
|
Alves AB, da Silva Bortoleti BT, Tomiotto-Pellissier F, Ganaza AFM, Gonçalves MD, Carloto ACM, Rodrigues ACJ, Silva TF, Nakazato G, Kobayashi RKT, Lazarin-Bidóia D, Miranda-Sapla MM, Costa IN, Pavanelli WR, Conchon-Costa I. Synergistic Antileishmanial Effect of Oregano Essential Oil and Silver Nanoparticles: Mechanisms of Action on Leishmania amazonensis. Pathogens 2023; 12:pathogens12050660. [PMID: 37242330 DOI: 10.3390/pathogens12050660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/18/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
American tegumentary leishmaniasis, a zoonotic disease caused by the Leishmania genus, poses significant challenges in treatment, including administration difficulty, low efficacy, and parasite resistance. Novel compounds or associations offer alternative therapies, and natural products such as oregano essential oil (OEO), extracted from Origanum vulgare, have been extensively researched due to biological effects, including antibacterial, antifungal, and antiparasitic properties. Silver nanoparticles (AgNp), a nanomaterial with compelling antimicrobial and antiparasitic activity, have been shown to exhibit potent leishmanicidal properties. We evaluated the in vitro effect of OEO and AgNp-Bio association on L. amazonensis and the death mechanisms of the parasite involved. Our results demonstrated a synergistic antileishmanial effect of OEO + AgNp on promastigote forms and L. amazonensis-infected macrophages, which induced morphological and ultrastructural changes in promastigotes. Subsequently, we investigated the mechanisms underlying parasite death and showed an increase in NO, ROS, mitochondrial depolarization, accumulation of lipid-storage bodies, autophagic vacuoles, phosphatidylserine exposure, and damage to the plasma membrane. Moreover, the association resulted in a reduction in the percentage of infected cells and the number of amastigotes per macrophage. In conclusion, our findings establish that OEO + AgNp elicits a late apoptosis-like mechanism to combat promastigote forms and promotes ROS and NO production in infected macrophages to target intracellular amastigote forms.
Collapse
Affiliation(s)
- Alex Barbosa Alves
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Bruna Taciane da Silva Bortoleti
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
- Carlos Chagas Institute (ICC-Fiocruz-Pr), Curitiba 81310-020, PR, Brazil
| | - Fernanda Tomiotto-Pellissier
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
- Carlos Chagas Institute (ICC-Fiocruz-Pr), Curitiba 81310-020, PR, Brazil
| | - Ana Flávia Marques Ganaza
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Manoela Daiele Gonçalves
- Laboratory of Biotransformation and Phytochemistry, Department of Chemistry, Center of Exact Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Amanda Cristina Machado Carloto
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Ana Carolina Jacob Rodrigues
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Taylon Felipe Silva
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | | | - Danielle Lazarin-Bidóia
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Milena Menegazzo Miranda-Sapla
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Idessania Nazareth Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Wander Rogério Pavanelli
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| | - Ivete Conchon-Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, PR, Brazil
| |
Collapse
|
35
|
Román-Álamo L, Allaw M, Avalos-Padilla Y, Manca ML, Manconi M, Fulgheri F, Fernández-Lajo J, Rivas L, Vázquez JA, Peris JE, Roca-Geronès X, Poonlaphdecha S, Alcover MM, Fisa R, Riera C, Fernàndez-Busquets X. In Vitro Evaluation of Aerosol Therapy with Pentamidine-Loaded Liposomes Coated with Chondroitin Sulfate or Heparin for the Treatment of Leishmaniasis. Pharmaceutics 2023; 15:pharmaceutics15041163. [PMID: 37111648 PMCID: PMC10147000 DOI: 10.3390/pharmaceutics15041163] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/09/2023] Open
Abstract
The second-line antileishmanial compound pentamidine is administered intramuscularly or, preferably, by intravenous infusion, with its use limited by severe adverse effects, including diabetes, severe hypoglycemia, myocarditis and renal toxicity. We sought to test the potential of phospholipid vesicles to improve the patient compliance and efficacy of this drug for the treatment of leishmaniasis by means of aerosol therapy. The targeting to macrophages of pentamidine-loaded liposomes coated with chondroitin sulfate or heparin increased about twofold (up to ca. 90%) relative to noncoated liposomes. The encapsulation of pentamidine in liposomes ameliorated its activity on the amastigote and promastigote forms of Leishmania infantum and Leishmania pifanoi, and it significantly reduced cytotoxicity on human umbilical endothelial cells, for which the concentration inhibiting 50% of cell viability was 144.2 ± 12.7 µM for pentamidine-containing heparin-coated liposomes vs. 59.3 ± 4.9 µM for free pentamidine. The deposition of liposome dispersions after nebulization was evaluated with the Next Generation Impactor, which mimics human airways. Approximately 53% of total initial pentamidine in solution reached the deeper stages of the impactor, with a median aerodynamic diameter of ~2.8 µm, supporting a partial deposition on the lung alveoli. Upon loading pentamidine in phospholipid vesicles, its deposition in the deeper stages significantly increased up to ~68%, and the median aerodynamic diameter decreased to a range between 1.4 and 1.8 µm, suggesting a better aptitude to reach the deeper lung airways in higher amounts. In all, nebulization of liposome-encapsulated pentamidine improved the bioavailability of this neglected drug by a patient-friendly delivery route amenable to self-administration, paving the way for the treatment of leishmaniasis and other infections where pentamidine is active.
Collapse
Affiliation(s)
- Lucía Román-Álamo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Mohamad Allaw
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, Italy
| | - Yunuen Avalos-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Maria Letizia Manca
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, Italy
| | - Maria Manconi
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, Italy
| | - Federica Fulgheri
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, Italy
| | - Jorge Fernández-Lajo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Luis Rivas
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - José Antonio Vázquez
- Group of Recycling and Valorization of Waste Materials (REVAL), Marine Research Institute (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain
| | - José Esteban Peris
- Department of Pharmacy and Pharmaceutical Technology, University of Valencia, 46100 Burjassot, Spain
| | - Xavier Roca-Geronès
- Section of Parasitology, Department of Biology, Health and Environment, Faculty of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - Srisupaph Poonlaphdecha
- Section of Parasitology, Department of Biology, Health and Environment, Faculty of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - Maria Magdalena Alcover
- Section of Parasitology, Department of Biology, Health and Environment, Faculty of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - Roser Fisa
- Section of Parasitology, Department of Biology, Health and Environment, Faculty of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - Cristina Riera
- Section of Parasitology, Department of Biology, Health and Environment, Faculty of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - Xavier Fernàndez-Busquets
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| |
Collapse
|
36
|
Integrated computational and experimental approach for novel anti-leishmanial molecules by targeting Dephospho-coenzyme A kinase. Int J Biol Macromol 2023; 232:123441. [PMID: 36708902 DOI: 10.1016/j.ijbiomac.2023.123441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/07/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
Coenzyme A acts as a necessary cofactor for many enzymes and is a part of many biochemical processes. One of the critical enzymes involved in Coenzyme A synthesis is Dephospho-coenzyme A-kinase (DPCK). In this study, we have used integrated computational and experimental approaches for promising inhibitors of DPCK using the natural products available in the ZINC database for anti-leishmanial drug development. The top hit compounds chosen after molecular docking were Veratramine, Azulene, Hupehenine, and Hederagenin. The free binding energy of Veratramine, Azulene, Hupehenine, and Hederagenin was estimated. Besides the favourable binding point, the ligands also showed good hydrogen bonding and other interactions with key residues of the enzyme's active site. The natural compounds were also experimentally investigated for their effect on the L. donovani promastigotes and murine macrophage (J774A.1). A good antileishmanial activity by the compounds on the promastigotes was observed as estimated by the MTT assay. The in-vitro experiments revealed that Hupehenine (IC50 = 7.34 ± 0.37 μM) and Veratramine (IC50 = 12.46 ± 2.28 μM) exhibited better inhibition than Hederagenin (IC50 = 23.36 ± 0.54 μM) and Azulene (IC50 = 24.42 ± 3.28 μM). This work has identified novel anti-leishmanial molecules possibly acting through the inhibition of DPCK.
Collapse
|
37
|
Hassan AHE, Mahmoud K, Phan TN, Shaldam MA, Lee CH, Kim YJ, Cho SB, Bayoumi WA, El-Sayed SM, Choi Y, Moon S, No JH, Lee YS. Bestatin analogs-4-quinolinone hybrids as antileishmanial hits: Design, repurposing rational, synthesis, in vitro and in silico studies. Eur J Med Chem 2023; 250:115211. [PMID: 36827952 DOI: 10.1016/j.ejmech.2023.115211] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/02/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023]
Abstract
Amongst different forms of leishmaniasis, visceral leishmaniasis caused by L. donovani is highly mortal. Identification of new hit compounds might afford new starting points to develop novel therapeutics. In this lieu, a rationally designed small library of bestatin analogs-4-quinolone hybrids were prepared and evaluated. Analysis of SAR unveiled distinct profiles for hybrids type 1 and type 2, which might arise from their different molecular targets. Amongst type 1 bestatin analog-4-quinolone hybrids, hybrid 1e was identified as potential hit inhibiting growth of L. donovani promastigotes by 91 and 53% at 50 and 25 μM concentrations, respectively. Meanwhile, hybrid 2j was identified amongst type 2 bestatin analog-4-quinolone hybrids as potential hit compound inhibiting growth of L. donovani promastigotes by 50 and 38% at 50 and 25 μM concentrations, respectively. Preliminary safety evaluation of the promising hit compounds showed that they are 50-100 folds safer against human derived monocytic THP-1 cells relative to the drug erufosine. In silico study was conducted to predict the possible binding of hybrid 1e with methionine aminopeptidases 1 and 2 of L. donovani. Molecular dynamic simulations verified the predicted binding modes and provide more in depth understanding of the impact of hybrid 1e on LdMetAP-1 and LdMetAP-2.
Collapse
Affiliation(s)
- Ahmed H E Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt; Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Kazem Mahmoud
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt
| | - Trong-Nhat Phan
- Host-Parasite Research Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Chae Hyeon Lee
- Department of Fundamental Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Yeon Ju Kim
- Department of Fundamental Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Soo Bin Cho
- Department of Fundamental Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Waleed A Bayoumi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Selwan M El-Sayed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Yeonwoo Choi
- Department of Fundamental Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Suyeon Moon
- Department of Fundamental Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Joo Hwan No
- Host-Parasite Research Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Yong Sup Lee
- Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Fundamental Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
38
|
de Sousa JMS, Nunes TADL, Rodrigues RRL, de Sousa JPA, Val MDCA, Coelho FADR, dos Santos ALS, Maciel NB, de Souza VMR, Machado YAA, Sousa PSDA, de Araújo AR, Rocha JA, de Sousa DP, da Silva MV, Arcanjo DDR, Rodrigues KADF. Cytotoxic and Antileishmanial Effects of the Monoterpene β-Ocimene. Pharmaceuticals (Basel) 2023; 16:183. [PMID: 37259336 PMCID: PMC9960243 DOI: 10.3390/ph16020183] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 08/05/2023] Open
Abstract
Leishmaniasis is a group of infectious-parasitic diseases with high mortality rates, and endemic in many regions of the globe. The currently available drugs present serious problems such as high toxicity, costs, and the emergence of drug resistance. This has stimulated research into new antileishmania drugs based on natural products and their derivatives. β-Ocimene is a monoterpene found naturally in the essential oils of many plant species which presents antileishmanial activity, and which has not yet been evaluated for its potential to inhibit the etiological agent of leishmaniasis. The aim of this work was to evaluate the activity of β-ocimene against Leishmania amazonensis, its cytotoxicity, and potential mechanisms of action. β-Ocimene presented direct activity against the parasite, with excellent growth inhibition of promastigotes (IC50 = 2.78 μM) and axenic amastigotes (EC50 = 1.12 μM) at concentrations non-toxic to RAW 264.7 macrophages (CC50 = 114.5 µM). The effect is related to changes in membrane permeability and resulting abnormalities in the parasitic cell shape. These were, respectively, observed in membrane integrity and atomic force microscopy assays. β-Ocimene was also shown to act indirectly, with greater activity against intra-macrophagic amastigotes (EC50 = 0.89 μM), increasing TNF-α, nitric oxide (NO), and reactive oxygen species (ROS), with lysosomal effects, as well as promoting decreases in IL-10 and IL-6. Against intra-macrophagic amastigote forms the selectivity index was higher than the reference drugs, being 469.52 times more selective than meglumine antimoniate, and 42.88 times more selective than amphotericin B. Our results suggest that β-ocimene possesses promising in vitro antileishmania activity and is a potential candidate for investigation in in vivo assays.
Collapse
Affiliation(s)
- Julyanne Maria Saraiva de Sousa
- Laboratory of Infectious Diseases, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - Thaís Amanda de Lima Nunes
- Laboratory of Infectious Diseases, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - Raiza Raianne Luz Rodrigues
- Laboratory of Infectious Diseases, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - João Paulo Araújo de Sousa
- Laboratory of Infectious Diseases, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | | | - Francisco Alex da Rocha Coelho
- Laboratory of Infectious Diseases, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - Airton Lucas Sousa dos Santos
- Laboratory of Infectious Diseases, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - Nicolle Barreira Maciel
- Laboratory of Infectious Diseases, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - Vanessa Maria Rodrigues de Souza
- Laboratory of Infectious Diseases, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - Yasmim Alves Aires Machado
- Laboratory of Infectious Diseases, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - Paulo Sérgio de Araújo Sousa
- Research Group on Medicinal Chemistry and Biotechnology, Federal University of Maranhão, São Bernardo 65550-000, MA, Brazil
| | - Alyne Rodrigues de Araújo
- Research Center in Biodiversity and Biotechnology, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - Jefferson Almeida Rocha
- Research Group on Medicinal Chemistry and Biotechnology, Federal University of Maranhão, São Bernardo 65550-000, MA, Brazil
- Research Center in Biodiversity and Biotechnology, Campus Ministro Reis Velloso, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil
| | - Damião Pergentino de Sousa
- Laboratory of Pharmaceutical Chemistry, Department of Pharmaceutical Sciences, Campus I, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| | - Marcos Vinicius da Silva
- Laboratory of Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | | | | |
Collapse
|
39
|
Miltefosine and Nifuratel Combination: A Promising Therapy for the Treatment of Leishmania donovani Visceral Leishmaniasis. Int J Mol Sci 2023; 24:ijms24021635. [PMID: 36675150 PMCID: PMC9865052 DOI: 10.3390/ijms24021635] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Visceral leishmaniasis is a neglected vector-borne tropical disease caused by Leishmania donovani and Leishmania infantum that is endemic not only in East African countries, but also in Asia, regions of South America and the Mediterranean Basin. For the pharmacological control of this disease, there is a limited number of old and, in general, poorly adherent drugs, with a multitude of adverse effects and low oral bioavailability, which favor the emergence of resistant pathogens. Pentavalent antimonials are the first-line drugs, but due to their misuse, resistant Leishmania strains have emerged worldwide. Although these drugs have saved many lives, it is recommended to reduce their use as much as possible and replace them with novel and more friendly drugs. From a commercial collection of anti-infective drugs, we have recently identified nifuratel-a nitrofurantoin used against vaginal infections-as a promising repurposing drug against a mouse model of visceral leishmaniasis. In the present work, we have tested combinations of miltefosine-the only oral drug currently used against leishmaniasis-with nifuratel in different proportions, both in axenic amastigotes from bone marrow and in intracellular amastigotes from infected Balb/c mouse spleen macrophages, finding a potent synergy in both cases. In vivo evaluation of oral miltefosine/nifuratel combinations using a bioimaging platform has revealed the potential of these combinations for the treatment of this disease.
Collapse
|
40
|
Crystal structure of Leishmania donovani glucose 6-phosphate dehydrogenase reveals a unique N-terminal domain. Commun Biol 2022; 5:1353. [PMID: 36494598 PMCID: PMC9734377 DOI: 10.1038/s42003-022-04307-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Since unicellular parasites highly depend on NADPH as a source for reducing equivalents, the pentose phosphate pathway, especially the first and rate-limiting NADPH-producing enzyme glucose 6-phosphate dehydrogenase (G6PD), is considered an excellent antitrypanosomatid drug target. Here we present the crystal structure of Leishmania donovani G6PD (LdG6PD) elucidating the unique N-terminal domain of Kinetoplastida G6PDs. Our investigations on the function of the N-domain suggest its involvement in the formation of a tetramer that is completely different from related Trypanosoma G6PDs. Structural and functional investigations further provide interesting insights into the binding mode of LdG6PD, following an ordered mechanism, which is confirmed by a G6P-induced domain shift and rotation of the helical N-domain. Taken together, these insights into LdG6PD contribute to the understanding of G6PDs' molecular mechanisms and provide an excellent basis for further drug discovery approaches.
Collapse
|
41
|
Magalhães LS, Melo EV, Damascena NP, Albuquerque ACB, Santos CNO, Rebouças MC, Bezerra MDO, Louzada da Silva R, de Oliveira FA, Santos PL, da Silva JS, Lipscomb MW, da Silva ÂM, de Jesus AR, de Almeida RP. Use of N-acetylcysteine as treatment adjuvant regulates immune response in visceral leishmaniasis: Pilot clinical trial and in vitro experiments. Front Cell Infect Microbiol 2022; 12:1045668. [PMID: 36506010 PMCID: PMC9730326 DOI: 10.3389/fcimb.2022.1045668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
This investigation aimed to assess the effect of N-acetylcysteine (NAC) as an adjuvant treatment to alleviate visceral leishmaniasis (VL). The present work includes both blinded randomized clinical intervention and experimental in vitro studies. The clinical trial included 60 patients with VL randomly allocated into two groups: a test group (n = 30) treated with meglumine antimoniate plus NAC (SbV + NAC) and a control group (n = 30) treated with meglumine antimoniate only (SbV). The primary outcome was clinical cure (absence of fever, spleen and liver sizes reduction, and hematological improvement) in 180 days. The cure rate did not differ between the groups; both groups had similar results in all readout indices. The immunological parameters of the patients treated with SbV + NAC showed higher sCD40L in sera during treatment, and the levels of sCD40L were negatively correlated with Interleukin-10 (IL-10) serum levels. In addition, data estimation showed a negative correlation between the sCD40L levels and the spleen size in patients with VL. For the in vitro experiments, peripheral blood mononuclear cells (PBMCs) or PBMC-derived macrophages from healthy donors were exposed to soluble Leishmania antigen (SLA) or infected with stationary promastigotes of Leishmania infantum in the presence or absence of NAC. Results revealed that NAC treatment of SLA-stimulated PBMCs reduces the frequency of monocytes producing IL-10 and lowers the frequency of CD4+ and CD8+ T cells expressing (pro-)inflammatory cytokines. Together, these results suggest that NAC treatment may modulate the immune response in patients with VL, thus warranting additional investigations to support its case use as an adjuvant to antimony therapy for VL.
Collapse
Affiliation(s)
- Lucas Sousa Magalhães
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil,Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Brazil,Sector of Parasitology and Pathology, Biological and Health Sciences Institute, Federal University of Alagoas, Maceió, Brazil
| | - Enaldo Vieira Melo
- Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil
| | - Nayra Prata Damascena
- Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil
| | - Adriana Cardoso Batista Albuquerque
- Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil
| | - Camilla Natália Oliveira Santos
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil,Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Brazil
| | - Mônica Cardozo Rebouças
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil
| | - Mariana de Oliveira Bezerra
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil,Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil
| | - Ricardo Louzada da Silva
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil,Department of Health Education, Federal University of Sergipe, Lagarto, Brazil
| | - Fabricia Alvisi de Oliveira
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil
| | - Priscila Lima Santos
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil,Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Brazil,Department of Health Education, Federal University of Sergipe, Lagarto, Brazil
| | - João Santana da Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Ângela Maria da Silva
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil,Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil
| | - Amélia Ribeiro de Jesus
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil,Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Brazil,Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil,Immunology Institute of Investigation (iii), National Institute of Science and Technology (INCT), Brazilian Research and Technology Council (CNPq), São Paulo, Brazil
| | - Roque Pacheco de Almeida
- Laboratory of Immunology and Molecular Biology, University Hospital, Federal University of Sergipe, Aracaju, Brazil,Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Brazil,Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil,Immunology Institute of Investigation (iii), National Institute of Science and Technology (INCT), Brazilian Research and Technology Council (CNPq), São Paulo, Brazil,*Correspondence: Roque Pacheco de Almeida,
| |
Collapse
|
42
|
Alonso L, Dorta ML, Alonso A. Ivermectin and curcumin cause plasma membrane rigidity in Leishmania amazonensis due to oxidative stress. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183977. [PMID: 35654148 DOI: 10.1016/j.bbamem.2022.183977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
Spin label electron paramagnetic resonance (EPR) spectroscopy was used to study the mechanisms of action of ivermectin and curcumin against Leishmania (L.) amazonensis promastigotes. EPR spectra showed that treatment of the parasites with both compounds results in plasma membrane rigidity due to oxidative processes. With the IC50 and EPR measurements for assays using different parasite concentrations, estimations could be made for the membrane-water partition coefficient (KM/W), and the concentration of the compound in the membrane (cm50) and in the aqueous phase (cw50), which inhibits cell growth by 50%. The KM/W values indicated that ivermectin has a greater affinity than curcumin for the parasite membrane. Therefore, the activity of ivermectin was higher for experiments with low cell concentrations, but for concentrations greater than 1.5 × 108 parasites/mL the compounds did not show significantly different results. The cm50 values indicated that the concentration of compound in the membrane leading to growth inhibition or membrane alteration is approximately 1 M for both ivermectin and curcumin. This high membrane concentration suggests that many ivermectin molecules per chlorine channel are needed to cause an increase in chlorine ion influx.
Collapse
Affiliation(s)
- Lais Alonso
- Instituto de Física, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Miriam Leandro Dorta
- Instituto de Patologia Tropical e Saúde Publica, Departamento de Imunologia e Patologia Geral, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Antonio Alonso
- Instituto de Física, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
43
|
Nettey H, Erskine IJ, Antwi Mensah A, Gyamera NKO, Obuobi CK, Kumadoh D, Asiedu-Gyekye IJ, Allotey-Babington GL. Oral amodiaquine microparticles repurposed for the treatment of visceral leishmaniasis. SCIENTIFIC AFRICAN 2022. [DOI: 10.1016/j.sciaf.2022.e01285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
44
|
Zahid F, Batool S, Ud-Din F, Ali Z, Nabi M, Khan S, Salman O, Khan GM. Antileishmanial Agents Co-loaded in Transfersomes with Enhanced Macrophage Uptake and Reduced Toxicity. AAPS PharmSciTech 2022; 23:226. [PMID: 35970966 DOI: 10.1208/s12249-022-02384-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/29/2022] [Indexed: 01/19/2023] Open
Abstract
The prime objective of this study was to develop amphotericin B (AMB) and rifampicin (RIF) co-loaded transfersomal gel (AMB-RIF co-loaded TFG) for effective treatment of cutaneous leishmaniasis (CL). AMB-RIF co-loaded TF was prepared by the thin-film hydration method and was optimized based on particle size, polydispersity index (PDI), zeta potential, entrapment efficiency (%EE), and deformability index. Similarly, AMB-RIF co-loaded TFG was characterized in terms of rheology, spread ability, and pH. In vitro, ex vivo, and in vivo assays were performed to evaluate AMB-RIF co-loaded TF as a potential treatment option for CL. The optimized formulation had vesicles in nanosize range (167 nm) with suitable PDI (0.106), zeta potential (- 19.05 mV), and excellent %EE of RIF (66%) and AMB (85%). Moreover, it had appropriate deformability index (0.952). Additionally, AMB-RIF co-loaded TFG demonstrated suitable rheological behavior for topical application. AMB-RIF co-loaded TF and AMB-RIF co-loaded TFG showed sustained release of the incorporated drugs as compared to AMB-RIF suspension. Furthermore, RIF permeation from AMB-RIF co-loaded TF and AMB-RIF co-loaded TFG was enhanced fivefold and threefold, whereas AMB permeation was enhanced by eightfold and 6.6-fold, respectively. The significantly different IC50, higher CC50, and FIC50 (p < 0.5) showed synergistic antileishmanial potential of AMB-RIF co-loaded TF. Likewise, reduced lesion size and parasitic burden in AMB-RIF co-loaded TF-treated mouse group further established the antileishmanial effect of the optimized formulation. Besides, AMB-RIF co-loaded TFG showed a better safety profile. This study concluded that TFG may be a suitable carrier for co-delivery of AMB-RIF when administered topically for the treatment of CL.
Collapse
Affiliation(s)
- Fatima Zahid
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Sibgha Batool
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Fakhar Ud-Din
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan.
| | - Zakir Ali
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Muhammad Nabi
- Institute of Pharmaceutical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Salman Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Omer Salman
- Department of Pharmacy, Forman Christian University, Lahore, Pakistan
| | - Gul Majid Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan. .,Islamia College University, Peshawar, Khyber Pakhtunkhwa, Pakistan.
| |
Collapse
|
45
|
Ibarra-Meneses AV, Corbeil A, Wagner V, Beaudry F, do Monte-Neto RL, Fernandez-Prada C. Exploring direct and indirect targets of current antileishmanial drugs using a novel thermal proteomics profiling approach. Front Cell Infect Microbiol 2022; 12:954144. [PMID: 35992178 PMCID: PMC9381709 DOI: 10.3389/fcimb.2022.954144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Visceral leishmaniasis (VL), caused by Leishmania infantum, is an oft-fatal neglected tropical disease. In the absence of an effective vaccine, the control of leishmaniasis relies exclusively on chemotherapy. Due to the lack of established molecular/genetic markers denoting parasite resistance, clinical treatment failure is often used as an indicator. Antimony-based drugs have been the standard antileishmanial treatment for more than seven decades, leading to major drug resistance in certain regions. Likewise, drug resistance to miltefosine and amphotericin B continues to spread at alarming rates. In consequence, innovative approaches are needed to accelerate the identification of antimicrobial drug targets and resistance mechanisms. To this end, we have implemented a novel approach based on thermal proteome profiling (TPP) to further characterize the mode of action of antileishmanials antimony, miltefosine and amphotericin B, as well as to better understand the mechanisms of drug resistance deployed by Leishmania. Proteins become more resistant to heat-induced denaturation when complexed with a ligand. In this way, we used multiplexed quantitative mass spectrometry-based proteomics to monitor the melting profile of thousands of expressed soluble proteins in WT, antimony-resistant, miltefosine-resistant, and amphotericin B-resistant L. infantum parasites, in the presence (or absence) of the above-mentioned drugs. Bioinformatics analyses were performed, including data normalization, melting profile fitting, and identification of proteins that underwent changes (fold change > 4) caused by complexation with a drug. With this unique approach, we were able to narrow down the regions of the L. infantum proteome that interact with antimony, miltefosine, and amphotericin B; validating previously-identified and unveiling novel drug targets. Moreover, analyses revealed candidate proteins potentially involved in drug resistance. Interestingly, we detected thermal proximity coaggregation for several proteins belonging to the same metabolic pathway (i.e., tryparedoxin peroxidase and aspartate aminotransferase in proteins exposed to antimony), highlighting the importance of these pathways. Collectively, our results could serve as a jumping-off point for the future development of innovative diagnostic tools for the detection and evaluation of antimicrobial-resistant Leishmania populations, as well as open the door for new on-target therapies.
Collapse
Affiliation(s)
- Ana Victoria Ibarra-Meneses
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Audrey Corbeil
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Victoria Wagner
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Francis Beaudry
- Département de Biomédecine, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- Centre de recherche sur le cerveau et l’apprentissage (CIRCA), Université de Montréal, Montréal, QC, Canada
| | - Rubens L. do Monte-Neto
- Biotechnology Applied to Pathogens (BAP) - Instituto René Rachou – Fundação Oswaldo Cruz/Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Christopher Fernandez-Prada
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- *Correspondence: Christopher Fernandez-Prada,
| |
Collapse
|
46
|
Kant V, Kumar P, Ranjan R, Kumar P, Mandal D, Vijayakumar S. In silico screening, molecular dynamic simulations, and in vitro activity of selected natural compounds as an inhibitor of Leishmania donovani 3-mercaptopyruvate sulfurtransferase. Parasitol Res 2022; 121:2093-2109. [PMID: 35536513 PMCID: PMC9085559 DOI: 10.1007/s00436-022-07532-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/20/2022] [Indexed: 11/26/2022]
Abstract
In Leishmania sp., the enzymes of de novo cysteine biosynthesis pathway require sulfide. Other organisms utilize sulfide through the sulfide reduction pathway, but Leishmania lacks the gene that encodes these enzymes. Hence, the major source of sulfide for Leishmania is believed to be from the action of 3-mercaptopyruvate sulfurtransferase (3MST) on 3-mercapto-pyruvate (3MP). There has been no effort reported in the past to screen inhibitors against L. donovani 3MST (Ld3MST). As a result, this study examines natural compounds that are potent against Ld3MST and validates it by in vitro activity and cytotoxicity tests. Initially, a library of ~ 5000 natural compounds was subjected to molecular docking approach for screening, and the best hit was validated using a long-term molecular dynamic simulation (MD). Among the docking results, quercetine-3-rutinoside (Rutin) was deemed the best hit. The results of the MD indicated that Rutin was highly capable of interacting with the varied active site segments, possibly blocking substrate access. Additionally, promastigotes and amastigotes were tested for Rutin activity and the IC50 was found to be 40.95 and 90.09 μM, respectively. Similarly, the cytotoxicity assay revealed that Rutin was not toxic even at a concentration of 819.00 μM to THP-1 cell lines. Additionally, the Ld3MST was cloned, purified, and evaluated for enzyme activity in the presence of Rutin. Reduction in the enzyme activity (~ 85%) was observed in the presence of ~ 40 μM Rutin. Thus, this study suggests that Rutin may act as a potent inhibitor of Ld3MST. With further in vivo investigations, Rutin could be a small molecule of choice for combating leishmaniasis.
Collapse
Affiliation(s)
- Vishnu Kant
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Pawan Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Ravi Ranjan
- Division of Bioinformatics, ICMR-Rajendra Memorial Institute of Medical Sciences, Patna, Bihar, India
| | - Prakash Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Debabrata Mandal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India.
| | - Saravanan Vijayakumar
- Division of Bioinformatics, ICMR-Rajendra Memorial Institute of Medical Sciences, Patna, Bihar, India.
| |
Collapse
|
47
|
N Cavallone I, Santos SK, Oliveira KS, D Passero LF, D Laurenti M, Jesus JA, P Marinsek G, Chucri TM, Mari RB. Histological and neuronal changes in the duodenum of hamsters infected with Leishmania (Leishmania) infantum. Exp Parasitol 2022; 239:108315. [PMID: 35780863 DOI: 10.1016/j.exppara.2022.108315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/07/2022] [Accepted: 06/20/2022] [Indexed: 11/26/2022]
Abstract
Visceral leishmaniasis is a neglected tropical disease caused by parasites belonging to the Leishmania genus that infect macrophages in different tissues such as the spleen, liver, lymph nodes, bone marrow, and intestine. Therefore, this study aimed to investigate the integrity of the intestinal tract and the nitrergic (NADPH-dp) and metabolically active (NADH-dp) myenteric neurons of the duodenum of golden hamsters infected with L. (L.) infantum. Therefore, thirty golden hamsters were divided into six groups (n = 5); three of them were infected with 2 × 107 promastigote forms of L. (L.) infantum by intraperitoneal route (Infected Group - IG) and three were inoculated with saline solution (control group - CG). After 30, 60 and 90 days post-infection (DPI) infected animals were euthanized and the liver, spleen and duodenum were collected to analyze tissue parasitism. The duodenum was processed using usual histological techniques to analyze the main changes that occurred during infection and histochemical techniques to phenotype myenteric neurons. Amastigote forms were observed in the spleen, liver, and duodenum during all experimental periods, and tissue parasitism in these organs increased significantly over time. At 30 DPI, reduction in muscle tunic, increase in the total intestinal wall and the number of goblet cells PAS+ was observed. At 60 DPI, an increase in intestinal crypts and intraepithelial lymphocytes was observed, and a reduction in intestinal villi was observed at 90 DPI, along with an increase in crypt size. Regarding neurons, an increase in the density of the NADPH-dp population was observed at 30 DPI, but at 60 and 90 DPI a significant reduction of this population was observed. In general, infection progression was observed to cause significant morphofunctional changes in the duodenum of infected hamsters.
Collapse
Affiliation(s)
- Italo N Cavallone
- Animal Morphophysiology Laboratory, Department of Biological and Environmental Sciences, São Paulo State University (UNESP), São Vicente, 11.380-97, Brazil
| | - Sarah K Santos
- Animal Morphophysiology Laboratory, Department of Biological and Environmental Sciences, São Paulo State University (UNESP), São Vicente, 11.380-97, Brazil
| | - Karine S Oliveira
- Animal Morphophysiology Laboratory, Department of Biological and Environmental Sciences, São Paulo State University (UNESP), São Vicente, 11.380-97, Brazil
| | - Luiz Felipe D Passero
- Animal Morphophysiology Laboratory, Department of Biological and Environmental Sciences, São Paulo State University (UNESP), São Vicente, 11.380-97, Brazil
| | - Márcia D Laurenti
- Laboratory of Pathology and Infectious Diseases, Department of Pathology, FMUSP, São Paulo, 01246903, Brazil
| | - Jéssica Adriana Jesus
- Laboratory of Pathology and Infectious Diseases, Department of Pathology, FMUSP, São Paulo, 01246903, Brazil
| | - Gabriela P Marinsek
- Animal Morphophysiology Laboratory, Department of Biological and Environmental Sciences, São Paulo State University (UNESP), São Vicente, 11.380-97, Brazil
| | - Thaís M Chucri
- Animal Morphophysiology Laboratory, Department of Biological and Environmental Sciences, São Paulo State University (UNESP), São Vicente, 11.380-97, Brazil
| | - Renata B Mari
- Animal Morphophysiology Laboratory, Department of Biological and Environmental Sciences, São Paulo State University (UNESP), São Vicente, 11.380-97, Brazil.
| |
Collapse
|
48
|
Mazire PH, Saha B, Roy A. Immunotherapy for visceral leishmaniasis: A trapeze of balancing counteractive forces. Int Immunopharmacol 2022; 110:108969. [PMID: 35738089 DOI: 10.1016/j.intimp.2022.108969] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022]
Abstract
The protozoan parasite Leishmania donovani, residing and replicating within the cells of the monocyte-macrophage (mono-mac) lineage, causes visceral leishmaniasis (VL) in humans. While, Leishmania infantum, is the main causative agent for zoonotic VL, where dogs are the main reservoirs of the disease. The chemotherapy is a serious problem because of restricted repertoire of drugs, drug-resistant parasites, drug-toxicity and the requirement for parenteral administration, which is a problem in resource-starved countries. Moreover, immunocompromised individuals, particularly HIV-1 infected are at higher risk of VL due to impairment in T-helper cell and regulatory cell responses. Furthermore, HIV-VL co-infected patients report poor response to conventional chemotherapy. Recent efforts are therefore directed towards devising both prophylactic and therapeutic immunomodulation. As far as prophylaxis is concerned, although canine vaccines for the disease caused by Leishmania infantum or Leishmania chagasi are available, no vaccine is available for use in humans till date. Therefore, anti-leishmanial immunotherapy triggering or manipulating the host's immune response is gaining momentum during the last two decades. Immunomodulators comprised of small molecules, anti-leishmanial peptides, complex ligands for host receptors, cytokines or their agonists and antibodies have been given trials both in experimental models and in humans. However, the success of immunotherapy in humans remains a far-off target. We, therefore, propose that devising a successful immunotherapy is an act of balancing enhanced beneficial Leishmania-specific responses and deleterious immune activation/hyperinflammation just as the swings in a trapeze.
Collapse
Affiliation(s)
- Priyanka H Mazire
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind Road, Pune 411007, India
| | - Amit Roy
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India.
| |
Collapse
|
49
|
Machado AS, Lage DP, Vale DL, Freitas CS, Linhares FP, Cardoso JM, Pereira IA, Ramos FF, Tavares GS, Ludolf F, Oliveira-da-Silva JA, Bandeira RS, Simões AC, Duarte MC, Oliveira JS, Christodoulides M, Chávez-Fumagalli MA, Roatt BM, Martins VT, Coelho EA. A recombinant Leishmania amastigote-specific protein, rLiHyG, with adjuvants, protects against infection with Leishmania infantum. Acta Trop 2022; 230:106412. [PMID: 35305943 DOI: 10.1016/j.actatropica.2022.106412] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/11/2022]
Abstract
Vaccination against visceral leishmaniasis (VL) should be considered as a control measure to protect against disease, and amastigote-specific proteins could help to develop such vaccines, since this parasite form is in contact with the host immune system during the active disease. In this study, a Leishmania amastigote-specific protein, LiHyG, was evaluated as recombinant protein (rLiHyG) as vaccine candidate against Leishmania infantum infection in BALB/c mice. The protein was associated with saponin (rLiHyG/Sap) or Poloxamer 407-based polymeric micelles (rLiHyG/Mic) as adjuvants, and animals receiving saline, saponin or micelle as controls. Immunological and parasitological analyses were performed before (n = 8 per group; as primary endpoint) and after (n = 8 per group; as secondary endpoint) infection. Results showed that, in both endpoints, rLiHyG/Sap and rLiHyG/Mic induced higher levels of IFN-γ, IL-12 and GM-CSF in spleen cell cultures from vaccinated animals, besides elevated presence of IgG2a isotype antibodies. Decreased hepatotoxicity and 'positive lymphoproliferative response were also found after challenge. Such findings reflected in significantly lower levels of parasite load found in their spleens, livers, bone marrows and draining lymph nodes. In conclusion, rLiHyG associated with Th1-type adjuvant could be considered for future studies as vaccine candidate to protect against VL.
Collapse
|
50
|
Bajwa HUR, Khan MK, Abbas Z, Riaz R, Rehman TU, Abbas RZ, Aleem MT, Abbas A, Almutairi MM, Alshammari FA, Alraey Y, Alouffi A. Nanoparticles: Synthesis and Their Role as Potential Drug Candidates for the Treatment of Parasitic Diseases. Life (Basel) 2022; 12:life12050750. [PMID: 35629416 PMCID: PMC9145985 DOI: 10.3390/life12050750] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/15/2022] Open
Abstract
Protozoa, helminths and ectoparasites are the major groups of parasites distributed worldwide. Currently, these parasites are treated with chemotherapeutic antiprotozoal drugs, anti-helminthic and anti-ectoparasitic agents, but, with the passage of time, resistance to these drugs has developed due to overuse. In this scenario, nanoparticles are proving to be a major breakthrough in the treatment and control of parasitic diseases. In the last decade, there has been enormous development in the field of nanomedicine for parasitic control. Gold and silver nanoparticles have shown promising results in the treatments of various types of parasitic infections. These nanoparticles are synthesized through the use of various conventional and molecular technologies and have shown great efficacy. They work in different ways, that include damaging the parasite membrane, DNA (Deoxyribonucleic acid) disruption, protein synthesis inhibition and free-radical formation. These agents are effective against intracellular parasites as well. Other nanoparticles, such as iron, nickel, zinc and platinum, have also shown good results in the treatment and control of parasitic infections. It is hoped that this research subject will become the future of modern drug development. This review summarizes the methods that are used to synthesize nanoparticles and their possible mechanisms of action against parasites.
Collapse
Affiliation(s)
| | - Muhammad Kasib Khan
- Department of Parasitology, University of Agriculture, Faisalabad 38040, Pakistan; (M.K.K.); (Z.A.); (R.Z.A.)
| | - Zaheer Abbas
- Department of Parasitology, University of Agriculture, Faisalabad 38040, Pakistan; (M.K.K.); (Z.A.); (R.Z.A.)
| | - Roshan Riaz
- Department of Animal Nutrition and Nutritional Diseases, Ankara University, Ankara 06100, Turkey;
| | - Tauseef ur Rehman
- Department of Parasitology, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- Correspondence: (T.u.R.); (A.A.)
| | - Rao Zahid Abbas
- Department of Parasitology, University of Agriculture, Faisalabad 38040, Pakistan; (M.K.K.); (Z.A.); (R.Z.A.)
| | - Muhammad Tahir Aleem
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China;
| | - Asghar Abbas
- Faculty of Veterinary and Animal Sciences, MNS-University of Agriculture Multan, Multan 60650, Pakistan;
| | - Mashal M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Fahdah Ayed Alshammari
- College of Sciences and Literature Microbiology, Arar Northern Border University, Arar 73211, Saudi Arabia;
| | - Yasser Alraey
- Department of Clinical Laboratory Sciences, Central Research Laboratory, College of Applied Medical Sciences, King Khalid University, Abha 62217, Saudi Arabia;
| | - Abdulaziz Alouffi
- King Abdulaziz City for Science and Technology, Riyadh 12354, Saudi Arabia
- Correspondence: (T.u.R.); (A.A.)
| |
Collapse
|