1
|
Goyzueta-Mamani LD, Pagliara Lage D, Barazorda-Ccahuana HL, Paco-Chipana M, Candia-Puma MA, Davila-Del-Carpio G, Galdino AS, Machado-de-Avila RA, Cordeiro Giunchetti R, D’Antonio EL, Ferraz Coelho EA, Chávez-Fumagalli MA. Exploring the Potential of Malvidin and Echiodinin as Probable Antileishmanial Agents Through In Silico Analysis and In Vitro Efficacy. Molecules 2025; 30:173. [PMID: 39795229 PMCID: PMC11722285 DOI: 10.3390/molecules30010173] [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: 12/02/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Leishmaniasis, a neglected tropical disease caused by Leishmania species, presents serious public health challenges due to limited treatment options, toxicity, high costs, and drug resistance. In this study, the in vitro potential of malvidin and echioidinin is examined as antileishmanial agents against L. amazonensis, L. braziliensis, and L. infantum, comparing their effects to amphotericin B (AmpB), a standard drug. Malvidin demonstrated greater potency than echioidinin across all parasite stages and species. Against L. amazonensis, malvidin's IC50 values were 197.71 ± 17.20 µM (stationary amastigotes) and 258.07 ± 17 µM (axenic amastigotes), compared to echioidinin's 272.99 ± 29.90 μM and 335.96 ± 19.35 μM. AmpB was more potent, with IC50 values of 0.06 ± 0.01 µM and 0.10 ± 0.03 µM. Malvidin exhibited lower cytotoxicity (CC50: 2920.31 ± 80.29 µM) than AmpB (1.06 ± 0.12 µM) and a favorable selectivity index. It reduced infection rates by 35.75% in L. amazonensis-infected macrophages. The in silico analysis revealed strong binding between malvidin and Leishmania arginase, with the residues HIS139 and PRO258 playing key roles. Gene expression analysis indicated malvidin's modulation of oxidative stress and DNA repair pathways, involving genes like GLO1 and APEX1. These findings suggest malvidin's potential as a safe, natural antileishmanial compound, warranting further in vivo studies to confirm its therapeutic efficacy and pharmacokinetics in animal models.
Collapse
Affiliation(s)
- Luis Daniel Goyzueta-Mamani
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| | - 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, Brazil; (D.P.L.); (E.A.F.C.)
| | - Haruna Luz Barazorda-Ccahuana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| | - Margot Paco-Chipana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| | - Mayron Antonio Candia-Puma
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Arequipa 04000, Peru;
| | - Gonzalo Davila-Del-Carpio
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Arequipa 04000, Peru;
| | - Alexsandro Sobreira Galdino
- Laboratório de Biotecnologia de Microrganismos, Universidade Federal São João Del-Rei, Divinópolis 35501-296, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biotecnologia Industrial (INCT-BI), Distrito Federal, Brasilia 70070-010, Brazil
| | | | - Rodolfo Cordeiro Giunchetti
- Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais (INCT-DT), Salvador 40110-160, Brazil
| | - Edward L. D’Antonio
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA;
| | - 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, Brazil; (D.P.L.); (E.A.F.C.)
| | - Miguel Angel Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| |
Collapse
|
2
|
Gupta D, Singh PK, Yadav PK, Narender T, Patil UK, Jain SK, Chourasia MK. Emerging strategies and challenges of molecular therapeutics in antileishmanial drug development. Int Immunopharmacol 2023; 115:109649. [PMID: 36603357 DOI: 10.1016/j.intimp.2022.109649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/16/2022] [Accepted: 12/24/2022] [Indexed: 01/05/2023]
Abstract
Molecular therapy refers to targeted therapies based on molecules which have been intelligently directed towards specific biomolecular structures and include small molecule drugs, monoclonal antibodies, proteins and peptides, DNA or RNA-based strategies, targeted chemotherapy and nanomedicines. Molecular therapy is emerging as the most effective strategy to combat the present challenges of life-threatening visceral leishmaniasis, where the successful human vaccine is currently unavailable. Moreover, current chemotherapy-based strategies are associated with the issues of ineffective targeting, unavoidable toxicities, invasive therapies, prolonged treatment, high treatment costs and the development of drug-resistant strains. Thus, the rational approach to antileishmanial drug development primarily demands critical exploration and exploitation of biochemical differences between host and parasite biology, immunocharacteristics of parasite homing, and host-parasite interactions at the molecular/cellular level. Following this, the novel technology-based designing and development of host and/or parasite-targeted therapeutics having leishmanicidal and immunomodulatory activity is utmost essential to improve treatment efficacy. Thus, the present review is focused on immunological and molecular checkpoint targets in host-pathogen interaction, and molecular therapeutic prospects for Leishmania intervention, and the challenges ahead.
Collapse
Affiliation(s)
- Deepak Gupta
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Pankaj K Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Tadigoppula Narender
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Umesh K Patil
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India.
| |
Collapse
|
3
|
Saraei S, Soozangar N, Miran M, Ghaffarifar F, Mohammadi-Ghalehbin B, Molaei S, Asfaram S. In vitro Evaluation of the Potent Antileishmanial Activity of Ferula tabasensis Alone or in Combination with Shark Cartilage Extract Against the Standard Iranian Strain of Leishmania major (MRHO/IR/75/ER). IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2023; 22:e136173. [PMID: 38116562 PMCID: PMC10728847 DOI: 10.5812/ijpr-136173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/28/2023] [Accepted: 06/06/2023] [Indexed: 12/21/2023]
Abstract
Background The available drugs for the treatment of leishmaniasis are highly toxic and extremely expensive, with low efficiency; therefore, the development of effective therapeutic compounds is essential. Objectives The present study aimed to explore the antileishmanial effects of ethyl acetate extract, methanol extract, and fractions 1-4 (F1-F4) of Ferula tabasensis, alone or in combination with shark cartilage extract (ShCE), on L. major in vitro. Methods In this study, ethyl acetate, methanol, and n-hexane extracts were extracted from the aerial roots of F. tabasensis by the maceration method. The silica gel column chromatography was used to separate n-hexane extracts at varying polarities (F1-F4 fractions). Subsequently, the effects of extracts and fractions against promastigotes were assessed by the parasite counting method microscopic inhibition test and MTT assay. Besides, their effects on the infected macrophage cells and the number of amastigotes were investigated. Cytotoxicity was evaluated in non-infected J774A.1 macrophage cells. Finally, apoptosis induction of promastigotes, including infected and non-infected macrophages, was evaluated. Results The results indicated the highly potent activity of F. tabasensis extracts and F1-F4 fractions, alone or in combination with ShCE, against L. major promastigotes and amastigotes in a dose-dependent manner (P < 0.05). The F1 fraction and methanol extract showed markedly higher toxicity compared to the other extracts and fractions, with 50% inhibitory concentrations (IC50/72h) of 2.4 ± 0.29 and 2.9 ± 0.55 µg/mL against promastigotes and 1.79 ± 0.27 µg/mL and 1.39 ± 0.27 µg/mL against amastigotes (P < 0.001). Moreover, they had a high selectivity index (SI) due to the low toxicity of macrophages (P < 0.0001). The results of flow cytometry indicated that the percentages of apoptotic promastigote cells in contact with IC50 concentrations of F1 and methanol extract alone after 72 h were 43.83 and 43.93%, as well as 78.4%, and 65.45% for their combination with ShCE, respectively.Also, apoptosis of infected macrophages induced by F1 and methanol extracts was estimated at 68.5% and 83.7%, respectively. Conclusions In this study, the F1 fraction and methanol extract of F. tabasensis showed potent efficacy against L. major, associated with low toxicity and apoptosis induction. Therefore, they can be promising therapeutic candidates in future animal and even human studies.
Collapse
Affiliation(s)
- Shaylin Saraei
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Narges Soozangar
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mansour Miran
- Department of Pharmacognosy and Biotechnology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Soheila Molaei
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shabnam Asfaram
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
4
|
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
|
5
|
Identification of L. infantum trypanothione synthetase inhibitors with leishmanicidal activity from a (non-biased) in-house chemical library. Eur J Med Chem 2022; 243:114675. [DOI: 10.1016/j.ejmech.2022.114675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022]
|
6
|
Ganguly S, Ghoshal B, Banerji I, Bhattacharjee S, Chakraborty S, Goswami A, Mukherjee K, Bhattacharyya SN. Leishmania survives by exporting miR-146a from infected to resident cells to subjugate inflammation. Life Sci Alliance 2022; 5:5/6/e202101229. [PMID: 35210329 PMCID: PMC8881743 DOI: 10.26508/lsa.202101229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/24/2022] Open
Abstract
Leishmania donovani, the causative agent of visceral leishmaniasis, infects and resides within tissue macrophage cells. It is not clear how the parasite infected cells crosstalk with the noninfected cells to regulate the infection process. During infection, Leishmania adopts a dual strategy for its survival by regulating the intercellular transport of host miRNAs to restrict inflammation. The parasite, by preventing mitochondrial function of host cells, restricts the entry of liver cell derived miR-122-containing extracellular vesicles in infected macrophages to curtail the inflammatory response associated with miR-122 entry. On contrary, the parasite up-regulates the export of miR-146a from the infected macrophages. The miR-146a, associated with the extracellular vesicles released by infected cells, restricts miR-122 production in hepatocytes while polarizing neighbouring naïve macrophages to the M2 state by affecting the cytokine expression. On entering the recipient macrophages, miR-146a dominates the miRNA antagonist RNA-binding protein HuR to inhibit the expression of proinflammatory cytokine mRNAs having HuR-interacting AU-rich elements whereas up-regulates anti-inflammatory IL-10 by exporting the miR-21 to polarize the recipient cells to M2 stage.
Collapse
Affiliation(s)
- Satarupa Ganguly
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Bartika Ghoshal
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Ishani Banerji
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Ghaziabad, India
| | - Shreya Bhattacharjee
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Ghaziabad, India
| | - Sreemoyee Chakraborty
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Ghaziabad, India
| | - Avijit Goswami
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Kamalika Mukherjee
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Suvendra N Bhattacharyya
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India .,Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Ghaziabad, India
| |
Collapse
|
7
|
Kumari D, Singh K. Exploring the paradox of defense between host and Leishmania parasite. Int Immunopharmacol 2021; 102:108400. [PMID: 34890999 DOI: 10.1016/j.intimp.2021.108400] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 01/04/2023]
Abstract
Leishmaniasis, a neglected tropical disease, still remains a global concern for the healthcare sector. The primary causative agents of the disease comprise diverse leishmanial species, leading to recurring failures in disease diagnosis and delaying the initiation of appropriate chemotherapy. Various species of the Leishmania parasite cause diverse clinical manifestations ranging from skin ulcers to systemic infections. Therefore, host immunity in response to different forms of infecting species of Leishmania becomes pivotal in disease progression or regression. Thus, understanding the paradox of immune arsenals during host and parasite interface becomes crucial to eliminate this deadly disease. In the present review, we have elaborated on the immunological perspectives of the disease and discussed primary host immune cells that form a defense line to counteract parasite infection. Furthermore, we also have shed light on the immune cells and effector molecules responsible for parasite survival in host lethal milieu/ environment. Next, we have highlighted recent molecules/compounds showing potent leishmanicidal activities pertaining to their pro-oxidant and immuno-modulatory mechanisms. This review addresses an immuno-biological overview of the factors influencing the parasitic disease, as this knowledge can aid in the unraveling/ identification of potential biomarkers, novel therapeutics, and vaccine candidates against leishmaniasis.
Collapse
Affiliation(s)
- Diksha Kumari
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
8
|
Kolářová I, Valigurová A. Hide-and-Seek: A Game Played between Parasitic Protists and Their Hosts. Microorganisms 2021; 9:2434. [PMID: 34946036 PMCID: PMC8707157 DOI: 10.3390/microorganisms9122434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022] Open
Abstract
After invading the host organism, a battle occurs between the parasitic protists and the host's immune system, the result of which determines not only whether and how well the host survives and recovers, but also the fate of the parasite itself. The exact weaponry of this battle depends, among others, on the parasite localisation. While some parasitic protists do not invade the host cell at all (extracellular parasites), others have developed successful intracellular lifestyles (intracellular parasites) or attack only the surface of the host cell (epicellular parasites). Epicellular and intracellular protist parasites have developed various mechanisms to hijack host cell functions to escape cellular defences and immune responses, and, finally, to gain access to host nutrients. They use various evasion tactics to secure the tight contact with the host cell and the direct nutrient supply. This review focuses on the adaptations and evasion strategies of parasitic protists on the example of two very successful parasites of medical significance, Cryptosporidium and Leishmania, while discussing different localisation (epicellular vs. intracellular) with respect to the host cell.
Collapse
Affiliation(s)
- Iva Kolářová
- Laboratory of Vector Biology, Department of Parasitology, Faculty of Science, Charles University, Albertov 6, 128 44 Prague, Czech Republic
| | - Andrea Valigurová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic
| |
Collapse
|
9
|
Richter R, Lehr CM. Extracellular vesicles as novel assay tools to study cellular interactions of anti-infective compounds - A perspective. Adv Drug Deliv Rev 2021; 173:492-503. [PMID: 33857554 DOI: 10.1016/j.addr.2021.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/21/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022]
Abstract
Sudden outbreaks of novel infectious diseases and the persistent evolution of antimicrobial resistant pathogens make it necessary to develop specific tools to quickly understand pathogen-cell interactions and to study appropriate drug delivery strategies. Extracellular vesicles (EVs) are cell-specific biogenic transport systems, which are gaining more and more popularity as either diagnostic markers or drug delivery systems. Apart from that, there are emerging possibilities for EVs as tools to study drug penetration, drug-membrane interactions as well as pathogen-membrane interactions. However, it appears that the potential of EVs for such applications has not been fully exploited yet. Considering the vast variety of cells that can be involved in an infection, vesicle-based analytical methods are just emerging and the number of reported applications is still relatively small. Aim of this review is to discuss the current state of the art of EV-based assays, especially in the context of antimicrobial research and therapy, and to present some new perspectives for a more exhaustive and creative exploration in the future.
Collapse
Affiliation(s)
- Robert Richter
- Department of Drug Delivery Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz-Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz-Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany.
| |
Collapse
|
10
|
Zayats R, Uzonna JE, Murooka TT. Visualizing the In Vivo Dynamics of Anti- Leishmania Immunity: Discoveries and Challenges. Front Immunol 2021; 12:671582. [PMID: 34093571 PMCID: PMC8172142 DOI: 10.3389/fimmu.2021.671582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/07/2021] [Indexed: 11/20/2022] Open
Abstract
Intravital microscopy, such as 2-photon microscopy, is now a mainstay in immunological research to visually characterize immune cell dynamics during homeostasis and pathogen infections. This approach has been especially beneficial in describing the complex process of host immune responses to parasitic infections in vivo, such as Leishmania. Human-parasite co-evolution has endowed parasites with multiple strategies to subvert host immunity in order to establish chronic infections and ensure human-to-human transmission. While much focus has been placed on viral and bacterial infections, intravital microscopy studies during parasitic infections have been comparatively sparse. In this review, we will discuss how in vivo microscopy has provided important insights into the generation of innate and adaptive immunity in various organs during parasitic infections, with a primary focus on Leishmania. We highlight how microscopy-based approaches may be key to providing mechanistic insights into Leishmania persistence in vivo and to devise strategies for better parasite control.
Collapse
Affiliation(s)
- Romaniya Zayats
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Jude E. Uzonna
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Thomas T. Murooka
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
11
|
Ftouh S, Bourgeade-Delmas S, Belkadi M, Deraeve C, Hemmert C, Valentin A, Gornitzka H. Synthesis, Characterization, and Antileishmanial Activity of Neutral Gold(I) Complexes with N-heterocyclic Carbene Ligands Bearing Sulfur-Containing Side Arms. Organometallics 2021. [DOI: 10.1021/acs.organomet.1c00113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Soumia Ftouh
- LCC-CNRS, Université de Toulouse, CNRS, UPS, Toulouse 31400, France
- (LSPBE), Département de Génie Chimique, Faculté de Chimie, Université des Sciences et de la Technologie d’Oran Mohamed Boudiaf, USTO-MB,
B.P 1505, El Mnaouer, Oran 31000, Algeria
| | | | - Mohamed Belkadi
- (LSPBE), Département de Génie Chimique, Faculté de Chimie, Université des Sciences et de la Technologie d’Oran Mohamed Boudiaf, USTO-MB,
B.P 1505, El Mnaouer, Oran 31000, Algeria
| | - Céline Deraeve
- LCC-CNRS, Université de Toulouse, CNRS, UPS, Toulouse 31400, France
| | | | - Alexis Valentin
- UMR 152 PharmaDev, Université de Toulouse, IRD, UPS, Toulouse 31400, France
| | - Heinz Gornitzka
- LCC-CNRS, Université de Toulouse, CNRS, UPS, Toulouse 31400, France
| |
Collapse
|
12
|
Mokdadi M, Abdelkrim YZ, Banroques J, Huvelle E, Oualha R, Yeter-Alat H, Guizani I, Barhoumi M, Tanner NK. The In Silico Identification of Potential Members of the Ded1/DDX3 Subfamily of DEAD-Box RNA Helicases from the Protozoan Parasite Leishmania infantum and Their Analyses in Yeast. Genes (Basel) 2021; 12:212. [PMID: 33535521 PMCID: PMC7912733 DOI: 10.3390/genes12020212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
DEAD-box RNA helicases are ubiquitous proteins found in all kingdoms of life and that are associated with all processes involving RNA. Their central roles in biology make these proteins potential targets for therapeutic or prophylactic drugs. The Ded1/DDX3 subfamily of DEAD-box proteins is of particular interest because of their important role(s) in translation. In this paper, we identified and aligned the protein sequences of 28 different DEAD-box proteins from the kinetoplast-protozoan parasite Leishmania infantum, which is the cause of the visceral form of leishmaniasis that is often lethal if left untreated, and compared them with the consensus sequence derived from DEAD-box proteins in general, and from the Ded1/DDX3 subfamily in particular, from a wide variety of other organisms. We identified three potential homologs of the Ded1/DDX3 subfamily and the equivalent proteins from the related protozoan parasite Trypanosoma brucei, which is the causative agent of sleeping sickness. We subsequently tested these proteins for their ability to complement a yeast strain deleted for the essential DED1 gene. We found that the DEAD-box proteins from Trypanosomatids are highly divergent from other eukaryotes, and consequently they are suitable targets for protein-specific drugs.
Collapse
Affiliation(s)
- Molka Mokdadi
- Expression Génétique Microbienne, UMR8261 CNRS, Université de Paris, Institut de Biologie Physico-Chimique, 13 rue Pierre et Marie Curie, 75005 Paris, France; (M.M.); (Y.Z.A.); (J.B.); (E.H.); (H.Y.-A.)
- PSL Research University, 75005 Paris, France
- Laboratory of Molecular Epidemiology and Experimental Pathology (LR16IPT04), Institut Pasteur de Tunis, Université de Tunis El Manar, 13 Place Pasteur, BP74 Tunis-Belvédère 1002, Tunisia; (R.O.); (I.G.)
- Institut National des Sciences Appliquées et Technologies, Université de Carthage, CEDEX, Tunis 1080, Tunisia
| | - Yosser Zina Abdelkrim
- Expression Génétique Microbienne, UMR8261 CNRS, Université de Paris, Institut de Biologie Physico-Chimique, 13 rue Pierre et Marie Curie, 75005 Paris, France; (M.M.); (Y.Z.A.); (J.B.); (E.H.); (H.Y.-A.)
- Laboratory of Molecular Epidemiology and Experimental Pathology (LR16IPT04), Institut Pasteur de Tunis, Université de Tunis El Manar, 13 Place Pasteur, BP74 Tunis-Belvédère 1002, Tunisia; (R.O.); (I.G.)
| | - Josette Banroques
- Expression Génétique Microbienne, UMR8261 CNRS, Université de Paris, Institut de Biologie Physico-Chimique, 13 rue Pierre et Marie Curie, 75005 Paris, France; (M.M.); (Y.Z.A.); (J.B.); (E.H.); (H.Y.-A.)
- PSL Research University, 75005 Paris, France
| | - Emmeline Huvelle
- Expression Génétique Microbienne, UMR8261 CNRS, Université de Paris, Institut de Biologie Physico-Chimique, 13 rue Pierre et Marie Curie, 75005 Paris, France; (M.M.); (Y.Z.A.); (J.B.); (E.H.); (H.Y.-A.)
- PSL Research University, 75005 Paris, France
| | - Rafeh Oualha
- Laboratory of Molecular Epidemiology and Experimental Pathology (LR16IPT04), Institut Pasteur de Tunis, Université de Tunis El Manar, 13 Place Pasteur, BP74 Tunis-Belvédère 1002, Tunisia; (R.O.); (I.G.)
| | - Hilal Yeter-Alat
- Expression Génétique Microbienne, UMR8261 CNRS, Université de Paris, Institut de Biologie Physico-Chimique, 13 rue Pierre et Marie Curie, 75005 Paris, France; (M.M.); (Y.Z.A.); (J.B.); (E.H.); (H.Y.-A.)
- PSL Research University, 75005 Paris, France
| | - Ikram Guizani
- Laboratory of Molecular Epidemiology and Experimental Pathology (LR16IPT04), Institut Pasteur de Tunis, Université de Tunis El Manar, 13 Place Pasteur, BP74 Tunis-Belvédère 1002, Tunisia; (R.O.); (I.G.)
| | - Mourad Barhoumi
- Laboratory of Molecular Epidemiology and Experimental Pathology (LR16IPT04), Institut Pasteur de Tunis, Université de Tunis El Manar, 13 Place Pasteur, BP74 Tunis-Belvédère 1002, Tunisia; (R.O.); (I.G.)
| | - N. Kyle Tanner
- Expression Génétique Microbienne, UMR8261 CNRS, Université de Paris, Institut de Biologie Physico-Chimique, 13 rue Pierre et Marie Curie, 75005 Paris, France; (M.M.); (Y.Z.A.); (J.B.); (E.H.); (H.Y.-A.)
- PSL Research University, 75005 Paris, France
| |
Collapse
|
13
|
Heat Shock Proteins as the Druggable Targets in Leishmaniasis: Promises and Perils. Infect Immun 2021; 89:IAI.00559-20. [PMID: 33139381 DOI: 10.1128/iai.00559-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Leishmania, the causative agent of leishmaniasis, is an intracellular pathogen that thrives in the insect gut and mammalian macrophages to complete its life cycle. Apart from temperature difference (26 to 37°C), it encounters several harsh conditions, including oxidative stress, inflammatory reactions, and low pH. Heat shock proteins (HSPs) play essential roles in cell survival by strategically reprogramming cellular processes and signaling pathways. HSPs assist cells in multiple functions, including differentiation, adaptation, virulence, and persistence in the host cell. Due to cyclical epidemiological patterns, limited chemotherapeutic options, drug resistance, and the absence of a vaccine, control of leishmaniasis remains a far-fetched dream. The essential roles of HSPs in parasitic differentiation and virulence and increased expression in drug-resistant strains highlight their importance in combating the disease. In this review, we highlighted the diverse physiological importance of HSPs present in Leishmania, emphasizing their significance in disease pathogenesis. Subsequently, we assessed the potential of HSPs as a chemotherapeutic target and underlined the challenges associated with it. Furthermore, we have summarized a few ongoing drug discovery initiatives that need to be explored further to develop clinically successful chemotherapeutic agents in the future.
Collapse
|
14
|
Campelo CM, Pinheiro IC, de Melo Tavares B, Alves de Lima Henn G, Fernandes C, Albuquerque-Pinto LC, Carneiro Câmara LM. Modulation in the expression of type 1 (CR1/CD35) and type 3 (CR3/CD11b) complement receptors on leukocytes from patients with Visceral leishmaniasis. Exp Parasitol 2020; 218:107970. [PMID: 32828829 DOI: 10.1016/j.exppara.2020.107970] [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: 12/13/2019] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 10/23/2022]
Abstract
Visceral leishmaniasis (VL) is an anthropozoonosis endemic in Brazil. We included 20 patients with confirmed diagnosis of VL and 20 healthy individuals to evaluate the expression levels of complement receptor 1 (CR1)/CD35 and CR3/CD11b on leukocytes in the peripheral blood and determine their correlation with the clinical state of patients. CR1/CD35 expression increased on CD11b+CD35+granulocytes of patients, while CR1/CD35 and CR3/CD11b expression levels increased on CD14+CD11b+CD35+ monocytes. Among patients, those with severe clinical state had higher expression of CR3/CD11b on CD14+monocytes. The count of CD19+CD35+B lymphocytes reduced in the blood samples from patients. These observed changes may indicate the modulation in CR1/CD35 and CR3/CD11b complement receptor expressionlevels on granulocyte and monocyte populations in response to Leishmania sp.
Collapse
Affiliation(s)
- Cássio Marinho Campelo
- Federal University of Ceará, Biomedicine Center, Laboratory of Medical Immunology, Fortaleza, Ceará, Brazil.
| | | | | | - Guilherme Alves de Lima Henn
- Federal University of Ceará, Biomedicine Center, Laboratory of Medical Immunology, Fortaleza, Ceará, Brazil; Hospital São José for Infectious Diseases(HSJ), Fortaleza, Ceará, Brazil.
| | - Camila Fernandes
- Federal University of Ceará, Biomedicine Center, Laboratory of Medical Immunology, Fortaleza, Ceará, Brazil.
| | | | - Lilia Maria Carneiro Câmara
- Federal University of Ceará, Biomedicine Center, Laboratory of Medical Immunology, Fortaleza, Ceará, Brazil.
| |
Collapse
|
15
|
Histone deacetylases inhibitors as new potential drugs against Leishmania braziliensis, the main causative agent of new world tegumentary leishmaniasis. Biochem Pharmacol 2020; 180:114191. [PMID: 32777278 DOI: 10.1016/j.bcp.2020.114191] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022]
Abstract
The protozoan parasite Leishmania braziliensis is a major causative agent of the neglected tropical diseases Cutaneous and Mucocutaneous Leishmaniases in the New World. There are no vaccines to prevent the infection and the treatment relies on few drugs that often display high toxicity and costs. Thus, chemotherapeutic alternatives are required. Histone Deacetylases (HDACs) are epigenetic enzymes involved in the control of chromatin structure. In this work, we tested an in-house library of 78 hydroxamic acid derivatives as putative inhibitors of L. braziliensis HDACs (HDACi). The compounds were evaluated in relation to the toxicity to the host cell macrophage and to the leishmanicidal effect against L. braziliensis during in vitro infection. Eight HDACi showed significant leishmanicidal effects and the top 5 compounds showed effective concentrations (EC50) in the range of 4.38 to 10.21 μM and selectivity indexes (SI) from of 6 to 21.7. Analyses by Transmission Electron Microscopy (TEM) indicated induction of apoptotic cell death of L. braziliensis amastigotes with a necrotic phenotype. An altered chromatin condensation pattern and cellular disorganization of intracellular amastigotes was also observed. A tight connection between the mitochondrion and nuclear protrusions, presumably of endoplasmic reticulum origin, was found in parasites but not in the host cell. In flow cytometry (FC) analyses, HDACi promoted parasite cell cycle arrest in the G2-M phase and no changes were found in macrophages. In addition, the direct effect of HDACi against the promastigotes showed apoptosis as the main mechanism of cell death. The FC results corroborate the TEM analyses indicating that the HDACi lead to changes in the cell cycle and induction of apoptosis of L. braziliensis. The production of nitric oxide by the infected macrophages was not altered after treatment with the top 5 compounds. Taken together, our results evidenced new HDACi as promising agents for the development of new treatments for American Tegumentary Leishmaniasis caused by L. braziliensis.
Collapse
|
16
|
Hurdayal R, Nieuwenhuizen NE, Khutlang R, Brombacher F. Inflammatory Dendritic Cells, Regulated by IL-4 Receptor Alpha Signaling, Control Replication, and Dissemination of Leishmania major in Mice. Front Cell Infect Microbiol 2020; 9:479. [PMID: 32039054 PMCID: PMC6992597 DOI: 10.3389/fcimb.2019.00479] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/27/2019] [Indexed: 11/16/2022] Open
Abstract
Leishmaniasis is a vector-borne disease caused by Leishmania parasites. Macrophages are considered the primary parasite host cell, but dendritic cells (DCs) play a critical role in initiating adaptive immunity and controlling Leishmania infection. Accordingly, our previous study in CD11ccreIL-4Rα−/lox mice, which have impaired IL-4 receptor alpha (IL-4Rα) expression on CD11c+ cells including DCs, confirmed a protective role for IL-4/IL-13-responsive DCs in replication and dissemination of parasites during cutaneous leishmaniasis. However, it was unclear which DC subset/s was executing this function. To investigate this, we infected CD11ccreIL-4Rα−/lox and control mice with L. major GFP+ parasites and identified subsets of infected DCs by flow cytometry. Three days after infection, CD11b+ DCs and CD103+ DCs were the main infected DC subsets in the footpad and draining lymph node, respectively and by 4 weeks post-infection, Ly6C+ and Ly6C− CD11b+ DCs were the main infected DC populations in both the lymph nodes and footpads. Interestingly, Ly6C+CD11b+ inflammatory monocyte-derived DCs but not Ly6C−CD11b+ DCs hosted parasites in the spleen. Importantly, intracellular parasitism was significantly higher in IL-4Rα-deficient DCs. In terms of DC effector function, we found no change in the expression of pattern-recognition receptors (TLR4 and TLR9) nor in expression of the co-stimulatory marker, CD80, but MHCII expression was lower in CD11ccreIL-4Rα−/lox mice at later time-points compared to the controls. Interestingly, in CD11ccreIL-4Rα−/lox mice, which have reduced Th1 responses, CD11b+ DCs had impaired iNOS production, suggesting that DC IL-4Rα expression and NO production is important for controlling parasite numbers and preventing dissemination. Expression of the alternative activation marker arginase was unchanged in CD11b+ DCs in CD11creIL-4Rα−/lox mice compared to littermate controls, but RELM-α was upregulated, suggesting IL-4Rα-independent alternative activation. In summary, L. major parasites may use Ly6C+CD11b+ inflammatory DCs derived from monocytes recruited to infection as “Trojan horses” to migrate to secondary lymphoid organs and peripheral sites, and DC IL-4Rα expression is important for controlling infection.
Collapse
Affiliation(s)
- Ramona Hurdayal
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Natalie Eva Nieuwenhuizen
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Rethabile Khutlang
- Identity Authentication Research Group, Defence and Security, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
17
|
Tracking of quiescence in Leishmania by quantifying the expression of GFP in the ribosomal DNA locus. Sci Rep 2019; 9:18951. [PMID: 31831818 PMCID: PMC6908629 DOI: 10.1038/s41598-019-55486-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/27/2019] [Indexed: 01/22/2023] Open
Abstract
Under stressful conditions some microorganisms adopt a quiescent stage characterized by a reversible non or slow proliferative condition that allows their survival. This adaptation was only recently discovered in Leishmania. We developed an in vitro model and a biosensor to track quiescence at population and single cell levels. The biosensor is a GFP reporter gene integrated within the 18S rDNA locus, which allows monitoring the expression of 18S rRNA (rGFP expression). We showed that rGFP expression decreased significantly and rapidly during the transition from extracellular promastigotes to intracellular amastigotes and that it was coupled in vitro with a decrease in replication as measured by BrdU incorporation. rGFP expression was useful to track the reversibility of quiescence in live cells and showed for the first time the heterogeneity of physiological stages among the population of amastigotes in which shallow and deep quiescent stages may coexist. We also validated the use of rGFP expression as a biosensor in animal models of latent infection. Our models and biosensor should allow further characterization of quiescence at metabolic and molecular level.
Collapse
|
18
|
Anas M, Kumari V, Gupta N, Dube A, Kumar N. Protein quality control machinery in intracellular protozoan parasites: hopes and challenges for therapeutic targeting. Cell Stress Chaperones 2019; 24:891-904. [PMID: 31228085 PMCID: PMC6717229 DOI: 10.1007/s12192-019-01016-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 01/28/2023] Open
Abstract
Intracellular protozoan parasites have evolved an efficient protein quality control (PQC) network comprising protein folding and degradation machineries that protect the parasite's proteome from environmental perturbations and threats posed by host immune surveillance. Interestingly, the components of PQC machinery in parasites have acquired sequence insertions which may provide additional interaction interfaces and diversify the repertoire of their biological roles. However, the auxiliary functions of PQC machinery remain poorly explored in parasite. A comprehensive understanding of this critical machinery may help to identify robust biological targets for new drugs against acute or latent and drug-resistant infections. Here, we review the dynamic roles of PQC machinery in creating a safe haven for parasite survival in hostile environments, serving as a metabolic sensor to trigger transformation into phenotypically distinct stages, acting as a lynchpin for trafficking of parasite cargo across host membrane for immune evasion and serving as an evolutionary capacitor to buffer mutations and drug-induced proteotoxicity. Versatile roles of PQC machinery open avenues for exploration of new drug targets for anti-parasitic intervention and design of strategies for identification of potential biomarkers for point-of-care diagnosis.
Collapse
Affiliation(s)
- Mohammad Anas
- Department of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Varsha Kumari
- Department of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Niharika Gupta
- Department of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Anuradha Dube
- Department of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Niti Kumar
- Academy of Scientific and Innovative Research (AcSIR), Delhi, India.
| |
Collapse
|
19
|
Chowdhury FT, Shohan MU, Islam T, Mimu TT, Palit P. A Therapeutic Approach Against Leishmania donovani by Predicting RNAi Molecules Against the Surface Protein, gp63. Curr Bioinform 2019. [DOI: 10.2174/1574893613666180828095737] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background:
Leishmaniasis is a disease caused by the Leishmania sp. and can be
classified into two major types: cutaneous and visceral leismaniasis. Visceral leishmaniasis is the
deadlier type and is mediated by Leishmania donovani and involves the establishment of persistent
infection and causes damage to the liver, spleen and bone marrow. With no vaccine yet available
against leishmaniasis and the current therapeutic drugs of leishmaniasis being toxic and expensive;
an alternative treatment is necessary.
Objective:
Surface glycocalyx protein gp63, plays a major role in the virulence and resulting
pathogenicity associated with the disease. Henceforth, silencing the gp63 mRNA through the RNA
interference system was the aim of this study.
Methods:
In this study two competent siRNAs and three miRNAs have been designed against gp63
for five different strains of L. donovani by using various computational methods. Target specific
siRNAs were designed using siDirect 2.0 and to design possible miRNA, another tool named IDT
(IntegratedDNA Technology). Screening for off-target similarity was done by BLAST and the GC
contents and the secondary structures of the designed RNAs were determined. RNA-RNA
interaction was calculated by RNAcofold and IntraRNA, followed by the determination of heat
capacity and the concentration of duplex by DNAmelt web server.
Results:
The selected RNAi molecules; two siRNA and three miRNA had no off-target in human
genome and the ones with lower GC content were selected for efficient RNAi function. The
selected ones showed proper thermodynamic characteristics to suppress the expression of the
pathogenic gene of gp63.
Collapse
Affiliation(s)
- Farhana T. Chowdhury
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Mohammad U.S. Shohan
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Tasmia Islam
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Taisha T. Mimu
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Parag Palit
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
20
|
Zamani S, Hoseini AZ, Namin AM. Glucose-6-phosphate dehydrogenase (G6PD) activity can modulate macrophage response to Leishmania major infection. Int Immunopharmacol 2019; 69:178-183. [DOI: 10.1016/j.intimp.2019.01.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/22/2018] [Accepted: 01/18/2019] [Indexed: 10/27/2022]
|
21
|
Dai W, Wu Y, Bi J, Wang S, Li F, Kong W, Barbier J, Cintrat JC, Gao F, Gillet D, Su W, Jiang C. Antiviral Effects of ABMA against Herpes Simplex Virus Type 2 In Vitro and In Vivo. Viruses 2018. [PMID: 29522484 PMCID: PMC5869512 DOI: 10.3390/v10030119] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Herpes simplex virus type 2 (HSV-2) is the causative pathogen of genital herpes and is closely associated with the occurrence of cervical cancer and human immunodeficiency virus (HIV) infection. The absence of an effective vaccine and the emergence of drug resistance to commonly used nucleoside analogs emphasize the urgent need for alternative antivirals against HSV-2. Recently, ABMA [1-adamantyl (5-bromo-2-methoxybenzyl) amine] has been demonstrated to be an inhibitor of several pathogens exploiting host-vesicle transport, which also participates in the HSV-2 lifecycle. Here, we showed that ABMA inhibited HSV-2-induced cytopathic effects and plaque formation with 50% effective concentrations of 1.66 and 1.08 μM, respectively. We also preliminarily demonstrated in a time of compound addition assay that ABMA exerted a dual antiviral mechanism by impairing virus entry, as well as the late stages of the HSV-2 lifecycle. Furthermore, in vivo studies showed that ABMA protected BALB/c mice from intravaginal HSV-2 challenge with an improved survival rate of 50% at 5 mg/kg (8.33% for the untreated virus infected control). Consequently, our study has identified ABMA as an effective inhibitor of HSV-2, both in vitro and in vivo, for the first time and presents an alternative to nucleoside analogs for HSV-2 infection treatment.
Collapse
Affiliation(s)
- Wenwen Dai
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
| | - Yu Wu
- SIMOPRO, CEA, Université Paris-Saclay, F-91191 Gif Sur Yvette, France.
| | - Jinpeng Bi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
| | - Shuai Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
| | - Fang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
| | - Julien Barbier
- SIMOPRO, CEA, Université Paris-Saclay, F-91191 Gif Sur Yvette, France.
| | | | - Feng Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
| | - Daniel Gillet
- SIMOPRO, CEA, Université Paris-Saclay, F-91191 Gif Sur Yvette, France.
| | - Weiheng Su
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, Jilin, China.
| |
Collapse
|
22
|
Thompson AM, O'Connor PD, Marshall AJ, Blaser A, Yardley V, Maes L, Gupta S, Launay D, Braillard S, Chatelain E, Wan B, Franzblau SG, Ma Z, Cooper CB, Denny WA. Development of (6 R)-2-Nitro-6-[4-(trifluoromethoxy)phenoxy]-6,7-dihydro-5 H-imidazo[2,1- b][1,3]oxazine (DNDI-8219): A New Lead for Visceral Leishmaniasis. J Med Chem 2018; 61:2329-2352. [PMID: 29461823 PMCID: PMC5867678 DOI: 10.1021/acs.jmedchem.7b01581] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
![]()
Discovery
of the potent antileishmanial effects of antitubercular
6-nitro-2,3-dihydroimidazo[2,1-b][1,3]oxazoles and
7-substituted 2-nitro-5,6-dihydroimidazo[2,1-b][1,3]oxazines
stimulated the examination of further scaffolds (e.g., 2-nitro-5,6,7,8-tetrahydroimidazo[2,1-b][1,3]oxazepines), but the results for these seemed less
attractive. Following the screening of a 900-compound pretomanid analogue
library, several hits with more suitable potency, solubility, and
microsomal stability were identified, and the superior efficacy of
newly synthesized 6R enantiomers with phenylpyridine-based
side chains was established through head-to-head assessments in a Leishmania donovani mouse model. Two such leads (R-84 and R-89) displayed promising activity in the more stringent Leishmania
infantum hamster model but were unexpectedly found to be
potent inhibitors of hERG. An extensive structure–activity
relationship investigation pinpointed two compounds (R-6 and pyridine R-136)
with better solubility and pharmacokinetic properties that also provided
excellent oral efficacy in the same hamster model (>97% parasite
clearance
at 25 mg/kg, twice daily) and exhibited minimal hERG inhibition. Additional
profiling earmarked R-6 as the favored
backup development candidate.
Collapse
Affiliation(s)
- Andrew M Thompson
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| | - Patrick D O'Connor
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| | - Andrew J Marshall
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| | - Adrian Blaser
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| | - Vanessa Yardley
- Faculty of Infectious & Tropical Diseases , London School of Hygiene & Tropical Medicine , Keppel Street , London WC1E 7HT , United Kingdom
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences , University of Antwerp , Universiteitsplein 1 , B-2610 Antwerp , Belgium
| | - Suman Gupta
- Division of Parasitology , CSIR-Central Drug Research Institute , Lucknow 226031 , India
| | - Delphine Launay
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant , 1202 Geneva , Switzerland
| | - Stephanie Braillard
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant , 1202 Geneva , Switzerland
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant , 1202 Geneva , Switzerland
| | - Baojie Wan
- Institute for Tuberculosis Research, College of Pharmacy , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | - Scott G Franzblau
- Institute for Tuberculosis Research, College of Pharmacy , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | - Zhenkun Ma
- Global Alliance for TB Drug Development , 40 Wall Street , New York , New York 10005 , United States
| | - Christopher B Cooper
- Global Alliance for TB Drug Development , 40 Wall Street , New York , New York 10005 , United States
| | - William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| |
Collapse
|
23
|
Vogel G, Cuénod A, Mouchet R, Strauss A, Daubenberger C, Pflüger V, Portevin D. Functional characterization and phenotypic monitoring of human hematopoietic stem cell expansion and differentiation of monocytes and macrophages by whole-cell mass spectrometry. Stem Cell Res 2017; 26:47-54. [PMID: 29227832 DOI: 10.1016/j.scr.2017.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/10/2017] [Accepted: 11/20/2017] [Indexed: 10/18/2022] Open
Abstract
The different facets of macrophages allow them to play distinct roles in tissue homeostasis, tissue repair and in response to infections. Individuals displaying dysregulated macrophage functions are proposed to be prone to inflammatory disorders or infections. However, this being a cause or a consequence of the pathology remains often unclear. In this context, we isolated and expanded CD34+ HSCs from healthy blood donors and derived them into CD14+ myeloid progenitors which were further enriched and differentiated into macrophages. Aiming for a comprehensive phenotypic profiling, we generated whole-cell mass spectrometry (WCMS) fingerprints of cell samples collected along the different stages of the differentiation process to build a predictive model using a linear discriminant analysis based on principal components. Through the capacity of the model to accurately predict sample's identity of a validation set, we demonstrate that WCMS profiles obtained from bona fide blood monocytes and respectively derived macrophages mirror profiles obtained from equivalent HSC derivatives. Finally, HSC-derived macrophage functionalities were assessed by quantifying cytokine and chemokine responses to a TLR agonist in a 34-plex luminex assay and by measuring their capacity to phagocytise mycobacteria. These functional read-outs could not discriminate blood monocytes-derived from HSC-derived macrophages. To conclude, we propose that this method opens new avenues to distinguish the impact of human genetics on the dysregulated biological properties of macrophages in pathological conditions.
Collapse
Affiliation(s)
| | - Aline Cuénod
- Department of Medical Parasitology and Infection Biology, Swiss TPH, Basel, Switzerland; University of Basel, 4002 Basel, Switzerland
| | | | | | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Swiss TPH, Basel, Switzerland; University of Basel, 4002 Basel, Switzerland
| | | | - Damien Portevin
- Department of Medical Parasitology and Infection Biology, Swiss TPH, Basel, Switzerland; University of Basel, 4002 Basel, Switzerland.
| |
Collapse
|
24
|
ABMA, a small molecule that inhibits intracellular toxins and pathogens by interfering with late endosomal compartments. Sci Rep 2017; 7:15567. [PMID: 29138439 PMCID: PMC5686106 DOI: 10.1038/s41598-017-15466-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/24/2017] [Indexed: 12/30/2022] Open
Abstract
Intracellular pathogenic microorganisms and toxins exploit host cell mechanisms to enter, exert their deleterious effects as well as hijack host nutrition for their development. A potential approach to treat multiple pathogen infections and that should not induce drug resistance is the use of small molecules that target host components. We identified the compound 1-adamantyl (5-bromo-2-methoxybenzyl) amine (ABMA) from a cell-based high throughput screening for its capacity to protect human cells and mice against ricin toxin without toxicity. This compound efficiently protects cells against various toxins and pathogens including viruses, intracellular bacteria and parasite. ABMA provokes Rab7-positive late endosomal compartment accumulation in mammalian cells without affecting other organelles (early endosomes, lysosomes, the Golgi apparatus, the endoplasmic reticulum or the nucleus). As the mechanism of action of ABMA is restricted to host-endosomal compartments, it reduces cell infection by pathogens that depend on this pathway to invade cells. ABMA may represent a novel class of broad-spectrum compounds with therapeutic potential against diverse severe infectious diseases.
Collapse
|
25
|
Characterization of the Protein Tyrosine Phosphatase LmPRL-1 Secreted by Leishmania major via the Exosome Pathway. Infect Immun 2017; 85:IAI.00084-17. [PMID: 28507071 DOI: 10.1128/iai.00084-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/11/2017] [Indexed: 01/02/2023] Open
Abstract
Similar to other intracellular pathogens, Leishmania parasites are known to evade the antimicrobial effector functions of host immune cells. To date, however, only a few virulence factors have been described for Leishmania major, one of the causative agents of cutaneous leishmaniasis. Here, we have characterized the expression and function of an L. major phosphatase, which we termed LmPRL-1. This enzyme shows a strong structural similarity to the human phosphatases of regenerating liver (PRL-1, -2, and -3) that regulate the proliferation, differentiation, and motility of cells. The biochemical characterization of the L. major phosphatase revealed that the enzyme is redox sensitive. When analyzing the subcellular localization of LmPRL-1 in promastigotes, amastigotes, and infected macrophages, we found that the phosphatase was predominantly expressed and secreted by promastigotes via the exosome route. Finally, we observed that ectopic expression of LmPRL-1 in L. major led to an increased number of parasites in macrophages. From these data, we conclude that the L. major phosphatase LmPRL-1 contributes to the intracellular survival of the parasites in macrophages.
Collapse
|
26
|
Chakraborty A, Kurati SP, Mahata SK, Sundar S, Roy S, Sen M. Wnt5a Signaling Promotes Host Defense against Leishmania donovani Infection. THE JOURNAL OF IMMUNOLOGY 2017; 199:992-1002. [PMID: 28659356 DOI: 10.4049/jimmunol.1601927] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/30/2017] [Indexed: 11/19/2022]
Abstract
Leishmania donovani infects macrophages, disrupting immune homeostasis. The underlying mechanism that sustains infection remains unresolved. In view of the potential of Wnt5a signaling to support immune homeostasis, we evaluated the interrelationship of Wnt5a signaling and Leishmania donovani infection. Upon infecting macrophages separately with antimony drug-sensitive and -resistant L. donovani, we noted disruption in the steady-state level of Wnt5a. Moreover, inhibition of Wnt5a signaling by small interfering RNA transfection in vitro or by use of inhibitor of Wnt production in vivo led to an increase in cellular parasite load. In contrast, treatment of macrophages with recombinant Wnt5a caused a decrease in the load of antimony-sensitive and -resistant parasites, thus confirming that Wnt5a signaling antagonizes L. donovani infection. Using inhibitors of the Wnt5a signaling intermediates Rac1 and Rho kinase, we demonstrated that Wnt5a-mediated inhibition of parasite infection in macrophages is Rac1/Rho dependent. Furthermore, phalloidin staining and reactive oxygen species estimation of Wnt5a-treated macrophages suggested that a Wnt5a-Rac/Rho-mediated decrease in parasite load is associated with an increase in F- actin assembly and NADPH oxidase activity. Moreover, live microscopy of L. donovani-infected macrophages treated with Wnt5a demonstrated increased endosomal/lysosomal fusions with parasite-containing vacuoles (parasitophorous vacuoles [PV]). An increase in PV-endosomal/lysosomal fusion accompanied by augmented PV degradation in Wnt5a-treated macrophages was also apparent from transmission electron microscopy of infected cells. Our results suggest that, although L. donovani evades host immune response, at least in part through inhibition of Wnt5a signaling, revamping Wnt5a signaling can inhibit L. donovani infection, irrespective of drug sensitivity or resistance.
Collapse
Affiliation(s)
- Arijit Chakraborty
- Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India
| | - Sony Priya Kurati
- Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India
| | - Sushil K Mahata
- Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161.,Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Shyam Sundar
- Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Syamal Roy
- Division of Infectious Disease and Immunology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India; and.,Coochbehar Panchanan Barma University, Cooch Behar, West Bengal 736101, India
| | - Malini Sen
- Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India;
| |
Collapse
|
27
|
Lamotte S, Späth GF, Rachidi N, Prina E. The enemy within: Targeting host-parasite interaction for antileishmanial drug discovery. PLoS Negl Trop Dis 2017; 11:e0005480. [PMID: 28594938 PMCID: PMC5464532 DOI: 10.1371/journal.pntd.0005480] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The state of antileishmanial chemotherapy is strongly compromised by the emergence of drug-resistant Leishmania. The evolution of drug-resistant phenotypes has been linked to the parasites’ intrinsic genome instability, with frequent gene and chromosome amplifications causing fitness gains that are directly selected by environmental factors, including the presence of antileishmanial drugs. Thus, even though the unique eukaryotic biology of Leishmania and its dependence on parasite-specific virulence factors provide valid opportunities for chemotherapeutical intervention, all strategies that target the parasite in a direct fashion are likely prone to select for resistance. Here, we review the current state of antileishmanial chemotherapy and discuss the limitations of ongoing drug discovery efforts. We finally propose new strategies that target Leishmania viability indirectly via mechanisms of host–parasite interaction, including parasite-released ectokinases and host epigenetic regulation, which modulate host cell signaling and transcriptional regulation, respectively, to establish permissive conditions for intracellular Leishmania survival.
Collapse
Affiliation(s)
- Suzanne Lamotte
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Gerald F. Späth
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Najma Rachidi
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Eric Prina
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
- * E-mail:
| |
Collapse
|
28
|
Vermelho AB, Capaci GR, Rodrigues IA, Cardoso VS, Mazotto AM, Supuran CT. Carbonic anhydrases from Trypanosoma and Leishmania as anti-protozoan drug targets. Bioorg Med Chem 2017; 25:1543-1555. [PMID: 28161253 DOI: 10.1016/j.bmc.2017.01.034] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 01/18/2017] [Accepted: 01/19/2017] [Indexed: 01/24/2023]
Abstract
Trypanosoma cruzi and Leishmania spp. are protozoa of the Trypanosomatidae family, being the etiological agents of two widespread parasitic diseases, Chagas disease and leishmaniasis, respectively. Both parasites are the focus of worldwide research with the aim to find effective and less toxic drugs than the few ones available so far, and for controlling the spread of the diseases. Carbonic anhydrases (CAs, EC 4.2.1.1) belonging to the α- and β-class were recently identified in these protozoans and several studies suggested that they could be new targets for drug development. Sulfonamide, thiol and hydroxamate inhibitors effectively inhibited the α-CA from T. cruzi (TcCA) and the β-CA from L. donovani chagasi (LdccCA) in vitro, and some of them also showed in vivo efficacy in inhibiting the growth of the parasites in animal models of Chagas disease and leishmaniasis. As few therapeutic options are presently available for these orphan diseases, protozoan CA inhibition may represent a novel strategy to address this stringent health problem.
Collapse
Affiliation(s)
- Alane B Vermelho
- BIOINOVAR - Biotechnology Laboratories: Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Giseli R Capaci
- School of Science and Technology and Graduate Studies in Science Education Program, University of Rio Grande, Duque de Caxias, RJ, Brazil
| | - Igor A Rodrigues
- Department of Natural Products and Food, School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Verônica S Cardoso
- BIOINOVAR - Biotechnology Laboratories: Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Maria Mazotto
- BIOINOVAR - Biotechnology Laboratories: Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudiu T Supuran
- Neurofarba Department and Laboratorio di Chimica Bioinorganica, Università degli Studi di Firenze, Via U. Schiff 6, 50019 Sesto Fiorentino, Florence, Italy.
| |
Collapse
|
29
|
Gannavaram S, Bhattacharya P, Ismail N, Kaul A, Singh R, Nakhasi HL. Modulation of Innate Immune Mechanisms to Enhance Leishmania Vaccine-Induced Immunity: Role of Coinhibitory Molecules. Front Immunol 2016; 7:187. [PMID: 27242794 PMCID: PMC4865500 DOI: 10.3389/fimmu.2016.00187] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/02/2016] [Indexed: 12/14/2022] Open
Abstract
No licensed human vaccines are currently available against any parasitic disease including leishmaniasis. Several antileishmanial vaccine formulations have been tested in various animal models, including genetically modified live-attenuated parasite vaccines. Experimental infection studies have shown that Leishmania parasites utilize a broad range of strategies to undermine effector properties of host phagocytic cells, i.e., dendritic cells (DCs) and macrophages (MΦ). Furthermore, Leishmania parasites have evolved strategies to actively inhibit TH1 polarizing functions of DCs and to condition the infected MΦ toward anti-inflammatory/alternative/M2 phenotype. The altered phenotype of phagocytic cells is characterized by decreased production of antimicrobial reactive oxygen, nitrogen molecules, and pro-inflammatory cytokines, such as IFN-γ, IL-12, and TNF-α. These early events limit the activation of TH1-effector cells and set the stage for pathogenesis. Furthermore, this early control of innate immunity by the virulent parasites results in substantial alteration in the adaptive immunity characterized by reduced proliferation of CD4+ and CD8+ T cells and TH2-biased immunity that results in production of anti-inflammatory cytokines, such as TGF-β, and IL-10. More recent studies have also documented the induction of coinhibitory ligands, such as CTLA-4, PD-L1, CD200, and Tim-3, that induce exhaustion and/or non-proliferation in antigen-experienced T cells. Most of these studies focus on viral infections in chronic phase, thus limiting the direct application of these results to parasitic infections and much less to parasitic vaccines. However, these studies suggest that vaccine-induced protective immunity can be modulated using strategies that enhance the costimulation that might reduce the threshold necessary for T cell activation and conversely by strategies that reduce or block inhibitory molecules, such as PD-L1 and CD200. In this review, we will focus on the polarization of antigen-presenting cells and subsequent role of costimulatory and coinhibitory molecules in mediating vaccine-induced immunity using live-attenuated Leishmania parasites as specific examples.
Collapse
Affiliation(s)
- Sreenivas Gannavaram
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| | - Parna Bhattacharya
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| | - Nevien Ismail
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| | - Amit Kaul
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| | - Rakesh Singh
- Department of Biochemistry, Banaras Hindu University , Varanasi , India
| | - Hira L Nakhasi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| |
Collapse
|
30
|
Toledo DAM, D'Avila H, Melo RCN. Host Lipid Bodies as Platforms for Intracellular Survival of Protozoan Parasites. Front Immunol 2016; 7:174. [PMID: 27199996 PMCID: PMC4853369 DOI: 10.3389/fimmu.2016.00174] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 04/19/2016] [Indexed: 12/31/2022] Open
Abstract
Pathogens induce several changes in the host cell signaling and trafficking mechanisms in order to evade and manipulate the immune response. One prominent pathogen-mediated change is the formation of lipid-rich organelles, termed lipid bodies (LBs) or lipid droplets, in the host cell cytoplasm. Protozoan parasites, which contribute expressively to the burden of infectious diseases worldwide, are able to induce LB genesis in non-immune and immune cells, mainly macrophages, key players in the initial resistance to the infection. Under host–parasite interaction, LBs not only accumulate in the host cytoplasm but also relocate around and move into parasitophorous vacuoles. There is increasing evidence that protozoan parasites may target host-derived LBs either for gaining nutrients or for escaping the host immune response. Newly formed, parasite-induced LBs may serve as lipid sources for parasite growth and also produce inflammatory mediators that potentially act in the host immune response deactivation. In this mini review, we summarize current knowledge on the formation and role of host LBs as sites exploited by intracellular protozoan parasites as a strategy to maintain their own survival.
Collapse
Affiliation(s)
- Daniel A M Toledo
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences (ICB), Federal University of Juiz de Fora (UFJF) , Juiz de Fora, Minas Gerais , Brazil
| | - Heloísa D'Avila
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences (ICB), Federal University of Juiz de Fora (UFJF) , Juiz de Fora, Minas Gerais , Brazil
| | - Rossana C N Melo
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences (ICB), Federal University of Juiz de Fora (UFJF) , Juiz de Fora, Minas Gerais , Brazil
| |
Collapse
|
31
|
Barbier J, Cintrat JC, Gillet D. Intracellular pathogens convert macrophages from death traps into hospitable homes. FEBS J 2016; 283:595-7. [DOI: 10.1111/febs.13657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Julien Barbier
- CEA, Section of Molecular Engineering of Proteins (SIMOPRO); Institute of Biology and Biotechnology of Saclay (iBiTec-S); LERMIT LabEx; Paris Saclay University; Gif sur Yvette France
| | - Jean-Christophe Cintrat
- CEA, Section of Bioorganic Chemistry and Labelling (SCBM); Institute of Biology and Biotechnology of Saclay (iBiTec-S); LERMIT LabEx; Paris Saclay University; Gif sur Yvette France
| | - Daniel Gillet
- CEA, Section of Molecular Engineering of Proteins (SIMOPRO); Institute of Biology and Biotechnology of Saclay (iBiTec-S); LERMIT LabEx; Paris Saclay University; Gif sur Yvette France
| |
Collapse
|