1
|
Sannigrahi A, Ghosh S, Pradhan S, Jana P, Jawed JJ, Majumdar S, Roy S, Karmakar S, Mukherjee B, Chattopadhyay K. Leishmania protein KMP-11 modulates cholesterol transport and membrane fluidity to facilitate host cell invasion. EMBO Rep 2024:10.1038/s44319-024-00302-7. [PMID: 39482488 DOI: 10.1038/s44319-024-00302-7] [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: 07/16/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024] Open
Abstract
The first step of successful infection by any intracellular pathogen relies on its ability to invade its host cell membrane. However, the detailed structural and molecular understanding underlying lipid membrane modification during pathogenic invasion remains unclear. In this study, we show that a specific Leishmania donovani (LD) protein, KMP-11, forms oligomers that bridge LD and host macrophage (MΦ) membranes. This KMP-11 induced interaction between LD and MΦ depends on the variations in cholesterol (CHOL) and ergosterol (ERG) contents in their respective membranes. These variations are crucial for the subsequent steps of invasion, including (a) the initial attachment, (b) CHOL transport from MΦ to LD, and (c) detachment of LD from the initial point of contact through a liquid ordered (Lo) to liquid disordered (Ld) membrane-phase transition. To validate the importance of KMP-11, we generate KMP-11 depleted LD, which failed to attach and invade host MΦ. Through tryptophan-scanning mutagenesis and synthesized peptides, we develop a generalized mathematical model, which demonstrates that the hydrophobic moment and the symmetry sequence code at the membrane interacting protein domain are key factors in facilitating the membrane phase transition and, consequently, the host cell infection process by Leishmania parasites.
Collapse
Affiliation(s)
- Achinta Sannigrahi
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Souradeepa Ghosh
- School of Medical Science and Technology, IIT-Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Supratim Pradhan
- School of Medical Science and Technology, IIT-Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Pulak Jana
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India
| | - Junaid Jibran Jawed
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, 700156, India
| | - Subrata Majumdar
- Department of Molecular Medicine, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Syamal Roy
- Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India
- INSA Senior Scientist, Indian Association for the Cultivation of Science, Kolkata, West Bengal, 700032, India
| | - Sanat Karmakar
- Department of Physics, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India
| | - Budhaditya Mukherjee
- School of Medical Science and Technology, IIT-Kharagpur, Kharagpur, West Bengal, 721302, India.
| | - Krishnananda Chattopadhyay
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India.
| |
Collapse
|
2
|
Díaz-Varela M, Sanchez-Hidalgo A, Calderon-Copete S, Tacchini V, Shipley TR, Ramírez LG, Marquis J, Fernández OL, Saravia NG, Tacchini-Cottier F. The different impact of drug-resistant Leishmania on the transcription programs activated in neutrophils. iScience 2024; 27:109773. [PMID: 38711445 PMCID: PMC11070714 DOI: 10.1016/j.isci.2024.109773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024] Open
Abstract
Drug resistance threatens the effective control of infections, including parasitic diseases such as leishmaniases. Neutrophils are essential players in antimicrobial control, but their role in drug-resistant infections is poorly understood. Here, we evaluated human neutrophil response to clinical parasite strains having distinct natural drug susceptibility. We found that Leishmania antimony drug resistance significantly altered the expression of neutrophil genes, some of them transcribed by specific neutrophil subsets. Infection with drug-resistant parasites increased the expression of detoxification pathways and reduced the production of cytokines. Among these, the chemokine CCL3 was predominantly impacted, which resulted in an impaired ability of neutrophils to attract myeloid cells. Moreover, decreased myeloid recruitment when CCL3 levels are reduced was confirmed by blocking CCL3 in a mouse model. Collectively, these findings reveal that the interplay between naturally drug-resistant parasites and neutrophils modulates the infected skin immune microenvironment, revealing a key role of neutrophils in drug resistance.
Collapse
Affiliation(s)
- Míriam Díaz-Varela
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Andrea Sanchez-Hidalgo
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Sandra Calderon-Copete
- Lausanne Genomic Technologies Facility, University of Lausanne, 1015 Lausanne, Switzerland
| | - Virginie Tacchini
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Tobias R. Shipley
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Lady Giovanna Ramírez
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Julien Marquis
- Lausanne Genomic Technologies Facility, University of Lausanne, 1015 Lausanne, Switzerland
| | - Olga Lucía Fernández
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Nancy Gore Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Fabienne Tacchini-Cottier
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| |
Collapse
|
3
|
Ansari A, Seth A, Dutta M, Qamar T, Katiyar S, Jaiswal AK, Rani A, Majhi S, Kumar M, Bhatta RS, Guha R, Mitra K, Sashidhara KV, Kar S. Discovery, SAR and mechanistic studies of quinazolinone-based acetamide derivatives in experimental visceral leishmaniasis. Eur J Med Chem 2023; 257:115524. [PMID: 37290183 DOI: 10.1016/j.ejmech.2023.115524] [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: 04/17/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/10/2023]
Abstract
Towards identification of novel therapeutic candidates, a series of quinazolinone-based acetamide derivatives were synthesized and assessed for their anti-leishmanial efficacy. Amongst synthesized derivatives, compounds F12, F27 and F30 demonstrated remarkable activity towards intracellular L. donovani amastigotes in vitro, with IC50 values of 5.76 ± 0.84 μM, 3.39 ± 0.85 μM and 8.26 ± 1.23 μM against promastigotes, and 6.02 μM ± 0.52, 3.55 ± 0.22 μM and 6.23 ± 0.13 μM against amastigotes, respectively. Oral administration of compounds F12 and F27 entailed >85% reduction in organ parasite burden in L. donovani-infected BALB/c mice and hamsters, by promoting host-protective Th1 cytokine response. In host J774 macrophages, mechanistic studies revealed inhibition of PI3K/Akt/CREB axis, resulting in a decrease of IL-10 versus IL-12 release upon F27 treatment. In silico docking studies conducted with lead compound, F27 demonstrated plausible inhibition of Leishmania prolyl-tRNA synthetase, which was validated via detection of decreased proline levels in parasites and induction of amino acid starvation, leading to G1 cell cycle arrest and autophagy-mediated programmed cell death of L. donovani promastigotes. Structure-activity analysis and study of pharmacokinetic and physicochemical parameters suggest oral availability and underscore F27 as a promising lead for anti-leishmanial drug development.
Collapse
Affiliation(s)
- Alisha Ansari
- Medicinal and Process Chemistry Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Anuradha Seth
- Molecular Microbiology & Immunology Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Mukul Dutta
- Molecular Microbiology & Immunology Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Tooba Qamar
- Molecular Microbiology & Immunology Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Sarita Katiyar
- Medicinal and Process Chemistry Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Arvind K Jaiswal
- Medicinal and Process Chemistry Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Ankita Rani
- Molecular Microbiology & Immunology Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Swetapadma Majhi
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Mukesh Kumar
- Pharmacokinetics and Metabolism Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Rabi S Bhatta
- Pharmacokinetics and Metabolism Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rajdeep Guha
- Laboratory Animal Facility Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Kalyan Mitra
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Koneni V Sashidhara
- Medicinal and Process Chemistry Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India.
| | - Susanta Kar
- Molecular Microbiology & Immunology Division, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
4
|
Domagalska MA, Barrett MP, Dujardin JC. Drug resistance in Leishmania: does it really matter? Trends Parasitol 2023; 39:251-259. [PMID: 36803859 DOI: 10.1016/j.pt.2023.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/19/2023]
Abstract
Treatment failure (TF) jeopardizes the management of parasitic diseases, including leishmaniasis. From the parasite's point of view, drug resistance (DR) is generally considered as central to TF. However, the link between TF and DR, as measured by in vitro drug susceptibility assays, is unclear, some studies revealing an association between treatment outcome and drug susceptibility, others not. Here we address three fundamental questions aiming to shed light on these ambiguities. First, are the right assays being used to measure DR? Second, are the parasites studied, which are generally those that adapt to in vitro culture, actually appropriate? Finally, are other parasite factors - such as the development of quiescent forms that are recalcitrant to drugs - responsible for TF without DR?
Collapse
Affiliation(s)
| | - Michael P Barrett
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
5
|
Chowdhuri SP, Dhiman S, Das SK, Meena N, Das S, Kumar A, Das BB. Novel Pyrido[2',1':2,3]imidazo[4,5- c]quinoline Derivative Selectively Poisons Leishmania donovani Bisubunit Topoisomerase 1 to Inhibit the Antimony-Resistant Leishmania Infection in Vivo. J Med Chem 2023; 66:3411-3430. [PMID: 36823782 DOI: 10.1021/acs.jmedchem.2c01932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
The unique bisubunit structure of Leishmania donovani topoisomerase 1B (LdTop1) is a potential drug target in the parasites unlike the monomeric Top1 from its human host counterpart. Here, we report the design, synthesis, and validation of a chimeric pyrido[2',1':2,3]imidazo[4,5-c]quinoline derivative (C17) as a novel antileishmanial agent that poisons topoisomerase 1-DNA covalent complexes (LdTop1cc) inside the parasites and inhibits Top1 religation activity both in the drug sensitive and antimony-resistant L. donovani clinical isolates. Importantly, the human Top1 is not sensitive to C17. Further, C17 overcomes the chemical instability of camptothecin (CPT) by generating persistent LdTop1cc-induced DNA breaks inside the parasites even after 12 h of drug removal. Intraperitoneal administration of C17 results in marked reduction of the Leishmania amastigotes from the infected spleen and liver of BALB/c mice. C17 confers a host protective immune-response up-regulating the Th1 cytokines facilitating parasite clearance which can be exploited for treating drug-resistant leishmaniasis.
Collapse
Affiliation(s)
- Srijita Paul Chowdhuri
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & B, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India
| | - Shiv Dhiman
- Department of Chemistry, Birla Institute of Technology and Science, Pilani 333 031 Rajasthan, India
| | - Subhendu K Das
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & B, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India
| | - Neha Meena
- Department of Chemistry, Birla Institute of Technology and Science, Pilani 333 031 Rajasthan, India
| | - Sonali Das
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Anil Kumar
- Department of Chemistry, Birla Institute of Technology and Science, Pilani 333 031 Rajasthan, India
| | - Benu Brata Das
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & B, Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India
| |
Collapse
|
6
|
Goel N, Gupta VK, Garg A, Bhoumik A, Biswas R, Natarajan R, Majumder HK, Jaisankar P. Holanamine, a Steroidal Alkaloid from the Bark of Holarrhena pubescens Wall. ex G. Don Inhibits the Growth of Leishmania donovani by Targeting DNA Topoisomerase 1B. ACS Infect Dis 2023; 9:162-177. [PMID: 36417798 DOI: 10.1021/acsinfecdis.2c00562] [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: 11/24/2022]
Abstract
Leishmaniasis is a group of neglected tropical diseases (NTDs) caused by about 20 species of obligate intracellular protozoan parasites of the genus Leishmania, which occurs in cutaneous, mucocutaneous, and visceral forms. Many researchers have sought to utilize natural products for novel and effective treatments to combat many infectious diseases, including leishmaniasis. Holarrhena pubescens Wall. ex G. Don (Apocynaceae) bark is a rich source of bioactive steroidal alkaloids. The total alkaloidal extract (IC50 6.12 ± 0.117 μg/mL), and the isolated alkaloid, holanamine, showed significant antileishmanial activity (IC50 2.66 ± 0.112 μM against AG83 and 3.80 ± 0.126 μM against BHU-575) against the Leishmania donovani parasite, better than miltefosine (IC50 19.61 ± 0.093 μM against AG83 and 23.20 ± 0.094 μM against BHU-575). Holanamine inhibited the L. donovani topoisomerase 1B (LdToP1B) in a non-competitive manner (IC50 2.81 ± 0.105 μM), indicating that it interacts with the free enzyme and enzyme-DNA complex without inhibiting human topoisomerase. Hydrogen bonding and hydrophobic interactions of holanamine with the N-terminal and hinge region of the large subunit of LTop1B is responsible for its potent antileishmanial activity, as shown by docking studies. Treatment with holanamine causes apoptotic-like cell death by generating cellular and mitochondrial reactive oxygen species, disrupting the mitochondrial membrane potential and inducing ultrastructural alterations in the promastigotes. Holanamine effectively clears intracellular amastigotes but minimally affects host macrophages with no significant cytotoxicity in HEK 293 and L929 cell lines. Thus, our studies show that holanamine can further be used to develop effective antileishmanial agents against evolving drug-resistant parasites.
Collapse
Affiliation(s)
- Narender Goel
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168, Maniktala Main Road, Kolkata700054, India.,Laboratory of Catalysis and Chemical Biology, Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata700032, India
| | - Vivek Kumar Gupta
- Laboratory of Catalysis and Chemical Biology, Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata700032, India
| | - Aakriti Garg
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168, Maniktala Main Road, Kolkata700054, India
| | - Arpita Bhoumik
- Laboratory of Molecular Parasitology, Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata700032, India
| | - Raju Biswas
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata700032, India
| | - Ramalingam Natarajan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata700032, India
| | - Hemanta K Majumder
- Laboratory of Molecular Parasitology, Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata700032, India
| | - Parasuraman Jaisankar
- Laboratory of Catalysis and Chemical Biology, Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata700032, India
| |
Collapse
|
7
|
Khanra S, Das S, Sarraf NR, Datta S, Das AK, Manna M, Roy S. Antimony resistance mechanism in genetically different clinical isolates of Indian Kala-azar patients. Front Cell Infect Microbiol 2022; 12:1021464. [PMID: 36405965 PMCID: PMC9667115 DOI: 10.3389/fcimb.2022.1021464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/06/2022] [Indexed: 01/04/2024] Open
Abstract
The central theme of this enterprise is to find common features, if any, displayed by genetically different antimony (Sb)-resistant viscerotropic Leishmania parasites to impart Sb resistance. In a limited number of clinical isolates (n = 3), we studied the breadth of variation in the following dimensions: (a) intracellular thiol content, (b) cell surface expression of glycan having N-acetyl-D-galactosaminyl residue as the terminal sugar, and (c) gene expression of thiol-synthesizing enzymes (CBS, MST, gamma-GCS, ODC, and TR), antimony-reducing enzymes (TDR and ACR2), and antimonial transporter genes (AQP1, MRPA, and PRP1). One of the isolates, T5, that was genotypically characterized as Leishmania tropica, caused Indian Kala-azar and was phenotypically Sb resistant (T5-LT-SSG-R), while the other two were Leishmania donovani, out of which one isolate, AG83, is antimony sensitive (AG83-LD-SSG-S) and the other isolate, T8, is Sb resistant (T8-LD-SSG-R). Our study showed that the Sb-resistant parasites, regardless of their genotype, showed significantly higher intracellular thiol compared with Sb-sensitive AG83-LD-SSG-S. Seemingly, T5-LT-SSG-R showed about 1.9-fold higher thiol content compared with T8-LD-SSG-R which essentially mirrored cell surface N-acetyl-D-galactosaminyl expression. Except TR, the expression of the remaining thiol-synthesizing genes was significantly higher in T8-LD-SSG-R and T5-LT-SSG-R than the sensitive one, and between the Sb-resistant parasites, the latter showed a significantly higher expression. Furthermore, the genes for Sb-reducing enzymes increased significantly in resistant parasites regardless of genotype compared with the sensitive one, and between two resistant parasites, there was hardly any difference in expression. Out of three antimony transporters, AQP1 was decreased with the concurrent increase in MRPA and PRP1 in resistant isolates when compared with the sensitive counterpart. Interestingly, no difference in expression of the above-mentioned transporters was noted between two Sb-resistant isolates. The enduring image that resonated from our study is that the genetically diverse Sb-resistant parasites showed enhanced thiol-synthesizing and antimony transporter gene expression than the sensitive counterpart to confer a resistant phenotype.
Collapse
Affiliation(s)
- Supriya Khanra
- Department of Zoology, Barasat Government College, Kolkata, India
| | - Shantanabha Das
- Department of Zoology, Diamond Harbour Women’s University, Sarisha, West Bengal, India
| | | | - Sanchita Datta
- Department of Zoology, Barasat Government College, Kolkata, India
| | - Anjan Kumar Das
- Department of Medicine, Calcutta National Medical College, Kolkata, India
| | - Madhumita Manna
- Department of Zoology, Barasat Government College, Kolkata, India
| | - Syamal Roy
- Department of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
8
|
Wijnant GJ, Dumetz F, Dirkx L, Bulté D, Cuypers B, Van Bocxlaer K, Hendrickx S. Tackling Drug Resistance and Other Causes of Treatment Failure in Leishmaniasis. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.837460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Leishmaniasis is a tropical infectious disease caused by the protozoan Leishmania parasite. The disease is transmitted by female sand flies and, depending on the infecting parasite species, causes either cutaneous (stigmatizing skin lesions), mucocutaneous (destruction of mucous membranes of nose, mouth and throat) or visceral disease (a potentially fatal infection of liver, spleen and bone marrow). Although more than 1 million new cases occur annually, chemotherapeutic options are limited and their efficacy is jeopardized by increasing treatment failure rates and growing drug resistance. To delay the emergence of resistance to existing and new drugs, elucidating the currently unknown causes of variable drug efficacy (related to parasite susceptibility, host immunity and drug pharmacokinetics) and improved use of genotypic and phenotypic tools to define, measure and monitor resistance in the field are critical. This review highlights recent progress in our understanding of drug action and resistance in Leishmania, ongoing challenges (including setbacks related to the COVID-19 pandemic) and provides an overview of possible strategies to tackle this public health challenge.
Collapse
|
9
|
Chowdhury SR, Das SK, Banerjee B, Paul Chowdhuri S, Majumder HK, Das BB. TDP1 knockout Leishmania donovani accumulate topoisomerase 1-linked DNA damage and are hypersensitive to clinically used antileishmanial drugs. FASEB J 2022; 36:e22265. [PMID: 35319800 DOI: 10.1096/fj.202101668rr] [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: 10/28/2021] [Revised: 02/28/2022] [Accepted: 03/08/2022] [Indexed: 11/11/2022]
Abstract
Leishmania donovani, a unicellular protozoan parasite, causes a wide range of human diseases including fatal visceral leishmaniasis. Tyrosyl DNA-phosphodiesterase 1 (TDP1) hydrolyzes the phosphodiester bond between DNA 3'-end and a tyrosyl moiety of trapped topoisomerase I-DNA covalent complexes (Top1cc). We have previously shown Leishmania harbors a TDP1 gene (LdTDP1), however, the biological role of TDP1 remains largely unknown. In the present study, we have generated TDP1 knockout L. donovani (LdTDP1-/- ) promastigotes and have shown that LdTDP1-/- parasites are deficient in 3'-phosphodiesterase activities and were hypersensitive to Top1-poison like camptothecin (CPT), DNA alkylation agent like methyl methanesulfonate, and oxidative DNA lesions generated by hydrogen peroxide but were not sensitive to etoposide. We also detected elevated levels of CPT-induced reactive oxygen species triggering cell cycle arrest and cell death in LdTDP1-/- promastigotes. LdTDP1-/- promastigotes accumulate a significant change in the membrane morphology with the accumulation of membrane pores, which is associated with oxidative stress and lipid peroxidation. To our surprise, we detected that LdTDP1-/- parasites were hypersensitive to antileishmanial drugs like amphotericin B and miltefosine, which could be rescued by complementation of wild-type TDP1 gene in the LdTDP1-/- parasites. Notably, multidrug-resistant L. donovani clinical isolates showed a marked reduction in TDP1 expression and were sensitive to Top1 poisons. Taken together, our study provides a new role of LdTDP1 in protecting L. donovani parasites from oxidative stress-induced DNA damage and resistance to amphotericin B and miltefosine.
Collapse
Affiliation(s)
- Somenath Roy Chowdhury
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Subhendu K Das
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India
| | - Bijoylaxmi Banerjee
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Srijita Paul Chowdhuri
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India
| | - Hemanta K Majumder
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Benu Brata Das
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India
| |
Collapse
|
10
|
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: 2.5] [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
|
11
|
Abhishek K, Kumar A, Sardar AH, Vijayakumar S, Dikhit MR, Kumar A, Kumar V, Das S, Das P. Differential translational regulation of host exosomal proteins play key role in immunomodulation in antimony resistance in Visceral Leishmaniasis: A proteomic profiling study. Acta Trop 2022; 226:106268. [PMID: 34890541 DOI: 10.1016/j.actatropica.2021.106268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/25/2021] [Accepted: 12/05/2021] [Indexed: 11/01/2022]
Abstract
In host-pathogen interactions, exosomal secretions are crucial for cell to cell communication and have an established role in immunomodulation. Protozoans, including Leishmania, modulates their host vesicular secretions for better survival; although the role of exosomal secretions in unresponsive against sodium antimony gluconate (SAG) has never been documented. In this study, the exosomal proteome of RAW macrophages infected with either SAG responsive (SAGS) or SAG unresponsive (SAGR) L. donovani parasites has been compared with uninfected RAW macrophages. Proteins isolated from exosomes were labelled with iTRAQ reagents; followed by subsequent LC-TOF/-MS analysis. In total, 394 proteins (p < 0.05) were identified which were shared common among all sets. Highly differentially expressed proteins were sorted by log2 value -1 and +1 as down regulated and up regulated respectively which yielded 58 proteins in SAGR and 41 proteins during SAGS infection. Out of the 58 proteins identified during SAGR infection, 17 proteins were of immune modulatory function. Network visualization model and pathway analysis revealed the interactions among these proteins via different immunological pathways with reported involvement of some proteins in SAG resistance and host immune modulation. Hence, the differential abundance of immune pathway related proteins in exosomes of infected host during SAGR infection supports the immune modulatory strategy adopted by SAG resistant parasites for enhanced survival .
Collapse
|
12
|
Guzmán-Gutiérrez SL, Silva-Miranda M, Krengel F, Huerta-Salazar E, León-Santiago M, Díaz-Cantón JK, Espitia Pinzón C, Reyes-Chilpa R. Antimycobacterial Activity of Alkaloids and Extracts from Tabernaemontana alba and T. arborea. PLANTA MEDICA 2022; 88:53-61. [PMID: 32392600 DOI: 10.1055/a-1157-1732] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Tuberculosis is the main cause of death from a single infectious agent. Globally, according to the World Health Organization, in 2018, there were an estimated 1.2 million tuberculosis deaths. Moreover, there is a continuous appearance of drug-resistant strains. Thus, development of new antituberculosis medicines should receive high priority. Plant-derived natural products are promising candidates for this purpose. We therefore screened alkaloid extracts obtained from the root and stem barks of the Mexican Apocynaceae species Tabernaemontana alba and Tabernaemontana arborea, as well as the pure alkaloids ibogaine, voacangine, and voacamine, tested for activity against Mycobacterium tuberculosis H37Rv and cytotoxicity to mammalian Vero cells using the resazurin microtiter and the MTT assays, respectively. The extracts were analyzed by GC-MS and HPLC-UV. T. arborea root bark alkaloid extract showed the highest activity against M. tuberculosis (MIC100 = 7.8 µg/mL) of the four extracts tested. HPLC suggested that voacangine and voacamine were the major components. The latter was isolated by column chromatography, and its chemical structure was elucidated by 1H and 13C NMR, and MS. Unambiguous assignation was performed by HSQC, HMBC, and NOESY experiments. Voacamine is a dimeric bis-indole-type alkaloid and is 15 times more potent than the monomeric ibogan-type alkaloids ibogaine and voacangine (MIC100 = 15.6, 250.0, and 250.0 µg/mL, respectively). However, all of these compounds showed cytotoxicity to Vero cells, with a poor selectivity index of 1.00, 0.16, and 1.42, respectively. This is the first report of voacamine activity against M. tuberculosis.
Collapse
Affiliation(s)
- Silvia Laura Guzmán-Gutiérrez
- CONACyT - Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mayra Silva-Miranda
- CONACyT - Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Felix Krengel
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, México
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| | | | - Mayra León-Santiago
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Jessica Karina Díaz-Cantón
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, México
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Clara Espitia Pinzón
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Ricardo Reyes-Chilpa
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
13
|
Pereira LOR, Sousa CS, Ramos HCP, Torres-Santos EC, Pinheiro LS, Alves MR, Cuervo P, Romero GAS, Boité MC, Porrozzi R, Cupolillo E. Insights from Leishmania (Viannia) guyanensis in vitro behavior and intercellular communication. Parasit Vectors 2021; 14:556. [PMID: 34711290 PMCID: PMC8554959 DOI: 10.1186/s13071-021-05057-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pentavalent antimonial-based chemotherapy is the first-line approach for leishmaniasis treatment and disease control. Nevertheless antimony-resistant parasites have been reported in some endemic regions. Treatment refractoriness is complex and is associated with patient- and parasite-related variables. Although amastigotes are the parasite stage in the vertebrate host and, thus, exposed to the drug, the stress caused by trivalent antimony in promastigotes has been shown to promote significant modification in expression of several genes involved in various biological processes, which will ultimately affect parasite behavior. Leishmania (Viannia) guyanensis is one of the main etiological agents in the Amazon Basin region, with a high relapse rate (approximately 25%). METHODS Herein, we conducted several in vitro analyses with L. (V.) guyanensis strains derived from cured and refractory patients after treatment with standardized antimonial therapeutic schemes, in addition to a drug-resistant in vitro-selected strain. Drug sensitivity assessed through Sb(III) half-maximal inhibitory concentration (IC50) assays, growth patterns (with and without drug pressure) and metacyclic-like percentages were determined for all strains and compared to treatment outcomes. Finally, co-cultivation without intercellular contact was followed by parasitic density and Sb(III) IC50 measurements. RESULTS Poor treatment response was correlated with increased Sb(III) IC50 values. The decrease in drug sensitivity was associated with a reduced cell replication rate, increased in vitro growth ability, and higher metacyclic-like proportion. Additionally, in vitro co-cultivation assays demonstrated that intercellular communication enabled lower drug sensitivity and enhanced in vitro growth ability, regardless of direct cell contact. CONCLUSIONS Data concerning drug sensitivity in the Viannia subgenus are emerging, and L. (V.) guyanensis plays a pivotal epidemiological role in Latin America. Therefore, investigating the parasitic features potentially related to relapses is urgent. Altogether, the data presented here indicate that all tested strains of L. (V.) guyanensis displayed an association between treatment outcome and in vitro parameters, especially the drug sensitivity. Remarkably, sharing enhanced growth ability and decreased drug sensitivity, without intercellular communication, were demonstrated.
Collapse
Affiliation(s)
- Luiza O R Pereira
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.
| | - Cíntia S Sousa
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Hellen C P Ramos
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Liliane S Pinheiro
- Laboratório de Bioquímica de Tripanossomatídeos, IOC, FIOCRUZ, Rio de Janeiro, Brazil.,Instituto de Saúde e Biotecnologia, Universidade Federal do Amazonas, Campus Coari, Amazonas, Brazil
| | - Marcelo R Alves
- Laboratório de Pesquisa Clínica em DST-AIDS, Instituto Nacional de Infectologia Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brazil
| | - Patricia Cuervo
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Mariana C Boité
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Renato Porrozzi
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Elisa Cupolillo
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Rashidi S, Mansouri R, Ali-Hassanzadeh M, Ghani E, Barazesh A, Karimazar M, Nguewa P, Carrera Silva EA. Highlighting the interplay of microRNAs from Leishmania parasites and infected-host cells. Parasitology 2021; 148:1434-1446. [PMID: 34218829 PMCID: PMC11010138 DOI: 10.1017/s0031182021001177] [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: 03/27/2021] [Revised: 06/10/2021] [Accepted: 06/27/2021] [Indexed: 02/05/2023]
Abstract
Leishmania parasites, the causative agents of leishmaniasis, are protozoan parasites with the ability to modify the signalling pathway and cell responses of their infected host cells. These parasite strategies alter the host cell environment and conditions favouring their replication, survival and pathogenesis. Since microRNAs (miRNAs) are able to post-transcriptionally regulate gene expression processes, these biomolecules can exert critical roles in controlling Leishmania-host cell interplay. Therefore, the identification of relevant miRNAs differentially expressed in Leishmania parasites as well as in infected cells, which affect the host fitness, could be critical to understand the infection biology, pathogenicity and immune response against these parasites. Accordingly, the current review aims to address the differentially expressed miRNAs in both, the parasite and infected host cells and how these biomolecules change cell signalling and host immune responses during infection. A deep understanding of these processes could provide novel guidelines and therapeutic strategies for managing and treating leishmaniasis.
Collapse
Affiliation(s)
- Sajad Rashidi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Esmaeel Ghani
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afshin Barazesh
- Department of Microbiology and Parasitology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammadreza Karimazar
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paul Nguewa
- University of Navarra, ISTUN Instituto de Salud Tropical, Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), c/Irunlarrea 1, 31008Pamplona, Spain
| | | |
Collapse
|
15
|
Magalhães LS, Bomfim LGS, Santos CNO, Dos Santos PL, Tanajura DM, Lipscomb MW, de Jesus AR, de Almeida RP, de Moura TR. Antimony resistance associated with persistence of Leishmania (Leishmania) infantum infection in macrophages. Parasitol Res 2021; 120:2959-2964. [PMID: 34272999 DOI: 10.1007/s00436-021-07231-7] [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: 07/29/2020] [Accepted: 06/16/2021] [Indexed: 10/20/2022]
Abstract
Visceral leishmaniasis is a severe disease caused by protozoan parasites that include Leishmania (L.) infantum. The disease is established when parasites subvert the immune response of the host. Notably, chemotherapy-based use of antimonial compounds can partially alleviate disease burden. Unfortunately, the resistance to drug treatments is increasing in areas endemic to the disease. In this report, we investigated immune responses within macrophages infected with antimony-resistant L. infantum isolates from patients with a relapse in the disease. Results revealed that antimony-resistant parasites persist in the first 24 h of infection. Activation of macrophage or blocking of thiol production during infection shows enhanced clearance of parasites, which is coordinately associated with increased production of pro-inflammatory cytokines. Taken together, these results suggest that the mechanism of antimony resistance in L. infantum isolates may be related to a decrease in macrophage microbicidal functions.
Collapse
Affiliation(s)
- Lucas Sousa Magalhães
- Laboratory of Molecular Biology and Immunology, Federal University of Sergipe, Aracaju, Brazil
| | | | | | - Priscila Lima Dos Santos
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Brazil.,Department of Health Education, Federal University of Sergipe, Lagarto, Brazil
| | | | | | - Amélia Ribeiro de Jesus
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Brazil.,Instituto de Investigação Em Imunologia, São Paulo, Brazil
| | - Roque Pacheco de Almeida
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Brazil.,Instituto de Investigação Em Imunologia, São Paulo, Brazil
| | - Tatiana Rodrigues de Moura
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju, Brazil. .,Department of Morphology, Federal University of Sergipe, São Cristóvão, Brazil.
| |
Collapse
|
16
|
Modulation of Splenic B Cell Subsets during Experimental Leishmania donovani Infection in BALB/c Mice. Pathogens 2021; 10:pathogens10070814. [PMID: 34209841 PMCID: PMC8308600 DOI: 10.3390/pathogens10070814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/17/2021] [Accepted: 06/25/2021] [Indexed: 11/29/2022] Open
Abstract
Sodium antimonials are one of the major and common drugs used against visceral form leishmaniasis (VL). However, the development of drug resistance makes it difficult to manage this disease. Current work investigates the modulation of splenic B cells during experimental infection with antimony-sensitive and -resistant Leishmania donovani infection. Here we phenotypically characterized splenic B cell subsets in BALB/c mice infected with antimony drug-sensitive and -resistant VL strains using flow-cytometry method. In the splenocytes we noticed increased number of Transitional T3 B cells and B1a B cells in drug-resistant VL strain infection. Besides, we also observed alteration in Follicular B cell population of antimony-resistant strain infected mice. Drug-resistant strain induced secretion of elevated level of IL-10 from B1a B cells and IL-6 from Transitional T3 B cell subsets in the splenocytes. Purified splenic B cells from antimony drug-resistant strain infected mice showed decrease in the Lyn kinase gene expression compared to sensitive strain infected and uninfected mice. The current study provides insight into changes in host splenic B-cell subsets during experimental infection with antimony-sensitive and -resistant L. donovani in murine model.
Collapse
|
17
|
Role of chromatin modulation in the establishment of protozoan parasite infection for developing targeted chemotherapeutics. THE NUCLEUS 2021. [DOI: 10.1007/s13237-021-00356-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
18
|
Goel Y, Yadav S, Pandey SK, Temre MK, Singh VK, Kumar A, Singh SM. Methyl Jasmonate Cytotoxicity and Chemosensitization of T Cell Lymphoma In Vitro Is Facilitated by HK 2, HIF-1α, and Hsp70: Implication of Altered Regulation of Cell Survival, pH Homeostasis, Mitochondrial Functions. Front Pharmacol 2021; 12:628329. [PMID: 33716751 PMCID: PMC7954117 DOI: 10.3389/fphar.2021.628329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/19/2021] [Indexed: 12/31/2022] Open
Abstract
Methyl jasmonate (MJ) displays antineoplastic potential against numerous neoplastic cells. However, several mechanistic aspects of its antineoplastic action against malignancies of T cell origin remain elusive. The present investigation reports the novel targets of MJ and mechanistic pathways of MJ-mediated antineoplastic and chemosensitizing action against tumor cells derived from murine T-cell lymphoma, designated as Dalton's lymphoma (DL). The present study demonstrates that MJ directly docks to HIF-1α, hexokinase 2, and Hsp70 at prominent binding sites. MJ exhibits tumoricidal action against tumor cells via induction of apoptosis and necrosis through multiple pathways, including declined mitochondrial membrane potential, enhanced expression of ROS, altered pH homeostasis, an elevated level of cytosolic cytochrome c, and modulated expression of crucial cell survival and metabolism regulatory molecules. Additionally, this study also reports the chemosensitizing ability of MJ against T cell lymphoma accompanied by a declined expression of MDR1. This study sheds new light by demonstrating the implication of novel molecular mechanisms underlying the antitumor action of MJ against T-cell lymphoma and hence has immense translational significance.
Collapse
Affiliation(s)
- Yugal Goel
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Saveg Yadav
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Shrish Kumar Pandey
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Mithlesh Kumar Temre
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Vinay Kumar Singh
- Centre for Bioinformatics, School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ajay Kumar
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sukh Mahendra Singh
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
19
|
Barrow P, Dujardin JC, Fasel N, Greenwood AD, Osterrieder K, Lomonossoff G, Fiori PL, Atterbury R, Rossi M, Lalle M. Viruses of protozoan parasites and viral therapy: Is the time now right? Virol J 2020; 17:142. [PMID: 32993724 PMCID: PMC7522927 DOI: 10.1186/s12985-020-01410-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022] Open
Abstract
Infections caused by protozoan parasites burden the world with huge costs in terms of human and animal health. Most parasitic diseases caused by protozoans are neglected, particularly those associated with poverty and tropical countries, but the paucity of drug treatments and vaccines combined with increasing problems of drug resistance are becoming major concerns for their control and eradication. In this climate, the discovery/repurposing of new drugs and increasing effort in vaccine development should be supplemented with an exploration of new alternative/synergic treatment strategies. Viruses, either native or engineered, have been employed successfully as highly effective and selective therapeutic approaches to treat cancer (oncolytic viruses) and antibiotic-resistant bacterial diseases (phage therapy). Increasing evidence is accumulating that many protozoan, but also helminth, parasites harbour a range of different classes of viruses that are mostly absent from humans. Although some of these viruses appear to have no effect on their parasite hosts, others either have a clear direct negative impact on the parasite or may, in fact, contribute to the virulence of parasites for humans. This review will focus mainly on the viruses identified in protozoan parasites that are of medical importance. Inspired and informed by the experience gained from the application of oncolytic virus- and phage-therapy, rationally-driven strategies to employ these viruses successfully against parasitic diseases will be presented and discussed in the light of the current knowledge of the virus biology and the complex interplay between the viruses, the parasite hosts and the human host. We also highlight knowledge gaps that should be addressed to advance the potential of virotherapy against parasitic diseases.
Collapse
Affiliation(s)
- Paul Barrow
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Loughborough, Leicestershire, LE12 5RD, UK.
| | - Jean Claude Dujardin
- Molecular Parasitology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Nationalestraat, 155, 2000, Antwerpen, Belgium
| | - Nicolas Fasel
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Ch. des Boveresses 155, 1066, Epalinges, Switzerland
| | - Alex D Greenwood
- Department of Wildlife Diseases, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
- Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- Institut für Virologie, Robert Von Ostertag-Haus - Zentrum Fuer Infektionsmedizin, Robert von Ostertag-Str. 7-13, 14163, Berlin, Germany
| | - Klaus Osterrieder
- Institut für Virologie, Robert Von Ostertag-Haus - Zentrum Fuer Infektionsmedizin, Robert von Ostertag-Str. 7-13, 14163, Berlin, Germany
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, 31 To Yuen Street, Kowloon, Hong Kong
| | - George Lomonossoff
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Pier Luigi Fiori
- Dipartimento Di Scienze Biomedice, Universita Degli Studi Di Sassari, Sardinia, Italy
| | - Robert Atterbury
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Loughborough, Leicestershire, LE12 5RD, UK
| | - Matteo Rossi
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Ch. des Boveresses 155, 1066, Epalinges, Switzerland
| | - Marco Lalle
- Unit of Foodborne and Neglected Parasitic Diseases, European Union Reference Laboratory for Parasites, Department of Infectious Diseases, Istituto Superiore Di Sanità, viale Regina Elena 299, 00186, Rome, Italy.
| |
Collapse
|
20
|
Kröber-Boncardo C, Lorenzen S, Brinker C, Clos J. Casein kinase 1.2 over expression restores stress resistance to Leishmania donovani HSP23 null mutants. Sci Rep 2020; 10:15969. [PMID: 32994468 PMCID: PMC7525241 DOI: 10.1038/s41598-020-72724-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/04/2020] [Indexed: 01/25/2023] Open
Abstract
Leishmania donovani is a trypanosomatidic parasite and causes the lethal kala-azar fever, a neglected tropical disease. The Trypanosomatida are devoid of transcriptional gene regulation and rely on gene copy number variations and translational control for their adaption to changing conditions. To survive at mammalian tissue temperatures, L. donovani relies on the small heat shock protein HSP23, the loss of which renders the parasites stress sensitive and impairs their proliferation. Here, we analysed a spontaneous escape mutant with wild type-like in vitro growth. Further selection of this escape strains resulted in a complete reversion of the phenotype. Whole genome sequencing revealed a correlation between stress tolerance and the massive amplification of a six-gene cluster on chromosome 35, with further analysis showing over expression of the casein kinase 1.2 gene as responsible. In vitro phosphorylation experiments established both HSP23 and the related P23 co-chaperone as substrates and modulators of casein kinase 1.2, providing evidence for another crucial link between chaperones and signal transduction protein kinases in this early branching eukaryote.
Collapse
Affiliation(s)
- Constanze Kröber-Boncardo
- Leishmania Group, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht St 74, 20359, Hamburg, Germany
| | - Stephan Lorenzen
- Department of Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Christine Brinker
- Leishmania Group, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht St 74, 20359, Hamburg, Germany
| | - Joachim Clos
- Leishmania Group, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht St 74, 20359, Hamburg, Germany.
| |
Collapse
|
21
|
Mukherjee B, Mukherjee K, Nanda P, Mukhopadhayay R, Ravichandiran V, Bhattacharyya SN, Roy S. Probing the molecular mechanism of aggressive infection by antimony resistant Leishmania donovani. Cytokine 2020; 145:155245. [PMID: 32861564 DOI: 10.1016/j.cyto.2020.155245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/12/2020] [Accepted: 08/08/2020] [Indexed: 12/17/2022]
Abstract
The disease visceral leishmaniasis (VL) or kala azar is caused by the protozoan parasite, Leishmania donovani (LD). For many decades the pentavalent antimonial drugs countered the successive epidemics of the disease in the Indian sub-continent and elsewhere. With time, antimony resistant LD (LDR) developed and the drug in turn lost its efficacy. Infection of mammals with LDR gives rise to aggressive infection as compared to its sensitive counterpart (LDS) coupled with higher surge of IL-10 and TGF-β. The IL-10 causes upregulation of multidrug resistant protein-1 which causes efflux of antimonials from LDR infected cells. This is believed to be a key mechanism of antimony resistance. MicroRNAs (miRNAs) are tiny post-transcriptional regulators of gene expression in mammalian cells and in macrophage play a pivotal role in controlling the expression of cytokines involved in infection process. Therefore, a change in miRNA profiles of macrophages infected with LDS or LDR could explain the differential cytokine response observed. Interestingly, the outcome of LD infection is also governed by the critical balance of pro- and anti-inflammatory cytokines which is inturn regulated by miRNA-Ago2 or miRNP complex and its antagonist RNA binding protein HuR. Here Ago2 plays the fulcrum whose phosphorylation and de-phosphorylation dictates the process; which in turn is controlled by PP2A and HuR. LDS and LDR upregulate PP2A and downregulate HuR at different magnitude leading to various levels of anti-inflammatory to proinflammatory cytokine production and resulting pathology in the host. While ectopic HuR expression alone is sufficient to clear LDS infection, simultaneous upregulation of HuR and inhibition of PP2A is required to inhibit LDR mediated infection. Therefore, tampering with miRNA pathway could be a new strategy to control infection caused by LDR parasite.
Collapse
Affiliation(s)
- Budhaditya Mukherjee
- CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India; School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, India
| | | | - Piyush Nanda
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, India
| | | | - V Ravichandiran
- National Institute of Pharmaceutical Education & Research, Kolkata 700054, India
| | | | - Syamal Roy
- CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India; National Institute of Pharmaceutical Education & Research, Kolkata 700054, India.
| |
Collapse
|
22
|
Combination of Mycobacterium indicus pranii and Heat-Induced Promastigotes Cures Drug-Resistant Leishmania Infection: Critical Role of Interleukin-6-Producing Classical Dendritic Cells. Infect Immun 2020; 88:IAI.00222-19. [PMID: 32229617 DOI: 10.1128/iai.00222-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/24/2020] [Indexed: 12/18/2022] Open
Abstract
The major issues in available therapeutic modalities against leishmaniasis are cost, toxicity, and the emergence of drug resistance. The aim of this work was to develop a successful therapeutic adjuvant against drug-resistant Leishmania donovani infection by means of combining Mycobacterium indicus pranii with heat-induced promastigotes (HIP). One-month postinfected BALB/c mice were administered subcutaneously with M. indicus pranii (108 cells) and HIP (100 μg) for 5 days. Spleens were harvested for flow cytometric and reverse transcriptase PCR analysis. The antileishmanial effect of the combination strategy was associated with induction of a disease-resolving Th1 and Th17 response with simultaneous downregulation of CD4+ CD25+ Foxp3+ (nTreg) cells and CD4+ CD25- Foxp3- (Tr1) cells in the spleen. The significant expansion of CD4+ TCM (CD4+ CD44hi CD11ahi CD62Lhi) cells was a further interesting outcome of this therapeutic strategy in the context of long-term protection of hosts against secondary infection. Toll-like receptor 2 (TLR2) was also found instrumental in this antiparasitic therapy. Induced interleukin-6 (IL-6) production from expanded CD11c+ CD8α+ (cDC1) and CD11c+ CD11b+ (cDC2) dendritic cells (DCs) but not from the CD11b+ Ly6c+ inflammatory monocytes (iMOs), was found critical in the protective expansion of Th17 as evidenced by an in vivo IL-6 neutralization assay. It also promoted the hematopoietic conversion toward DC progenitors (pre-DCs) from common dendritic cell progenitors (CDPs), the immediate precursors, in bone marrow. This novel combinational strategy demonstrated that expansion of Th17 by IL-6 released from CD11c+ classical DCs is crucial, together with the conventional Th1 response, to control drug-resistant infection.
Collapse
|
23
|
Goswami A, Mukherjee K, Mazumder A, Ganguly S, Mukherjee I, Chakrabarti S, Roy S, Sundar S, Chattopadhyay K, Bhattacharyya SN. MicroRNA exporter HuR clears the internalized pathogens by promoting pro-inflammatory response in infected macrophages. EMBO Mol Med 2020; 12:e11011. [PMID: 32031337 PMCID: PMC7059013 DOI: 10.15252/emmm.201911011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 12/24/2019] [Accepted: 01/08/2020] [Indexed: 01/10/2023] Open
Abstract
HuR is a miRNA derepressor protein that can act as miRNA sponge for specific miRNAs to negate their action on target mRNAs. Here we have identified how HuR, by inducing extracellular vesicles‐mediated export of miRNAs, ensures robust derepression of miRNA‐repressed cytokines essential for strong pro‐inflammatory response in activated mammalian macrophages. Leishmania donovani, the causative agent of visceral leishmaniasis, on the contrary alters immune response of the host macrophage by a variety of complex mechanisms to promote anti‐inflammatory response essential for the survival of the parasite. We have found that during Leishmania infection, the pathogen targets HuR to promote onset of anti‐inflammatory response in mammalian macrophages. In infected macrophages, Leishmania also upregulate protein phosphatase 2A that acts on Ago2 protein to keep it in dephosphorylated and miRNA‐associated form. This causes robust repression of the miRNA‐targeted pro‐inflammatory cytokines to establish an anti‐inflammatory response in infected macrophages. HuR has an inhibitory effect on protein phosphatase 2A expression, and mathematical modelling of macrophage activation process supports antagonistic miRNA‐modulatory roles of HuR and protein phosphatase 2A which mutually balances immune response in macrophage by targeting miRNA function. Supporting this model, ectopic expression of the protein HuR and simultaneous inhibition of protein phosphatase 2A induce strong pro‐inflammatory response in the host macrophage to prevent the virulent antimonial drug‐sensitive or drug‐resistant form of L. donovani infection. Thus, HuR can act as a balancing factor of immune responses to curtail the macrophage infection process by the protozoan parasite.
Collapse
Affiliation(s)
- Avijit Goswami
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Kamalika Mukherjee
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Anup Mazumder
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Satarupa Ganguly
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Ishita Mukherjee
- Structural Biology and Bio-informatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Saikat Chakrabarti
- Structural Biology and Bio-informatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Syamal Roy
- National Institute of Pharmaceutical Educations and Research, Kolkata, India
| | - Shyam Sundar
- Department of Medicine, Banaras Hindu University, Varanasi, India
| | - Krishnananda Chattopadhyay
- Structural Biology and Bio-informatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Suvendra N Bhattacharyya
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
24
|
Patino LH, Muskus C, Ramírez JD. Transcriptional responses of Leishmania (Leishmania) amazonensis in the presence of trivalent sodium stibogluconate. Parasit Vectors 2019; 12:348. [PMID: 31300064 PMCID: PMC6626383 DOI: 10.1186/s13071-019-3603-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/06/2019] [Indexed: 12/13/2022] Open
Abstract
Background In the last decade, resistance to antimonials has become a serious problem due to the emergence of drug-resistant strains. Therefore, understanding the mechanisms used by Leishmania parasites to survive under drug pressure is essential, particularly for species of medical-veterinary importance such as L. amazonensis. Methods Here, we used RNA-seq technology to analyse transcriptome profiles and identify global changes in gene expression between antimony-resistant and -sensitive L. amazonensis promastigotes. Results A total of 723 differentially expressed genes were identified between resistant and sensitive lines. Comparative transcriptomic analysis revealed that genes encoding proteins involved in metabolism (fatty acids) and stress response, as well as those associated with antimony resistance in other Leishmania species, were upregulated in the antimony-resistant line. Most importantly, we observed upregulation of genes encoding autophagy proteins, suggesting that in the presence of trivalent stibogluconate (SbIII) L. amazonensis can activate these genes either as a survival strategy or to induce cell death, as has been observed in other parasites. Conclusions This work identified global transcriptomic changes in an in vitro-adapted strain in response to SbIII. Our results provide relevant information to continue understanding the mechanism used by parasites of the subgenus Leishmania (L. amazonensis) to generate an antimony-resistant phenotype. Electronic supplementary material The online version of this article (10.1186/s13071-019-3603-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Luz H Patino
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Carlos Muskus
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Juan David Ramírez
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
25
|
Nucleic acid sensing activates the innate cytosolic surveillance pathway and promotes parasite survival in visceral leishmaniasis. Sci Rep 2019; 9:9825. [PMID: 31285443 PMCID: PMC6614394 DOI: 10.1038/s41598-019-45800-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/12/2019] [Indexed: 01/08/2023] Open
Abstract
Microbial pattern recognition critically contributes to innate response, both at extracellular and intracellular cytosolic surveillance pathway (CSP) interface. However, the role of pattern recognition by host innate receptors in CSP is poorly understood in Leishmania donovani infection. Here, we have demonstrated that cytosolic targeting of L.donovani DNA (Ld-DNA) inhibits macrophage responsiveness to IFNɣ, through decreased MHC-II expression and lowered pSTAT1 (Y701) levels, involving host three-prime repair exonuclease-1 (TREX-1). The Ld-DNA potently induced type-1 IFNs, i.e. significant over-production of IFNβ through activation of the IRF pathway. Interestingly, knockdown of TRIF or MyD88 expression in macrophages had no effect on cytosolic Ld-DNA transfection-mediated IFN-β production, indicating involvement of a TLR independent pathway. Contrastingly, Ld-DNA failed to induce IFNβ in both TBK-1 and IRF3KO knockout macrophages. Although IFNβ was not induced by Ld-DNA in STING- knockout macrophages, STING alone was not enough for the induction. Evidently, besides STING, Ld-DNA recognition for induction of IFNβ critically required cytosolic cyclic GMP-AMP synthase (cGAS). Furthermore, the cGAS dependent targeting of Ld-DNA induced IFNβ over-production that contributed to antimony resistance in L.donovani infection. We provide the first evidence that enhanced cytosolic sensing of Ld-DNA in infection by antimony resistant (SBR-LD), but not antimony sensitive L.donovani strains (SBS-LD), was critically regulated by host MDRs, multi drug resistant associated protein 1 (MRP 1) and permeability glycoprotein (P-gp) in macrophages. Collectively, our results disclose Ld-DNA as a vital pathogen associated molecular pattern (PAMP) driving host Type-I IFN responses and antimony resistance. The findings may help in future development of policies for novel anti-leishmanial therapeutics.
Collapse
|
26
|
Chauhan K, Kaur G, Kaur S. Activity of rutin, a potent flavonoid against SSG-sensitive and -resistant Leishmania donovani parasites in experimental leishmaniasis. Int Immunopharmacol 2018; 64:372-385. [DOI: 10.1016/j.intimp.2018.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/02/2018] [Accepted: 09/17/2018] [Indexed: 12/20/2022]
|
27
|
Lipase Precursor-Like Protein Promotes Miltefosine Tolerance in Leishmania donovani by Enhancing Parasite Infectivity and Eliciting Anti-inflammatory Responses in Host Macrophages. Antimicrob Agents Chemother 2018; 62:AAC.00666-18. [PMID: 30297367 DOI: 10.1128/aac.00666-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 09/28/2018] [Indexed: 01/02/2023] Open
Abstract
The oral drug miltefosine (MIL) was introduced in the Indian subcontinent in the year 2002 for the treatment of visceral leishmaniasis (VL). However, recent reports on its declining efficacy and increasing relapse rates pose a serious concern. An understanding of the factors contributing to MIL tolerance in Leishmania parasites is critical. In the present study, we assessed the role of the lipase precursor-like protein (Lip) in conferring tolerance to miltefosine by episomally overexpressing Lip in Leishmania donovani (LdLip++). We observed a significant increase (∼3-fold) in the MIL 50% inhibitory concentration (IC50) at both the promastigote (3.90 ± 0.68 µM; P < 0.05) and intracellular amastigote (9.10 ± 0.60 µM; P < 0.05) stages compared to the wild-type counterpart (LdNeo) (MIL IC50s of 1.49 ± 0.20 µM at the promastigote stage and 3.95 ± 0.45 µM at the amastigote stage). LdLip++ parasites exhibited significantly (P < 0.05) increased infectivity to host macrophages and increased metacyclogenesis and tolerance to MIL-induced oxidative stress. The susceptibility of LdLip++ to other antileishmanial drugs (sodium antimony gluconate and amphotericin B) remained unchanged. In comparison to LdNeo, the LdLip++ parasites elicited high host interleukin-10 (IL-10) cytokine expression levels (1.6-fold; P < 0.05) with reduced expression of the cytokine tumor necrosis factor alpha (TNF-α) (1.5-fold; P < 0.05), leading to a significantly (P < 0.01) increased ratio of IL-10/TNF-α. The above-described findings suggest a role of lipase precursor-like protein in conferring tolerance to the oral antileishmanial drug MIL in L. donovani parasites.
Collapse
|
28
|
Rugani JN, Quaresma PF, Gontijo CF, Soares RP, Monte-Neto RL. Intraspecies susceptibility of Leishmania (Viannia) braziliensis to antileishmanial drugs: Antimony resistance in human isolates from atypical lesions. Biomed Pharmacother 2018; 108:1170-1180. [PMID: 30372818 DOI: 10.1016/j.biopha.2018.09.149] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
Abstract
Leishmania (Viannia) braziliensis is the most common etiological agent of cutaneous and mucocutaneous leishmaniasis (MCL) in Latin America. An interesting aspect of the disease outcome caused by this species is the appearance of non-ulcerated atypical cutaneous leishmaniasis. Atypical (AT) lesions are often associated with therapeutic failure when treated with antimony(Sb)-based drugs. Refractory cases are not necessarily due to intrinsic parasite drug resistance. The status of in vitro drug susceptibility from L. braziliensis field isolates is less assessed than patient treatment outcome. In this work, L. braziliensis isolated from typical CL (6), MCL (1) and AT (3) lesions and vector (1) were tested for their susceptibility to amphotericin B (AmB), miltefosine (MIL), glucantime (GLU) and non-comercial meglumine antimoniate (MA). Overall, intracellular amastigotes of all isolates were sensitive to the tested antileishmanial drugs except AT lesions-derived strains 316, 330 and 340 that presented in vitro resistance against SbV-based drugs. Although susceptible to miltefosine - based on phenotypic screening - intramacrophagic quiescent amastigotes could restore infection. L. braziliensis promastigotes isolated from AT lesions also displayed 29% reduced capacity to infect human monocyte-derived macrophages when compared with parasites obtained from patients with typical lesions, MCL or from sand-fly. These data indicate differences in drug susceptibility and infectiveness among L. braziliensis isolated from patients exhibiting different types of lesions and highlight the importance of its characterization for drug response prediction outcome in clinical practice.
Collapse
Affiliation(s)
- Jeronimo N Rugani
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, 30190-009, Belo Horizonte, MG, Brazil.
| | - Patrícia F Quaresma
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, 30190-009, Belo Horizonte, MG, Brazil.
| | - Célia F Gontijo
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, 30190-009, Belo Horizonte, MG, Brazil.
| | - Rodrigo P Soares
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, 30190-009, Belo Horizonte, MG, Brazil.
| | - Rubens L Monte-Neto
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, 30190-009, Belo Horizonte, MG, Brazil.
| |
Collapse
|
29
|
Importance of secondary screening with clinical isolates for anti-leishmania drug discovery. Sci Rep 2018; 8:11765. [PMID: 30082744 PMCID: PMC6078976 DOI: 10.1038/s41598-018-30040-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/10/2018] [Indexed: 11/17/2022] Open
Abstract
The growing drug resistance (DR) raises major concerns for the control of visceral leishmaniasis (VL), a neglected disease lethal in 95 percent of the cases if left untreated. Resistance has rendered antimonials (SSG) obsolete in the Indian Sub-Continent (ISC) and the first miltefosine-resistant Leishmania donovani were isolated. New chemotherapeutic options are needed and novel compounds are being identified by high-throughput screening (HTS). HTS is generally performed with old laboratory strains such as LdBOB and we aimed here to validate the activity of selected compounds against recent clinical isolates. In this academic/industrial collaboration, 130 compounds from the GSK “Leishbox” were screened against one SSG-sensitive and one SSG-resistant strain of L. donovani recently isolated from ISC patients, using an intracellular assay of L. donovani-infected THP1-derived macrophages. We showed that only 45% of the compounds were active in both clinical isolates and LdBOB. There were also different compound efficiencies linked to the SSG susceptibility background of the strains. In addition, our results suggested that the differential susceptibility profiles were chemical series-dependent. In conclusion, we demonstrate the potential value of including clinical isolates (as well as resistant strains) in the HTS progression cascade.
Collapse
|
30
|
Magalhães LS, Bomfim LG, Mota SG, Cruz GS, Corrêa CB, Tanajura DM, Lipscomb MW, Borges VM, Jesus ARD, Almeida RPD, Moura TRD. Increased thiol levels in antimony-resistant Leishmania infantum isolated from treatment-refractory visceral leishmaniasis in Brazil. Mem Inst Oswaldo Cruz 2018; 113:119-125. [PMID: 29236925 PMCID: PMC5722267 DOI: 10.1590/0074-02760170289] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/06/2017] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Treatment-refractory visceral leishmaniasis (VL) has become an important
problem in many countries. OBJECTIVES We evaluated the antimony-resistance mechanisms of Leishmania
infantum isolated from VL patients refractory or responsive to
treatment with pentavalent antimony. METHODS Strains isolated from antimony-refractory patients (in vitro
antimony-resistant isolates) and antimony-responsive patients (in
vitro antimony-sensitive isolates) were examined. Morphological
changes were evaluated by transmission electron microscopy after trivalent
antimony exposure. P-glycoprotein (P-gp) efflux pump activity was evaluated
using the pump-specific inhibitor verapamil hydrochloride, and the role of
thiol in trivalent antimony resistance was investigated using the enzymatic
inhibitor L-buthionine sulfoximine. FINDINGS Antimony treatment induced fewer alterations in the cellular structure of
L. infantum resistant isolates than in that of
sensitive isolates. P-gp efflux activity was not involved in antimony
resistance in these isolates. Importantly, the resistant isolates contained
higher levels of thiol compared to the sensitive isolates, and inhibition of
thiol synthesis in the resistant isolates recovered their sensitivity to
trivalent antimony treatment, and enhanced the production of reactive oxygen
species in promastigotes exposed to the drug. MAIN CONCLUSIONS Our results demonstrate that isolates from patients with antimony-refractory
VL exhibited higher thiol levels than antimony-sensitive isolates. This
indicates that redox metabolism plays an important role in the
antimony-resistance of New World VL isolates.
Collapse
Affiliation(s)
- Lucas S Magalhães
- Laboratório de Biologia Molecular, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Lays Gs Bomfim
- Laboratório de Biologia Molecular, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Sthefanne G Mota
- Laboratório de Biologia Molecular, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Geydson S Cruz
- Laboratório de Biologia Molecular, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Cristiane B Corrêa
- Laboratório de Biologia Molecular, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Diego M Tanajura
- Laboratório de Biologia Molecular, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Michael W Lipscomb
- Department of Biology, Howard University, Washington DC, United States of America
| | - Valéria M Borges
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz-Fiocruz, Salvador, BA, Brasil
| | - Amélia R de Jesus
- Laboratório de Biologia Molecular, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Roque P de Almeida
- Laboratório de Biologia Molecular, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Tatiana R de Moura
- Laboratório de Biologia Molecular, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| |
Collapse
|
31
|
Ponte-Sucre A, Gamarro F, Dujardin JC, Barrett MP, López-Vélez R, García-Hernández R, Pountain AW, Mwenechanya R, Papadopoulou B. Drug resistance and treatment failure in leishmaniasis: A 21st century challenge. PLoS Negl Trop Dis 2017; 11:e0006052. [PMID: 29240765 PMCID: PMC5730103 DOI: 10.1371/journal.pntd.0006052] [Citation(s) in RCA: 521] [Impact Index Per Article: 74.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Reevaluation of treatment guidelines for Old and New World leishmaniasis is urgently needed on a global basis because treatment failure is an increasing problem. Drug resistance is a fundamental determinant of treatment failure, although other factors also contribute to this phenomenon, including the global HIV/AIDS epidemic with its accompanying impact on the immune system. Pentavalent antimonials have been used successfully worldwide for the treatment of leishmaniasis since the first half of the 20th century, but the last 10 to 20 years have witnessed an increase in clinical resistance, e.g., in North Bihar in India. In this review, we discuss the meaning of “resistance” related to leishmaniasis and discuss its molecular epidemiology, particularly for Leishmania donovani that causes visceral leishmaniasis. We also discuss how resistance can affect drug combination therapies. Molecular mechanisms known to contribute to resistance to antimonials, amphotericin B, and miltefosine are also outlined. Chemotherapy is central to the control and management of leishmaniasis. Antimonials remain the primary drugs against different forms of leishmaniasis in several regions. However, resistance to antimony has necessitated the use of alternative medications, especially in the Indian subcontinent (ISC). Compounds, notably the orally available miltefosine (MIL), parenteral paromomycin, and amphotericin B (AmB), are increasingly used to treat leishmaniasis. Although treatment failure (TF) has been observed in patients treated with most anti-leishmanials, its frequency of appearance may be important in patients treated with MIL, which has replaced antimonials within the kala-azar elimination program in the ISC. AmB is highly efficacious, and the associated toxic effects—when administered in its free deoxycholate form—are somewhat ameliorated in its liposomal formulation. Regrettably, laboratory experimentation has demonstrated a risk of resistance towards AmB as well. The rise of drug resistance impacts treatment outcome, and understanding its causes, spread, and impact will help us manage the risks it imposes. Here, we review the problem of TF in leishmaniasis and the contribution of drug resistance to the problem. Molecular mechanisms causing resistance to anti-leishmanials are discussed along with the appropriate use of additional available drugs, as well as the urgent need to consolidate strategies to monitor drug efficacy, epidemiological surveillance, and local policies. Coordination of these activities in national and international programs against leishmaniasis might represent a successful guide to further research and prevention activities.
Collapse
Affiliation(s)
- Alicia Ponte-Sucre
- Department of Physiological Sciences, Laboratory of Molecular Physiology, Institute of Experimental Medicine, Luis Razetti School of Medicine, Universidad Central de Venezuela, Caracas, Venezuela
- * E-mail: (BP); (APS)
| | - Francisco Gamarro
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López-Neyra, Spanish National Research Council (IPBLN-CSIC), Granada, Spain
| | - Jean-Claude Dujardin
- Molecular Parasitology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Michael P. Barrett
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Rogelio López-Vélez
- Department of Infectious Diseases, National Referral Unit for Tropical Diseases, Ramón y Cajal University Hospital, Madrid, Spain
| | - Raquel García-Hernández
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López-Neyra, Spanish National Research Council (IPBLN-CSIC), Granada, Spain
| | - Andrew W. Pountain
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Roy Mwenechanya
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
| | - Barbara Papadopoulou
- Research Center in Infectious Diseases, CHU de Quebec Research Center and Department of Microbiology-Infectious Disease and Immunology, University Laval, Quebec, Canada
- * E-mail: (BP); (APS)
| |
Collapse
|
32
|
Profiling gene expression of antimony response genes in Leishmania (Viannia) panamensis and infected macrophages and its relationship with drug susceptibility. Acta Trop 2017; 176:355-363. [PMID: 28843396 DOI: 10.1016/j.actatropica.2017.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 08/04/2017] [Accepted: 08/17/2017] [Indexed: 12/31/2022]
Abstract
The mechanisms of Leishmania resistance to antimonials have been primarily determined in experimentally derived Leishmania strains. However, their participation in the susceptibility phenotype in field isolates has not been conclusively established. Being an intracellular parasite, the activity of antileishmanials is dependent on internalization of drugs into host cells and effective delivery to the intracellular compartments inhabited by the parasite. In this study we quantified and comparatively analyzed the gene expression of nine molecules involved in mechanisms of xenobiotic detoxification and Leishmania resistance to antimonial drugs in resistant and susceptible laboratory derived and clinical L.(Viannia) panamensis strains(n=19). In addition, we explored the impact of Leishmania susceptibility to antimonials on the expression of macrophage gene products having putative functions in transport, accumulation and metabolism of antimonials. As previously shown for other Leishmania species, a trend of increased abcc3 and lower aqp-1 expression was observed in the laboratory derived Sb-resistant L.(V.) panamensis line. However, this was not found in clinical strains, in which the expression of abca2 was significantly higher in resistant strains as both, promastigotes and intracellular amastigotes. The effect of drug susceptibility on host cell gene expression was evaluated on primary human macrophages from patients with cutaneous leishmaniasis (n=17) infected ex-vivo with the matched L.(V.) panamensis strains isolated at diagnosis, and in THP-1 cells infected with clinical strains (n=6) and laboratory adapted L.(V.) panamensis lines. Four molecules, abcb1 (p-gp), abcb6, aqp-9 and mt2a were differentially modulated by drug resistant and susceptible parasites, and among these, a consistent and significantly increased expression of the xenobiotic scavenging molecule mt2a was observed in macrophages infected with Sb-susceptible L. (V.) panamensis. Our results substantiate that different mechanisms of drug resistance operate in laboratory adapted and clinical Leishmania strains, and provide evidence that parasite-mediated modulation of host cell gene expression of molecules involved in drug transport and metabolism could contribute to the mechanisms of drug resistance and susceptibility in Leishmania.
Collapse
|
33
|
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: 3.1] [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
|
34
|
Verma S, Das S, Mandal A, Ansari MY, Kumari S, Mansuri R, Kumar A, Singh R, Saini S, Abhishek K, Kumar V, Sahoo GC, Das P. Role of inhibitors of serine peptidases in protecting Leishmania donovani against the hydrolytic peptidases of sand fly midgut. Parasit Vectors 2017; 10:303. [PMID: 28645315 PMCID: PMC5481909 DOI: 10.1186/s13071-017-2239-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/11/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In vector-borne diseases such as leishmaniasis, the sand fly midgut is considered to be an important site for vector-parasite interaction. Digestive enzymes including serine peptidases such as trypsin and chymotrypsin, which are secreted in the midgut are one of the obstacles for Leishmania in establishing a successful infection. The presence of some natural inhibitors of serine peptidases (ISPs) has recently been reported in Leishmania. In the present study, we deciphered the role of these ISPs in the survival of Leishmania donovani in the hostile sand fly midgut environment. METHODS In silico and co-immunoprecipitation studies were performed to observe the interaction of L. donovani ISPs with trypsin and chymotrypsin. Zymography and in vitro enzyme assays were carried out to observe the inhibitory effect of purified recombinant ISPs of L. donovani (rLdISPs) on trypsin, chymotrypsin and the sand fly midgut peptidases. The expression of ISPs in the amastigote to promastigote transition stages were studied by semi-quantitative RT-PCR and Western blot. The role of LdISP on the survival of ISP overexpressed (OE) and ISP knocked down (KD) Leishmania parasites inside the sand fly gut was investigated by in vitro and in vivo cell viability assays. RESULTS We identified two ecotin-like genes in L. donovani, LdISP1 and LdISP2. In silico and co-immunoprecipitation results clearly suggest a strong interaction of LdISP molecules with trypsin and chymotrypsin. Zymography and in vitro enzyme assay confirmed the inhibitory effect of rLdISP on trypsin, chymotrypsin and the sand fly midgut peptidases. The expression of LdISP2 was found to be strongly associated with the amastigote to promastigote phase transition. The activities of the digestive enzymes were found to be significantly reduced in the infected sand flies when compared to uninfected. To our knowledge, our study is the first report showing the possible reduction of chymotrypsin activity in L. donovani infected sand flies compared to uninfected. Interestingly, during the early transition stage, substantial killing was observed in ISP2 knocked down (ISP2KD) parasites compared to wild type (WT), whereas ISP1 knocked down (ISP1KD) parasites remained viable. Therefore, our study clearly indicates that LdISP2 is a more effective inhibitor of serine peptidases than LdISP1. CONCLUSION Our results suggest that the lack of ISP2 is detrimental to the parasites during the early transition from amastigotes to promastigotes. Moreover, the results of the present study demonstrated for the first time that LdISP2 has an important role in the inhibition of peptidases and promoting L. donovani survival inside the Phlebotomus argentipes midgut.
Collapse
Affiliation(s)
- Sudha Verma
- Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Agamkuan, Patna, Bihar 800007 India
| | - Sushmita Das
- Department of Microbiology, All India Institute of Medical Sciences, Patna, Bihar 801105 India
| | - Abhishek Mandal
- Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Agamkuan, Patna, Bihar 800007 India
| | - Md Yousuf Ansari
- National Institute of Pharmaceutical Education and Research, Hajipur, Bihar 844101 India
- MM College of Pharmacy, Maharishi Markandeshwar University, Mullana, Ambala, 133207 India
| | - Sujata Kumari
- Department of Vector Biology, Rajendra Memorial Research Institute of Medical Sciences, (ICMR), Agamkuan, Patna, Bihar 800007 India
| | - Rani Mansuri
- National Institute of Pharmaceutical Education and Research, Hajipur, Bihar 844101 India
| | - Ajay Kumar
- Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Agamkuan, Patna, Bihar 800007 India
| | - Ruby Singh
- Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Agamkuan, Patna, Bihar 800007 India
| | - Savita Saini
- National Institute of Pharmaceutical Education and Research, Hajipur, Bihar 844101 India
| | - Kumar Abhishek
- Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Agamkuan, Patna, Bihar 800007 India
| | - Vijay Kumar
- Department of Vector Biology, Rajendra Memorial Research Institute of Medical Sciences, (ICMR), Agamkuan, Patna, Bihar 800007 India
| | - Ganesh Chandra Sahoo
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Agamkuan, Patna, Bihar 800007 India
| | - Pradeep Das
- Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Agamkuan, Patna, Bihar 800007 India
| |
Collapse
|
35
|
Chowdhury SR, Kumar A, Godinho JLP, De Macedo Silva ST, Zuma AA, Saha S, Kumari N, Rodrigues JCF, Sundar S, Dujardin JC, Roy S, De Souza W, Mukhopadhyay S, Majumder HK. Voacamine alters Leishmania ultrastructure and kills parasite by poisoning unusual bi-subunit topoisomerase IB. Biochem Pharmacol 2017; 138:19-30. [PMID: 28483460 DOI: 10.1016/j.bcp.2017.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/03/2017] [Indexed: 01/01/2023]
Abstract
Indole alkaloids possess a large spectrum of biological activities including anti-protozoal action. Here we report for the first time that voacamine, isolated from the plant Tabernaemontana coronaria, is an antiprotozoal agent effective against a large array of trypanosomatid parasites including Indian strain of Leishmania donovani and Brazilian strains of Leishmania amazonensis and Trypanosoma cruzi. It inhibits the relaxation activity of topoisomerase IB of L. donovani (LdTop1B) and stabilizes the cleavable complex. Voacamine is probably the first LdTop1B-specific poison to act uncompetitively. It has no impact on human topoisomerase I and II up to 200μM concentrations. The study also provides a thorough insight into ultrastructural alterations induced in three kinetoplastid parasites by a specific inhibitor of LdTop1B. Voacamine is also effective against intracellular amastigotes of different drug unresponsive field isolates of Leishmania donovani obtained from endemic zones of India severely affected with visceral leishmaniasis. Most importantly, this is the first report demonstrating the efficacy of a compound to reduce the burden of drug resistant parasites, unresponsive to SAG, amphotericin B and miltefosine, in experimental BALB/c mice model of visceral leishmaniasis. The findings cumulatively provide a strong evidence that voacamine can be a promising drug candidate against trypanosomatid infections.
Collapse
Affiliation(s)
- Somenath Roy Chowdhury
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Ashish Kumar
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Joseane Lima Prado Godinho
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro 21941-902, Brazil
| | - Sara Teixeira De Macedo Silva
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro 21941-902, Brazil
| | - Aline Araujo Zuma
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro 21941-902, Brazil
| | - Sourav Saha
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Neha Kumari
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Juliany Cola Fernandes Rodrigues
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro 21941-902, Brazil
| | - Shyam Sundar
- Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Jean-Claude Dujardin
- Department of Parasitology, Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Syamal Roy
- Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal 726 101, India
| | - Wanderley De Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro 21941-902, Brazil
| | - Sibabrata Mukhopadhyay
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Hemanta K Majumder
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India.
| |
Collapse
|
36
|
DeMars KM, Yang C, Hawkins KE, McCrea AO, Siwarski DM, Candelario-Jalil E. Spatiotemporal Changes in P-glycoprotein Levels in Brain and Peripheral Tissues Following Ischemic Stroke in Rats. J Exp Neurosci 2017; 11:1179069517701741. [PMID: 28469478 PMCID: PMC5398227 DOI: 10.1177/1179069517701741] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/02/2017] [Indexed: 01/19/2023] Open
Abstract
P-glycoprotein (P-gp) is known to transport a diverse array of xenobiotics, including therapeutic drugs. A member of the ATP-binding cassette (ABC) transporter family, P-gp is a protein encoded by the gene Mdr1 in humans and Abcb1 in rodents (represented by 2 isoforms Abcb1a and Abcb1b). Lining the luminal and abluminal membrane of brain capillary endothelial cells, P-gp is a promiscuous efflux pump extruding a variety of exogenous toxins and drugs. In this study, we measured dynamic changes in Abcb1a and Abcb1b transcripts and P-gp protein in the brain, liver, and kidney after experimental stroke. P-glycoprotein has been shown to increase in brain endothelial cells following hypoxia in vitro or after exposure to proinflammatory cytokines. Using a rat model of ischemic stroke, we hypothesized that P-gp expression will be increased in the brain, liver, and kidney in response to neuroinflammation following ischemic stroke. Adult Sprague Dawley rats underwent middle cerebral artery occlusion (MCAO) for 90 minutes and were killed at 4, 14, 24, and 48 hours postreperfusion onset to determine the time course of P-gp expression. To mimic ischemia occurring at the blood-brain barrier, rat brain endothelial (RBE4) cells were subjected to hypoxia and low glucose (HLG) for 16 hours. Immunoblotting analyses showed P-gp increases in brain and liver following 90-minute MCAO, as well as in cultured RBE4 cells after 16-hour HLG treatment, but fluctuated in the kidney depending on the time point. The relative roles of each isoform in the protein expression were analyzed with quantitative reverse transcriptase polymerase chain reaction. Ischemic stroke leads to significant increases in P-gp levels not only in the brain but also in the liver. The increase in P-gp could dramatically reduce the bioavailability and efficacy of neuroprotective drugs. Therefore, P-gp represents a big hurdle to drug delivery to the ischemic brain.
Collapse
Affiliation(s)
- Kelly M DeMars
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kimberly E Hawkins
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Austin O McCrea
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - David M Siwarski
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
37
|
Snapshot Profiling of the Antileishmanial Potency of Lead Compounds and Drug Candidates against Intracellular Leishmania donovani Amastigotes, with a Focus on Human-Derived Host Cells. Antimicrob Agents Chemother 2017; 61:AAC.01228-16. [PMID: 28069646 DOI: 10.1128/aac.01228-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 12/28/2016] [Indexed: 01/01/2023] Open
Abstract
This study characterized the in vitro potencies of antileishmanial agents against intracellular Leishmania donovani amastigotes in primary human macrophages, obtained with or without CD14-positive monocyte enrichment, phorbol 12-myristate 13-acetate (PMA)-differentiated THP-1 cells, and mouse peritoneal exudate macrophages (PEMs). Host cell-dependent potency was confirmed for pentavalent and trivalent antimony. Fexinidazole was inactive against intracellular amastigotes across the host cell panel. Fexinidazole sulfone, (R)-PA-824, (S)-PA-824, and VL-2098 displayed similar potency in all of the host cells tested.
Collapse
|
38
|
Hefnawy A, Berg M, Dujardin JC, De Muylder G. Exploiting Knowledge on Leishmania Drug Resistance to Support the Quest for New Drugs. Trends Parasitol 2016; 33:162-174. [PMID: 27993477 DOI: 10.1016/j.pt.2016.11.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/04/2016] [Accepted: 11/08/2016] [Indexed: 12/20/2022]
Abstract
New drugs are needed to control leishmaniasis and efforts are currently on-going to counter the neglect of this disease. We discuss here the utility and the impact of associating drug resistance (DR) studies to drug discovery pipelines. We use as paradigm currently used drugs, antimonials and miltefosine, and complement our reflection by interviewing three experts in the field. We suggest DR studies to be involved at two different stages of drug development: (i) the efficiency of novel compounds should be confirmed on sets of strains including recent clinical isolates with DR; (ii) experimental DR should be generated to promising compounds at an early stage of their development, to further optimize them and monitor clinical trials.
Collapse
Affiliation(s)
- Aya Hefnawy
- Institute of Tropical Medicine, Antwerp, Belgium
| | - Maya Berg
- Institute of Tropical Medicine, Antwerp, Belgium
| | | | | |
Collapse
|
39
|
Abstract
For decades antimonials were the drugs of choice for the treatment of visceral
leishmaniasis (VL), but the recent emergence of resistance has made them redundant as
first-line therapy in the endemic VL region in the Indian subcontinent. The application of
other drugs has been limited due to adverse effects, perceived high cost, need for
parenteral administration and increasing rate of treatment failures. Liposomal
amphotericin B (AmB) and miltefosine (MIL) have been positioned as the effective
first-line treatments; however, the number of monotherapy MIL-failures has increased after
a decade of use. Since no validated molecular resistance markers are yet available,
monitoring and surveillance of changes in drug sensitivity and resistance still depends on
standard phenotypic in vitro promastigote or amastigote susceptibility
assays. Clinical isolates displaying defined MIL- or AmB-resistance are still fairly
scarce and fundamental and applied research on resistance mechanisms and dynamics remains
largely dependent on laboratory-generated drug resistant strains. This review addresses
the various challenges associated with drug susceptibility and -resistance monitoring in
VL, with particular emphasis on the choice of strains, susceptibility model selection and
standardization of procedures with specific read-out parameters and well-defined threshold
criteria. The latter are essential to support surveillance systems and safeguard the
limited number of currently available antileishmanial drugs.
Collapse
|
40
|
PI3K signaling in Leishmania infections. Cell Immunol 2016; 309:19-22. [PMID: 27622385 DOI: 10.1016/j.cellimm.2016.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/01/2016] [Accepted: 09/04/2016] [Indexed: 11/22/2022]
Abstract
PI3K signaling plays a role in the host response to Leishmania infections. At the cellular level PI3K signaling is engaged by the parasite to control several cellular processes, which ensures parasite persistence. At the systemic level, there is evidence that recruitment of regulatory cells into lesions is impaired in the absence of robust PI3K signaling. In this mini-review the more recent studies that investigated the roles of PI3K signaling in Leishmania infections are discussed.
Collapse
|
41
|
de Moura TR, Santos MLB, Braz JM, Santos LFVC, Aragão MT, de Oliveira FA, Santos PL, da Silva ÂM, de Jesus AR, de Almeida RP. Cross-resistance of Leishmania infantum isolates to nitric oxide from patients refractory to antimony treatment, and greater tolerance to antileishmanial responses by macrophages. Parasitol Res 2016; 115:713-21. [PMID: 26481489 DOI: 10.1007/s00436-015-4793-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/12/2015] [Indexed: 11/24/2022]
Abstract
Visceral leishmaniasis is a life-threatening disease characterized by intense parasitism of the spleen, liver, and bone marrow. Antimonials have served as front-line antileishmanial therapeutics for decades, but the increasing failure rates under antimonial treatment have challenged the continued use of these drugs. Pentavalent antimonials are known to reinforce the killing mechanisms of macrophages, although the associated mechanism remains unclear. Here, for the first time, we determined whether Leishmania infantum strains isolated from patients refractory to antimony treatment (relapse cases) were cross-resistant to antimonials, liposomal amphotericin B, and/or nitric oxide, and also whether these strains modulate macrophage infection. We selected four clinical isolates from relapse cases and two clinical isolates from antimony-responsive patients (control group) for the present study. The L. infantum promastigotes from all four relapse cases were resistant to trivalent antimonial treatment and nitric oxide, while only one isolate was resistant to liposomal amphotericin B. We evaluated whether the resistant strains from relapse cases showed enhanced infectivity and amastigote survival in macrophages, or macrophage-killing mechanisms in macrophages activated by lipopolysaccharide plus interferon gamma. Infection indexes calculated using macrophages infected with isolates from relapse were higher than those observed with control strains that were stimulated independently. Macrophage infection was higher with L. infantum isolates from relapse cases and correlated with enhanced interleukin 1-β production but showed similar nitrite production. Our results demonstrate that L. infantum field isolates from relapse cases were resistant to antimonials and nitric oxide and that these parasites stimulated inflammatory cytokines and were resistant to macrophage-killing mechanisms, factors that may contribute to disease severity.
Collapse
|
42
|
A Telomeric Cluster of Antimony Resistance Genes on Chromosome 34 of Leishmania infantum. Antimicrob Agents Chemother 2016; 60:5262-75. [PMID: 27324767 DOI: 10.1128/aac.00544-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/11/2016] [Indexed: 11/20/2022] Open
Abstract
The mechanisms underlying the drug resistance of Leishmania spp. are manifold and not completely identified. Apart from the highly conserved multidrug resistance gene family known from higher eukaryotes, Leishmania spp. also possess genus-specific resistance marker genes. One of them, ARM58, was first identified in Leishmania braziliensis using a functional cloning approach, and its domain structure was characterized in L. infantum Here we report that L. infantum ARM58 is part of a gene cluster at the telomeric end of chromosome 34 also comprising the neighboring genes ARM56 and HSP23. We show that overexpression of all three genes can confer antimony resistance to intracellular amastigotes. Upon overexpression in L. donovani, ARM58 and ARM56 are secreted via exosomes, suggesting a scavenger/secretion mechanism of action. Using a combination of functional cloning and next-generation sequencing, we found that the gene cluster was selected only under antimonyl tartrate challenge and weakly under Cu(2+) challenge but not under sodium arsenite, Cd(2+), or miltefosine challenge. The selective advantage is less pronounced in intracellular amastigotes treated with the sodium stibogluconate, possibly due to the known macrophage-stimulatory activity of this drug, against which these resistance markers may not be active. Our data point to the specificity of these three genes for antimony resistance.
Collapse
|
43
|
Caljon G, De Muylder G, Durnez L, Jennes W, Vanaerschot M, Dujardin JC. Alice in microbes' land: adaptations and counter-adaptations of vector-borne parasitic protozoa and their hosts. FEMS Microbiol Rev 2016; 40:664-85. [PMID: 27400870 DOI: 10.1093/femsre/fuw018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2016] [Indexed: 12/24/2022] Open
Abstract
In the present review, we aim to provide a general introduction to different facets of the arms race between pathogens and their hosts/environment, emphasizing its evolutionary aspects. We focus on vector-borne parasitic protozoa, which have to adapt to both invertebrate and vertebrate hosts. Using Leishmania, Trypanosoma and Plasmodium as main models, we review successively (i) the adaptations and counter-adaptations of parasites and their invertebrate host, (ii) the adaptations and counter-adaptations of parasites and their vertebrate host and (iii) the impact of human interventions (chemotherapy, vaccination, vector control and environmental changes) on these adaptations. We conclude by discussing the practical impact this knowledge can have on translational research and public health.
Collapse
Affiliation(s)
- Guy Caljon
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Health, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Géraldine De Muylder
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium
| | - Lies Durnez
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium
| | - Wim Jennes
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium
| | - Manu Vanaerschot
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium Columbia University, College of Physicians and Surgeons, Department of Microbiology and Immunology, Fidock Lab, New York, NY 10032, USA
| | - Jean-Claude Dujardin
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Health, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| |
Collapse
|
44
|
Kauffmann F, Dumetz F, Hendrickx S, Muraille E, Dujardin JC, Maes L, Magez S, De Trez C. In vivocharacterization of two additionalLeishmania donovanistrains using the murine and hamster model. Parasite Immunol 2016; 38:290-302. [DOI: 10.1111/pim.12316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/19/2016] [Indexed: 11/29/2022]
Affiliation(s)
- F. Kauffmann
- Laboratory of Cellular and Molecular Immunology (CMIM); Vrije Universiteit Brussel (VUB); Brussels Belgium
- Department of Structural Biology Research Center; Vlaams Instituut voor Biotechnologie (VIB); Brussels Belgium
| | - F. Dumetz
- Molecular Parasitology Unit; Department of Biomedical Sciences; Institute of Tropical Medicine; Antwerpen Belgium
| | - S. Hendrickx
- Laboratory of Microbiology; Parasitology and Hygiene (LMPH); University of Antwerp; Antwerpen Belgium
| | - E. Muraille
- Laboratory of Parasitology; Université Libre de Bruxelles; Brussels Belgium
| | - J.-C. Dujardin
- Molecular Parasitology Unit; Department of Biomedical Sciences; Institute of Tropical Medicine; Antwerpen Belgium
| | - L. Maes
- Laboratory of Parasitology; Université Libre de Bruxelles; Brussels Belgium
| | - S. Magez
- Laboratory of Cellular and Molecular Immunology (CMIM); Vrije Universiteit Brussel (VUB); Brussels Belgium
- Department of Structural Biology Research Center; Vlaams Instituut voor Biotechnologie (VIB); Brussels Belgium
| | - C. De Trez
- Laboratory of Cellular and Molecular Immunology (CMIM); Vrije Universiteit Brussel (VUB); Brussels Belgium
- Department of Structural Biology Research Center; Vlaams Instituut voor Biotechnologie (VIB); Brussels Belgium
| |
Collapse
|
45
|
Hendrickx S, Beyers J, Mondelaers A, Eberhardt E, Lachaud L, Delputte P, Cos P, Maes L. Evidence of a drug-specific impact of experimentally selected paromomycin and miltefosine resistance on parasite fitness in Leishmania infantum. J Antimicrob Chemother 2016; 71:1914-21. [PMID: 27084919 DOI: 10.1093/jac/dkw096] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 02/29/2016] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Although miltefosine and paromomycin were only recently introduced to treat visceral leishmaniasis, increasing numbers of miltefosine treatment failures and occasional primary resistance to both drugs have been reported. Understanding alterations in parasite behaviour linked to drug resistance is essential to assess the propensity for emergence and spread of resistant strains, particularly since a positive effect on fitness has been reported for antimony-resistant parasites. This laboratory study compared the fitness of a drug-susceptible parent WT clinical Leishmania infantum isolate (MHOM/FR/96/LEM3323) and derived miltefosine and paromomycin drug-resistant lines that were experimentally selected at the intracellular amastigote level. METHODS Parasite fitness of WT, paromomycin-resistant and miltefosine-resistant strains, in vitro and in vivo parasite growth, metacyclogenesis, infectivity and macrophage stress responses were comparatively evaluated. RESULTS No significant differences in promastigote fitness were noted between the WT and paromomycin-resistant strain, while clear benefits could be demonstrated for paromomycin-resistant amastigotes in terms of enhanced in vitro and in vivo growth potential and intracellular stress response. The miltefosine-resistant phenotype showed incomplete promastigote metacyclogenesis, decreased intracellular growth and weakened stress response, revealing a reduced fitness compared with WT parent parasites. CONCLUSIONS The rapid selection and fitness advantages of paromomycin-resistant amastigotes endorse the current use of paromomycin in combination therapy. Although a reduced fitness of miltefosine-resistant strains may explain the difficulty of miltefosine resistance selection in vitro, the growing number of miltefosine treatment failures in the field still requires further exploratory research.
Collapse
Affiliation(s)
- S Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - J Beyers
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - A Mondelaers
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - E Eberhardt
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - L Lachaud
- Laboratoire de Parasitologie-Mycologie et Centre National de Référence des Leishmanioses, Centre Hospitalier Universitaire et Université de Montpellier, Montpellier, France
| | - P Delputte
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - P Cos
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - L Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
46
|
Shaw CD, Lonchamp J, Downing T, Imamura H, Freeman TM, Cotton JA, Sanders M, Blackburn G, Dujardin JC, Rijal S, Khanal B, Illingworth CJR, Coombs GH, Carter KC. In vitro selection of miltefosine resistance in promastigotes of Leishmania donovani from Nepal: genomic and metabolomic characterization. Mol Microbiol 2016; 99:1134-48. [PMID: 26713880 PMCID: PMC4832254 DOI: 10.1111/mmi.13291] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2015] [Indexed: 12/17/2022]
Abstract
In this study, we followed the genomic, lipidomic and metabolomic changes associated with the selection of miltefosine (MIL) resistance in two clinically derived Leishmania donovani strains with different inherent resistance to antimonial drugs (antimony sensitive strain Sb-S; and antimony resistant Sb-R). MIL-R was easily induced in both strains using the promastigote-stage, but a significant increase in MIL-R in the intracellular amastigote compared to the corresponding wild-type did not occur until promastigotes had adapted to 12.2 μM MIL. A variety of common and strain-specific genetic changes were discovered in MIL-adapted parasites, including deletions at the LdMT transporter gene, single-base mutations and changes in somy. The most obvious lipid changes in MIL-R promastigotes occurred to phosphatidylcholines and lysophosphatidylcholines and results indicate that the Kennedy pathway is involved in MIL resistance. The inherent Sb resistance of the parasite had an impact on the changes that occurred in MIL-R parasites, with more genetic changes occurring in Sb-R compared with Sb-S parasites. Initial interpretation of the changes identified in this study does not support synergies with Sb-R in the mechanisms of MIL resistance, though this requires an enhanced understanding of the parasite's biochemical pathways and how they are genetically regulated to be verified fully.
Collapse
Affiliation(s)
- C D Shaw
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - J Lonchamp
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - T Downing
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- College of Science, NUI Galway, Galway, Ireland
| | - H Imamura
- Department of Biomedical Sciences, Instituut voor Tropische Geneeskunde Nationalestraat, Antwerpen, Belgium
| | - T M Freeman
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - J A Cotton
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
| | - M Sanders
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
| | - G Blackburn
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Antwerpen, Belgium
- Glasgow Polyomics, University of Glasgow, Glasgow
| | - J C Dujardin
- Department of Biomedical Sciences, Instituut voor Tropische Geneeskunde Nationalestraat, Antwerpen, Belgium
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Antwerpen, Belgium
| | - S Rijal
- BP Koirala Institute of Health Sciences, Dharan, Nepal
| | - B Khanal
- BP Koirala Institute of Health Sciences, Dharan, Nepal
| | | | - G H Coombs
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - K C Carter
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| |
Collapse
|
47
|
Hendrickx S, Leemans A, Mondelaers A, Rijal S, Khanal B, Dujardin JC, Delputte P, Cos P, Maes L. Comparative Fitness of a Parent Leishmania donovani Clinical Isolate and Its Experimentally Derived Paromomycin-Resistant Strain. PLoS One 2015; 10:e0140139. [PMID: 26469696 PMCID: PMC4607421 DOI: 10.1371/journal.pone.0140139] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/22/2015] [Indexed: 11/19/2022] Open
Abstract
Paromomycin has recently been introduced for the treatment of visceral leishmaniasis and emergence of drug resistance can only be appropriately judged upon its long term routine use in the field. Understanding alterations in parasite behavior linked to paromomycin-resistance may be essential to assess the propensity for emergence and spread of resistant strains. A standardized and integrated laboratory approach was adopted to define and assess parasite fitness of both promastigotes and amastigotes using an experimentally induced paromomycin-resistant Leishmania donovani strain and its paromomycin-susceptible parent wild-type clinical isolate. Primary focus was placed on parasite growth and virulence, two major components of parasite fitness. The combination of in vitro and in vivo approaches enabled detailed comparison of wild-type and resistant strains for which no differences could be demonstrated with regard to promastigote growth, metacyclogenesis, in vitro infectivity, multiplication in primary peritoneal mouse macrophages and infectivity for Balb/c mice upon infection with 2 x 107 metacyclic promastigotes. Monitoring of in vitro intracellular amastigote multiplication revealed a consistent decrease in parasite burden over time for both wild-type and resistant parasites, an observation that was subsequently also confirmed in a larger set of L. donovani clinical isolates. Though the impact of these findings should be further explored, the study results suggest that the epidemiological implications of acquired paromomycin-resistance may remain minimal other than the loss of one of the last remaining drugs effective against visceral leishmaniasis.
Collapse
Affiliation(s)
- Sarah Hendrickx
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Annelies Leemans
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Annelies Mondelaers
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Suman Rijal
- BP Koirala Institute of Health Sciences, Dharan, Nepal
| | | | - Jean-Claude Dujardin
- Molecular Parasitology Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter Delputte
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
- * E-mail:
| |
Collapse
|
48
|
Das S, Shah P, Tandon R, Yadav NK, Sahasrabuddhe AA, Sundar S, Siddiqi MI, Dube A. Over-Expression of Cysteine Leucine Rich Protein Is Related to SAG Resistance in Clinical Isolates of Leishmania donovani. PLoS Negl Trop Dis 2015; 9:e0003992. [PMID: 26295340 PMCID: PMC4546639 DOI: 10.1371/journal.pntd.0003992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/16/2015] [Indexed: 01/26/2023] Open
Abstract
Background Resistance emergence against antileishmanial drugs, particularly Sodium Antimony Gluconate (SAG) has severely hampered the therapeutic strategy against visceral leishmaniasis, the mechanism of resistance being indistinguishable. Cysteine leucine rich protein (CLrP), was recognized as one of the overexpressed proteins in resistant isolates, as observed in differential proteomics between sensitive and resistant isolates of L. donovani. The present study deals with the characterization of CLrP and for its possible connection with SAG resistance. Methodology and Principal Findings In pursuance of deciphering the role of CLrP in SAG resistance, gene was cloned, over-expressed in E. coli system and thereafter antibody was raised. The expression profile of CLrP and was found to be over-expressed in SAG resistant clinical isolates of L. donovani as compared to SAG sensitive ones when investigated by real-time PCR and western blotting. CLrP has been characterized through bioinformatics, immunoblotting and immunolocalization analysis, which reveals its post-translational modification along with its dual existence in the nucleus as well as in the membrane of the parasite. Further investigation using a ChIP assay confirmed its DNA binding potential. Over-expression of CLrP in sensitive isolate of L. donovani significantly decreased its responsiveness to SAG (SbV and SbIII) and a shift towards the resistant mode was observed. Further, a significant increase in its infectivity in murine macrophages has been observed. Conclusion/Significance The study reports the differential expression of CLrP in SAG sensitive and resistant isolates of L. donovani. Functional intricacy of CLrP increases with dual localization, glycosylation and DNA binding potential of the protein. Further over-expressing CLrP in sensitive isolate of L. donovani shows significantly decreased sensitivity towards SAG and increased infectivity as well, thus assisting the parasite in securing a safe niche. Results indicates the possible contribution of CLrP to antimonial resistance in L. donovani by assisting the parasite growth in the macrophages. Leishmania causes complex of pathologies called Leishmaniasis and among the several forms visceral leishmaniasis is the precarious one as being fatal, if left untreated. Emergence of resistance against several antileishmanials particularly Sodium Antimony Gluconate (SAG) has severely battered the therapeutic strategy against VL and the resistance mechanism is still vague. Thus, to apprehend the underlying mechanism, previously, a differential proteomics of SAG unresponsive versus SAG sensitive isolates of L.donovani was done wherein overexpression of Cysteine Leucine Rich protein (CLrP), a member of Leucine rich repeat superfamily, was observed. To scrutinize its involvement in the SAG resistance mechanism, which is till date not investigated, the characterization of CLrP was carried out which revealed its post-translational modification along with its dual existence in the nucleus and in the membrane of the parasite. Further investigation using a ChIP assay confirmed its DNA binding potential. Over-expression of CLrP in sensitive isolate of L. donovani significantly decreased its SAG sensitivity. CLrP overexpressed parasites have increased infectivity. These results point out towards the CLrP’s contribution to antimonial resistance in L. donovani by facilitating parasites growth through macrophages. Further studies are required to depict CLrP as a potential drug target to strengthen the present arsenal against L donovani.
Collapse
Affiliation(s)
- Sanchita Das
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Priyanka Shah
- Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rati Tandon
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, India
| | | | | | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | | - Anuradha Dube
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, India
- * E-mail: ,
| |
Collapse
|
49
|
Mukherjee B, Paul J, Mukherjee S, Mukhopadhyay R, Das S, Naskar K, Sundar S, Dujardin JC, Saha B, Roy S. Antimony-Resistant Leishmania donovani Exploits miR-466i To Deactivate Host MyD88 for Regulating IL-10/IL-12 Levels during Early Hours of Infection. THE JOURNAL OF IMMUNOLOGY 2015; 195:2731-42. [PMID: 26283478 DOI: 10.4049/jimmunol.1402585] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 07/15/2015] [Indexed: 01/29/2023]
Abstract
Infection with antimony-resistant Leishmania donovani (Sb(R)LD) induces aggressive pathology in the mammalian hosts as compared with ones with antimony-sensitive L. donovani (Sb(S)LD) infection. Sb(R)LD, but not Sb(S)LD, interacts with TLR2/TLR6 to induce IL-10 by exploiting p50/c-Rel subunits of NF-κB in infected macrophages (Mϕs). Most of the TLRs exploit the universal adaptor protein MyD88 to activate NF-κB. We now show that infection of Mϕs from MyD88(-/-) mice with Sb(R)LD gave rise to significantly higher intracellular parasite number coupled with elevated IL-10/IL-12 ratio in the culture supernatant as compared with infection in wild type (WT) Mϕs. Τhese attributes were not seen with Sb(S)LD in similar experiments. Further, Sb(R)LD infection upregulated miR-466i, which binds with 3'-untranslated region, leading to the downregulation of MyD88. Infection of MyD88(-/-) Mϕ or IL-12(-/-) Mϕ with Sb(R)LD induced IL-10 surge at 4 h, whereas the same in WT Mϕ started from 12 h. Thus, absence of IL-12 in MyD88(-/-) mice favored early binding of NF-κB subunits to the IL-10 promoter, resulting in IL-10 surge. Infection of MyD88(-/-) mice with Sb(R)LD showed significantly higher organ parasites coupled with ill-defined and immature hepatic granulomas, whereas in WT mice there were less organ parasites and the granulomas were well defined. From the survival kinetics it was observed that Sb(R)LD-infected MyD88(-/-) mice died by 60 d postinfection, whereas the WT mice continued to survive. Our results demonstrate that Sb(R)LD has evolved a unique strategy to evade host antileishmanial immune repertoire by manipulating host MyD88 to its advantage.
Collapse
Affiliation(s)
- Budhaditya Mukherjee
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Joydeep Paul
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sandip Mukherjee
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Rupkatha Mukhopadhyay
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Shantanabha Das
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Kshudiram Naskar
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Shyam Sundar
- Institute of Medical Sciences, Benaras Hindu University, Varanasi 221005, India
| | - Jean-Claude Dujardin
- Institute of Tropical Medicine, Antwerp 2000, Belgium; University of Antwerp, Antwerp 2000, Belgium; and
| | - Bhaskar Saha
- National Centre for Cell Science, Pune 411007, India
| | - Syamal Roy
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India;
| |
Collapse
|
50
|
A Multiplatform Metabolomic Approach to the Basis of Antimonial Action and Resistance in Leishmania infantum. PLoS One 2015; 10:e0130675. [PMID: 26161866 PMCID: PMC4498920 DOI: 10.1371/journal.pone.0130675] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/25/2015] [Indexed: 12/20/2022] Open
Abstract
There is a rising resistance against antimony drugs, the gold-standard for treatment until some years ago. That is a serious problem due to the paucity of drugs in current clinical use. In a research to reveal how these drugs affect the parasite during treatment and to unravel the underlying basis for their resistance, we have employed metabolomics to study treatment in Leishmania infantum promastigotes. This was accomplished first through the untargeted analysis of metabolic snapshots of treated and untreated parasites both resistant and responders, utilizing a multiplatform approach to give the widest as possible coverage of the metabolome, and additionally through novel monitoring of the origin of the detected alterations through a 13C traceability experiment. Our data stress a multi-target metabolic alteration with treatment, affecting in particular the cell redox system that is essential to cope with detoxification and biosynthetic processes. Additionally, relevant changes were noted in amino acid metabolism. Our results are in agreement with other authors studying other Leishmania species.
Collapse
|