1
|
Azevedo LG, Sosa E, de Queiroz ATL, Barral A, Wheeler RJ, Nicolás MF, Farias LP, Do Porto DF, Ramos PIP. High-throughput prioritization of target proteins for development of new antileishmanial compounds. Int J Parasitol Drugs Drug Resist 2024; 25:100538. [PMID: 38669848 PMCID: PMC11068527 DOI: 10.1016/j.ijpddr.2024.100538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/11/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Leishmaniasis, a vector-borne disease, is caused by the infection of Leishmania spp., obligate intracellular protozoan parasites. Presently, human vaccines are unavailable, and the primary treatment relies heavily on systemic drugs, often presenting with suboptimal formulations and substantial toxicity, making new drugs a high priority for LMIC countries burdened by the disease, but a low priority in the agenda of most pharmaceutical companies due to unattractive profit margins. New ways to accelerate the discovery of new, or the repositioning of existing drugs, are needed. To address this challenge, our study aimed to identify potential protein targets shared among clinically-relevant Leishmania species. We employed a subtractive proteomics and comparative genomics approach, integrating high-throughput multi-omics data to classify these targets based on different druggability metrics. This effort resulted in the ranking of 6502 ortholog groups of protein targets across 14 pathogenic Leishmania species. Among the top 20 highly ranked groups, metabolic processes known to be attractive drug targets, including the ubiquitination pathway, aminoacyl-tRNA synthetases, and purine synthesis, were rediscovered. Additionally, we unveiled novel promising targets such as the nicotinate phosphoribosyltransferase enzyme and dihydrolipoamide succinyltransferases. These groups exhibited appealing druggability features, including less than 40% sequence identity to the human host proteome, predicted essentiality, structural classification as highly druggable or druggable, and expression levels above the 50th percentile in the amastigote form. The resources presented in this work also represent a comprehensive collection of integrated data regarding trypanosomatid biology.
Collapse
Affiliation(s)
- Lucas G Azevedo
- Center for Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil; Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil.
| | - Ezequiel Sosa
- Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Artur T L de Queiroz
- Center for Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil; Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil.
| | - Aldina Barral
- Laboratório de Medicina e Saúde Pública de Precisão (MeSP2), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil.
| | - Richard J Wheeler
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| | - Marisa F Nicolás
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil.
| | - Leonardo P Farias
- Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil; Laboratório de Medicina e Saúde Pública de Precisão (MeSP2), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil.
| | | | - Pablo Ivan P Ramos
- Center for Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil; Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil.
| |
Collapse
|
2
|
Scaramele NF, Troiano JA, Felix JDS, Costa SF, Almeida MC, Florencio de Athayde FR, Soares MF, Lopes MFDS, Furlan ADO, de Lima VMF, Lopes FL. Leishmania infantum infection modulates messenger RNA, microRNA and long non-coding RNA expression in human neutrophils in vitro. PLoS Negl Trop Dis 2024; 18:e0012318. [PMID: 39028711 PMCID: PMC11259272 DOI: 10.1371/journal.pntd.0012318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/25/2024] [Indexed: 07/21/2024] Open
Abstract
In the Americas, L. infantum (syn. chagasi) is the main cause of human visceral leishmaniasis. The role of neutrophils as part of the innate response to Leishmania spp. infection is dubious and varies according to the species causing the infection. Global expression of coding RNAs, microRNAs and long non-coding RNAs changes as part of the immune response against pathogens. Changes in mRNA and non-coding RNA expression resulting from infection by Leishmania spp. are widely studied in macrophages, but scarce in neutrophils, the first cell to encounter the trypanosomatid, especially following infection by L. infantum. Herein, we aimed to understand the expression patterns of coding and non-coding transcripts during acute in vitro infection of human neutrophils by L. infantum. We isolated neutrophils from whole blood of healthy male donors (n = 5) and split into groups: 1) infected with L. infantum (MOI = 5:1), and 2) uninfected controls. After 3 hours of exposure of infected group to promastigotes of L. infantum, followed by 17 hours of incubation, total RNA was extracted and total RNA-Seq and miRNA microarray were performed. A total of 212 genes were differentially expressed in neutrophils following RNA-Seq analysis (log2(FC)±0.58, FDR≤0.05). In vitro infection with L. infantum upregulated the expression of 197 and reduced the expression of 92 miRNAs in human neutrophils (FC±2, FDR≤0.01). Lastly, 5 downregulated genes were classified as lncRNA, and of the 10 upregulated genes, there was only 1 lncRNA. Further bioinformatic analysis indicated that changes in the transcriptome and microtranscriptome of neutrophils, following in vitro infection with L. infantum, may impair phagocytosis, apoptosis and decrease nitric oxide production. Our work sheds light on several mechanisms used by L. infantum to control neutrophil-mediated immune response and identifies several targets for future functional studies, aiming at the development of preventive or curative treatments for this prevalent zoonosis.
Collapse
Affiliation(s)
- Natália Francisco Scaramele
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Jéssica Antonini Troiano
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Juliana de Souza Felix
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Sidnei Ferro Costa
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Mariana Cordeiro Almeida
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Flávia Regina Florencio de Athayde
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Matheus Fujimura Soares
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Maria Fernanda da Silva Lopes
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Amanda de Oliveira Furlan
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Valéria Marçal Felix de Lima
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Flavia Lombardi Lopes
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| |
Collapse
|
3
|
de Vrij N, Pollmann J, Rezende AM, Ibarra-Meneses AV, Pham TT, Hailemichael W, Kassa M, Bogale T, Melkamu R, Yeshanew A, Mohammed R, Diro E, Maes I, Domagalska MA, Landuyt H, Vogt F, van Henten S, Laukens K, Cuypers B, Meysman P, Beyene H, Sisay K, Kibret A, Mersha D, Ritmeijer K, van Griensven J, Adriaensen W. Persistent T cell unresponsiveness associated with chronic visceral leishmaniasis in HIV-coinfected patients. Commun Biol 2024; 7:524. [PMID: 38702419 PMCID: PMC11068874 DOI: 10.1038/s42003-024-06225-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
A large proportion of HIV-coinfected visceral leishmaniasis (VL-HIV) patients exhibit chronic disease with frequent VL recurrence. However, knowledge on immunological determinants underlying the disease course is scarce. We longitudinally profiled the circulatory cellular immunity of an Ethiopian HIV cohort that included VL developers. We show that chronic VL-HIV patients exhibit high and persistent levels of TIGIT and PD-1 on CD8+/CD8- T cells, in addition to a lower frequency of IFN-γ+ TIGIT- CD8+/CD8- T cells, suggestive of impaired T cell functionality. At single T cell transcriptome and clonal resolution, the patients show CD4+ T cell anergy, characterised by a lack of T cell activation and lymphoproliferative response. These findings suggest that PD-1 and TIGIT play a pivotal role in VL-HIV chronicity, and may be further explored for patient risk stratification. Our findings provide a strong rationale for adjunctive immunotherapy for the treatment of chronic VL-HIV patients to break the recurrent disease cycle.
Collapse
Affiliation(s)
- Nicky de Vrij
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | - Julia Pollmann
- Department of Medical Oncology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT) Heidelberg, 69120, Heidelberg, Germany
| | - Antonio M Rezende
- Department of Microbiology, Aggeu Magalhães Institute-FIOCRUZ/PE, Recife, Brazil
| | - Ana V Ibarra-Meneses
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Thao-Thy Pham
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Wasihun Hailemichael
- Department of Immunology and Molecular Biology, Faculty of Biomedical Sciences, University of Gondar, Gondar, Ethiopia
| | - Mekibib Kassa
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Tadfe Bogale
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Roma Melkamu
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Arega Yeshanew
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Rezika Mohammed
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Ermias Diro
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Ilse Maes
- Molecular Parasitology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Malgorzata A Domagalska
- Molecular Parasitology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Hanne Landuyt
- Clinical Trial Unit, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Florian Vogt
- National Centre for Epidemiology and Population Health, The Australian National University, Canberra, 2601, Australia
- The Kirby Institute, University of New South Wales, Sydney, 2052, Australia
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Saskia van Henten
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Kris Laukens
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | - Bart Cuypers
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | - Pieter Meysman
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | | | | | | | | | | | - Johan van Griensven
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Wim Adriaensen
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium.
| |
Collapse
|
4
|
Diotallevi A, Bruno F, Castelli G, Persico G, Buffi G, Ceccarelli M, Ligi D, Mannello F, Vitale F, Magnani M, Galluzzi L. Transcriptional signatures in human macrophage-like cells infected by Leishmania infantum, Leishmania major and Leishmania tropica. PLoS Negl Trop Dis 2024; 18:e0012085. [PMID: 38578804 PMCID: PMC11023634 DOI: 10.1371/journal.pntd.0012085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 04/17/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND In the Mediterranean basin, three Leishmania species have been identified: L. infantum, L. major and L. tropica, causing zoonotic visceral leishmaniasis (VL), zoonotic cutaneous leishmaniasis (CL) and anthroponotic CL, respectively. Despite animal models and genomic/transcriptomic studies provided important insights, the pathogenic determinants modulating the development of VL and CL are still poorly understood. This work aimed to identify host transcriptional signatures shared by cells infected with L. infantum, L. major, and L. tropica, as well as specific transcriptional signatures elicited by parasites causing VL (i.e., L. infantum) and parasites involved in CL (i.e., L. major, L. tropica). METHODOLOGY/PRINCIPAL FINDINGS U937 cells differentiated into macrophage-like cells were infected with L. infantum, L. major and L. tropica for 24h and 48h, and total RNA was extracted. RNA sequencing, performed on an Illumina NovaSeq 6000 platform, was used to evaluate the transcriptional signatures of infected cells with respect to non-infected cells at both time points. The EdgeR package was used to identify differentially expressed genes (fold change > 2 and FDR-adjusted p-values < 0.05). Then, functional enrichment analysis was employed to identify the enriched ontology terms in which these genes are involved. At 24h post-infection, a common signature of 463 dysregulated genes shared among all infection conditions was recognized, while at 48h post-infection the common signature was reduced to 120 genes. Aside from a common transcriptional response, we evidenced different upregulated functional pathways characterizing L. infantum-infected cells, such as VEGFA-VEGFR2 and NFE2L2-related pathways, indicating vascular remodeling and reduction of oxidative stress as potentially important factors for visceralization. CONCLUSIONS The identification of pathways elicited by parasites causing VL or CL could lead to new therapeutic strategies for leishmaniasis, combining the canonical anti-leishmania compounds with host-directed therapy.
Collapse
Affiliation(s)
- Aurora Diotallevi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Federica Bruno
- Centro di Referenza Nazionale per le Leishmaniosi (C.Re.Na.L.), OIE Leishmania Reference Laboratory, Istituto Zooprofilattico Sperimentale della Sicilia A Mirri, Palermo, Italy
| | - Germano Castelli
- Centro di Referenza Nazionale per le Leishmaniosi (C.Re.Na.L.), OIE Leishmania Reference Laboratory, Istituto Zooprofilattico Sperimentale della Sicilia A Mirri, Palermo, Italy
| | - Giuseppe Persico
- Department of Experimental Oncology, IRCCS, European Institute of Oncology, Milan, Italy
| | - Gloria Buffi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Marcello Ceccarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Daniela Ligi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ferdinando Mannello
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Fabrizio Vitale
- Centro di Referenza Nazionale per le Leishmaniosi (C.Re.Na.L.), OIE Leishmania Reference Laboratory, Istituto Zooprofilattico Sperimentale della Sicilia A Mirri, Palermo, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Luca Galluzzi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
5
|
Shen Z, Ke Z, Yang Q, Ghebremichael ST, Li T, Li T, Chen J, Meng X, Xiang H, Li C, Zhou Z, Pan G, Chen P. Transcriptomic changes in the microsporidia proliferation and host responses in congenitally infected embryos and larvae. BMC Genomics 2024; 25:321. [PMID: 38556880 PMCID: PMC10983672 DOI: 10.1186/s12864-024-10236-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
Congenital infection caused by vertical transmission of microsporidia N. bombycis can result in severe economic losses in the silkworm-rearing industry. Whole-transcriptome analyses have revealed non-coding RNAs and their regulatory networks in N. bombycis infected embryos and larvae. However, transcriptomic changes in the microsporidia proliferation and host responses in congenitally infected embryos and larvae remains unclear. Here, we simultaneously compared the transcriptomes of N. bombycis and its host B. mori embryos of 5-day and larvae of 1-, 5- and 10-day during congenital infection. For the transcriptome of N. bombycis, a comparison of parasite expression patterns between congenital-infected embryos and larva showed most genes related to parasite central carbon metabolism were down-regulated in larvae during infection, whereas the majority of genes involved in parasite proliferation and growth were up-regulated. Interestingly, a large number of distinct or shared differentially expressed genes (DEGs) were revealed by the Venn diagram and heat map, many of them were connected to infection related factors such as Ricin B lectin, spore wall protein, polar tube protein, and polysaccharide deacetylase. For the transcriptome of B. mori infected with N. bombycis, beyond numerous DEGs related to DNA replication and repair, mRNA surveillance pathway, RNA transport, protein biosynthesis, and proteolysis, with the progression of infection, a large number of DEGs related to immune and infection pathways, including phagocytosis, apoptosis, TNF, Toll-like receptor, NF-kappa B, Fc epsilon RI, and some diseases, were successively identified. In contrast, most genes associated with the insulin signaling pathway, 2-oxacarboxylic acid metabolism, amino acid biosynthesis, and lipid metabolisms were up-regulated in larvae compared to those in embryos. Furthermore, dozens of distinct and three shared DEGs that were involved in the epigenetic regulations, such as polycomb, histone-lysine-specific demethylases, and histone-lysine-N-methyltransferases, were identified via the Venn diagram and heat maps. Notably, many DEGs of host and parasite associated with lipid-related metabolisms were verified by RT-qPCR. Taken together, simultaneous transcriptomic analyses of both host and parasite genes lead to a better understanding of changes in the microsporidia proliferation and host responses in embryos and larvae in N. bombycis congenital infection.
Collapse
Affiliation(s)
- Zigang Shen
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Tiansheng Street, Chongqing, 400716, China
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Zhuojun Ke
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Qiong Yang
- Sericulture and Agri-food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Samson Teweldeberhan Ghebremichael
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Tangxin Li
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Tian Li
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Jie Chen
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Xianzhi Meng
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Heng Xiang
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Chunfeng Li
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Zeyang Zhou
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
- College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Guoqing Pan
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China.
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China.
| | - Ping Chen
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Tiansheng Street, Chongqing, 400716, China.
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China.
| |
Collapse
|
6
|
Palacios G, Diaz-Solano R, Valladares B, Dorta-Guerra R, Carmelo E. Evolving immunometabolic response to the early Leishmania infantum infection in the spleen of BALB/c mice described by gene expression profiling. Acta Trop 2023; 247:107005. [PMID: 37619900 DOI: 10.1016/j.actatropica.2023.107005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/27/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Transcriptional analysis is a useful approximation towards the identification of global changes in host-pathogen interaction, in order to elucidate tissue-specific immune responses that drive the immunopathology of the disease. For this purpose, expression of 223 genes involved in innate and adaptive immune response, lipid metabolism, prostaglandin synthesis, C-type lectin receptors and MAPK signaling pathway, among other processes, were analyzed during the early infection in spleens of BALB/c mice infected by Leishmania infantum. Our results highlight the activation of immune responses in spleen tissue as early as 1 day p.i., but a mixed pro-inflammatory and regulatory response at day 10 p.i., failing to induce an effective response towards control of Leishmania infection in the spleen. This ineffective response is coupled to downregulation of metabolic markers relevant for pathways related to icosanoid biosynthesis, adipocytokine signaling or HIF-1 signaling, among others. Interestingly, the over-representation of processes related to immune response, revealed Il21 as a potential early biomarker of L. infantum infection in the spleen. These results provide insights into the relationships between immune and metabolic responses at transcriptional level during the first days of infection in the L. infantum-BALB/c experimental model, revealing the deregulation of many important pathways and processes crucial for parasitic control in infected tissues.
Collapse
Affiliation(s)
- Génesis Palacios
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n (Tenerife), La Laguna 38200, Spain
| | - Raquel Diaz-Solano
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n (Tenerife), La Laguna 38200, Spain
| | - Basilio Valladares
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n (Tenerife), La Laguna 38200, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, (Tenerife), La Laguna 38200, Spain
| | - Roberto Dorta-Guerra
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n (Tenerife), La Laguna 38200, Spain; Departamento de Matemáticas, Estadística e Investigación Operativa, Facultad de Ciencias, Universidad de La Laguna, (Tenerife), La Laguna 38200, Spain
| | - Emma Carmelo
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n (Tenerife), La Laguna 38200, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, (Tenerife), La Laguna 38200, Spain.
| |
Collapse
|
7
|
Vargas DA, Gregory DJ, Koren RN, Zilberstein D, Belew AT, El-Sayed NM, Gómez MA. Macrophage metallothioneins participate in the antileishmanial activity of antimonials. FRONTIERS IN PARASITOLOGY 2023; 2:1242727. [PMID: 38239429 PMCID: PMC10795579 DOI: 10.3389/fpara.2023.1242727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
Abstract
Host cell functions that participate in the pharmacokinetics and pharmacodynamics (PK/PD) of drugs against intracellular pathogen infections are critical for drug efficacy. In this study, we investigated whether macrophage mechanisms of xenobiotic detoxification contribute to the elimination of intracellular Leishmania upon exposure to pentavalent antimonials (SbV). Primary macrophages from patients with cutaneous leishmaniasis (CL) (n=6) were exposed ex vivo to L. V. panamensis infection and SbV, and transcriptomes were generated. Seven metallothionein (MT) genes, potent scavengers of heavy metals and central elements of the mammalian cell machinery for xenobiotic detoxification, were within the top 20 up-regulated genes. To functionally validate the participation of MTs in drug-mediated killing of intracellular Leishmania, tandem knockdown (KD) of MT2-A and MT1-E, MT1-F, and MT1-X was performed using a pan-MT shRNA approach in THP-1 cells. Parasite survival was unaffected in tandem-KD cells, as a consequence of strong transcriptional upregulation of MTs by infection and SbV, overcoming the KD effect. Gene silencing of the metal transcription factor-1 (MTF-1) abrogated expression of MT1 and MT2-A genes, but not ZnT-1. Upon exposure to SbV, intracellular survival of Leishmania in MTF-1KD cells was significantly enhanced. Results from this study highlight the participation of macrophage MTs in Sb-dependent parasite killing.
Collapse
Affiliation(s)
- Deninson Alejandro Vargas
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - David J. Gregory
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Roni Nitzan Koren
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Dan Zilberstein
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ashton Trey Belew
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| | - Najib M. El-Sayed
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| | - María Adelaida Gómez
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| |
Collapse
|
8
|
Venugopal G, Bird JT, Roys H, Bowlin A, Fry L, Byrum SD, Weinkopff T. BOTH THE INFECTION STATUS AND INFLAMMATORY MICROENVIRONMENT INDUCE TRANSCRIPTIONAL REMODELING IN MACROPHAGES IN MURINE LEISHMANIAL LESIONS. J Parasitol 2023; 109:200-210. [PMID: 37270767 DOI: 10.1645/22-94] [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: 06/05/2023] Open
Abstract
Cutaneous leishmaniasis is caused by infection with the protozoan parasite Leishmania, which resides intracellularly in dermal macrophages (Mø), producing lesions. The skin lesions are characterized by proinflammatory cytokines and growth factors as well as inflammatory hypoxia, creating a stressful microenvironment for Mø. Of importance, not all Mø in lesions harbor parasites. To distinguish the influence of the parasite from the inflammatory microenvironment after Leishmania major (LM) infection on the Mø, we performed single-cell RNA sequencing and compared Mø associated with LM transcripts (or 'infected' Mø) with Mø not associated with LM transcripts (or 'bystander' Mø) within the lesions. Our findings show coordinated lysosomal expression and regulation signaling with increased cathepsin and H+-ATPase transcripts are upregulated in infected compared with bystander Mø. Additionally, eukaryotic initiation factor 2 (EIF2) signaling is downregulated in infected compared with bystander Mø, which includes many small and large ribosomal subunit (Rps and Rpl) transcripts being decreased in Mø harboring parasites. Furthermore, we also find EIF2 signaling including EIF, Rps, and Rpl transcripts being downregulated in bystander Mø compared with Mø from naïve skin. These data suggest that both the parasite and the inflammatory host microenvironment affect the transcription of ribosomal machinery in lesional Mø, thereby potentially affecting the ability of these cells to perform translation, protein synthesis, and thus function. Altogether, these results suggest that both the parasite and host inflammatory microenvironment independently drive transcriptional remodeling in Mø during LM infection in vivo.
Collapse
Affiliation(s)
- Gopinath Venugopal
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Jordan T Bird
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
- Arkansas Children's Research Institute, Little Rock, Arkansas 72202
| | - Hayden Roys
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Anne Bowlin
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Lucy Fry
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
- Arkansas Children's Research Institute, Little Rock, Arkansas 72202
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| |
Collapse
|
9
|
Solier S, Müller S, Cañeque T, Versini A, Mansart A, Sindikubwabo F, Baron L, Emam L, Gestraud P, Pantoș GD, Gandon V, Gaillet C, Wu TD, Dingli F, Loew D, Baulande S, Durand S, Sencio V, Robil C, Trottein F, Péricat D, Näser E, Cougoule C, Meunier E, Bègue AL, Salmon H, Manel N, Puisieux A, Watson S, Dawson MA, Servant N, Kroemer G, Annane D, Rodriguez R. A druggable copper-signalling pathway that drives inflammation. Nature 2023; 617:386-394. [PMID: 37100912 PMCID: PMC10131557 DOI: 10.1038/s41586-023-06017-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 03/27/2023] [Indexed: 04/28/2023]
Abstract
Inflammation is a complex physiological process triggered in response to harmful stimuli1. It involves cells of the immune system capable of clearing sources of injury and damaged tissues. Excessive inflammation can occur as a result of infection and is a hallmark of several diseases2-4. The molecular bases underlying inflammatory responses are not fully understood. Here we show that the cell surface glycoprotein CD44, which marks the acquisition of distinct cell phenotypes in the context of development, immunity and cancer progression, mediates the uptake of metals including copper. We identify a pool of chemically reactive copper(II) in mitochondria of inflammatory macrophages that catalyses NAD(H) redox cycling by activating hydrogen peroxide. Maintenance of NAD+ enables metabolic and epigenetic programming towards the inflammatory state. Targeting mitochondrial copper(II) with supformin (LCC-12), a rationally designed dimer of metformin, induces a reduction of the NAD(H) pool, leading to metabolic and epigenetic states that oppose macrophage activation. LCC-12 interferes with cell plasticity in other settings and reduces inflammation in mouse models of bacterial and viral infections. Our work highlights the central role of copper as a regulator of cell plasticity and unveils a therapeutic strategy based on metabolic reprogramming and the control of epigenetic cell states.
Collapse
Affiliation(s)
- Stéphanie Solier
- Equipe Labellisée Ligue Contre le Cancer, Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Sebastian Müller
- Equipe Labellisée Ligue Contre le Cancer, Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Tatiana Cañeque
- Equipe Labellisée Ligue Contre le Cancer, Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Antoine Versini
- Equipe Labellisée Ligue Contre le Cancer, Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Arnaud Mansart
- Paris Saclay University, UVSQ, INSERM, 2I, Montigny-le-Bretonneux, France
| | - Fabien Sindikubwabo
- Equipe Labellisée Ligue Contre le Cancer, Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Leeroy Baron
- Equipe Labellisée Ligue Contre le Cancer, Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Laila Emam
- Paris Saclay University, UVSQ, INSERM, 2I, Montigny-le-Bretonneux, France
| | - Pierre Gestraud
- CBIO-Centre for Computational Biology, Institut Curie, INSERM, Mines ParisTech, Paris, France
| | - G Dan Pantoș
- Department of Chemistry, University of Bath, Bath, UK
| | - Vincent Gandon
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, CNRS, Paris Saclay University, Orsay, France
- Laboratoire de Chimie Moléculaire, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Christine Gaillet
- Equipe Labellisée Ligue Contre le Cancer, Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Ting-Di Wu
- Institut Curie, PSL Research University, Paris, France
- Multimodal Imaging Center, Paris Saclay University, CNRS, INSERM, Orsay, France
| | - Florent Dingli
- CurieCoreTech Mass Spectrometry Proteomic, Institut Curie, PSL Research University, Paris, France
| | - Damarys Loew
- CurieCoreTech Mass Spectrometry Proteomic, Institut Curie, PSL Research University, Paris, France
| | - Sylvain Baulande
- ICGex Next-Generation Sequencing Platform, Institut Curie, PSL Research University, Paris, France
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Valentin Sencio
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, CIIL, Lille, France
| | - Cyril Robil
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, CIIL, Lille, France
| | - François Trottein
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, CIIL, Lille, France
| | - David Péricat
- Institut of Pharmacology and Structural Biology, University of Toulouse, CNRS, Toulouse, France
| | - Emmanuelle Näser
- Institut of Pharmacology and Structural Biology, University of Toulouse, CNRS, Toulouse, France
- Cytometry and Imaging Core facility, Institute of Pharmacology and Structural Biology, University of Toulouse, CNRS, Toulouse, France
| | - Céline Cougoule
- Institut of Pharmacology and Structural Biology, University of Toulouse, CNRS, Toulouse, France
| | - Etienne Meunier
- Institut of Pharmacology and Structural Biology, University of Toulouse, CNRS, Toulouse, France
| | | | - Hélène Salmon
- Institut Curie, INSERM, PSL Research University, Paris, France
| | - Nicolas Manel
- Institut Curie, INSERM, PSL Research University, Paris, France
| | - Alain Puisieux
- Equipe Labellisée Ligue Contre le Cancer, Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Sarah Watson
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France
| | - Mark A Dawson
- Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, Melbourne, Victoria, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicolas Servant
- CBIO-Centre for Computational Biology, Institut Curie, INSERM, Mines ParisTech, Paris, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, University of Paris, Sorbonne University, INSERM, Institut Universitaire de France, Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Djillali Annane
- Paris Saclay University, UVSQ, INSERM, 2I, Montigny-le-Bretonneux, France
- Department of Intensive Care, Hôpital Raymond Poincaré, AP-HP, Garches, France
| | - Raphaël Rodriguez
- Equipe Labellisée Ligue Contre le Cancer, Institut Curie, CNRS, INSERM, PSL Research University, Paris, France.
| |
Collapse
|
10
|
Dual RNA sequencing of group B Streptococcus-infected human monocytes reveals new insights into host-pathogen interactions and bacterial evasion of phagocytosis. Sci Rep 2023; 13:2137. [PMID: 36747074 PMCID: PMC9902490 DOI: 10.1038/s41598-023-28117-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/13/2023] [Indexed: 02/08/2023] Open
Abstract
Streptococcus agalactiae, also known as Group B Streptococcus (GBS) is a frequent cause of infections, including bacteraemia and other acute diseases in adults and immunocompromised individuals. We developed a novel system to study GBS within human monocytes to define the co-transcriptome of intracellular GBS (iGBS) and host cells simultaneously using dual RNA-sequencing (RNA-seq) to better define how this pathogen responds to host cells. Using human U937 monocytes and genome-sequenced GBS reference strain 874,391 in antibiotic protection assays we validated a system for dual-RNA seq based on measures of GBS and monocyte viability to ensure that the bacterial and host cell co-transcriptome reflected mainly intracellular (iGBS) rather than extracellular GBS. Elucidation of the co-transcriptome revealed 1119 dysregulated transcripts in iGBS with most genes, including several that encode virulence factors (e.g., scpB, hvgA, ribD, pil2b) exhibiting activation by upregulated expression. Infection with iGBS resulted in significant remodelling of the monocyte transcriptome, with 7587 transcripts differentially expressed including 7040 up-regulated and 547 down-regulated. qPCR confirmed that the most strongly activated genes included sht, encoding Streptococcal Histidine Triad Protein. An isogenic GBS mutant strain deficient in sht revealed a significant effect of this gene on phagocytosis of GBS and survival of the bacteria during systemic infection in mice. Identification of a novel contribution of sht to GBS virulence shows the co-transcriptome responses elucidated in GBS-infected monocytes help to shape the host-pathogen interaction and establish a role for sht in the response of the bacteria to phagocytic uptake. This study provides comprehension of concurrent transcriptional responses that occur in GBS and human monocytes that shape the host-pathogen interaction.
Collapse
|
11
|
Fernandes JCR, Gonçalves ANA, Floeter-Winter LM, Nakaya HI, Muxel SM. Comparative transcriptomic analysis of long noncoding RNAs in Leishmania-infected human macrophages. Front Genet 2023; 13:1051568. [PMID: 36685903 PMCID: PMC9845402 DOI: 10.3389/fgene.2022.1051568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/25/2022] [Indexed: 01/05/2023] Open
Abstract
It is well established that infection with Leishmania alters the host cell's transcriptome. Since mammalian cells have multiple mechanisms to control gene expression, different molecules, such as noncoding RNAs, can be involved in this process. MicroRNAs have been extensively studied upon Leishmania infection, but whether long noncoding RNAs (lncRNAs) are also altered in macrophages is still unexplored. We performed RNA-seq from THP-1-derived macrophages infected with Leishmania amazonensis (La), L. braziliensis (Lb), and L. infantum (Li), investigating a previously unappreciated fraction of macrophage transcriptome. We found that more than 24% of the total annotated transcripts and 30% of differentially expressed (DE) RNAs in Leishmania-infected macrophage correspond to lncRNAs. LncRNAs and protein coding RNAs with altered expression are similar among macrophages infected with the Leishmania species. Still, some species-specific alterations could occur due to distinct pathophysiology in which Li infection led to a more significant number of exclusively DE RNAs. The most represented classes among DE lncRNAs were intergenic and antisense lncRNAs. We also found enrichment for immune response-related pathways in the DE protein coding RNAs, as well as putative targets of the lncRNAs. We performed a coexpression analysis to explore potential cis regulation of coding and antisense noncoding transcripts. We identified that antisense lncRNAs are similarly regulated as its neighbor protein coding genes, such as the BAALC/BAALC-AS1, BAALC/BAALC-AS2, HIF1A/HIF1A-AS1, HIF1A/HIF1A-AS3 and IRF1/IRF1-AS1 pairs, which can occur as a species-specific modulation. These findings are a novelty in the field because, to date, no study has focused on analyzing lncRNAs in Leishmania-infected macrophage. Our results suggest that lncRNAs may account for a novel mechanism by which Leishmania can control macrophage function. Further research must validate putative lncRNA targets and provide additional prospects in lncRNA function during Leishmania infection.
Collapse
Affiliation(s)
- Juliane C. R. Fernandes
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil,Instituto de Medicina Tropical da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Lucile M. Floeter-Winter
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | | - Sandra M. Muxel
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil,*Correspondence: Sandra M. Muxel,
| |
Collapse
|
12
|
New Insights on Heme Uptake in Leishmania spp. Int J Mol Sci 2022; 23:ijms231810501. [PMID: 36142411 PMCID: PMC9504327 DOI: 10.3390/ijms231810501] [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: 06/20/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/20/2022] Open
Abstract
The protozoan parasite Leishmania, responsible for leishmaniasis, is one of the few aerobic organisms that cannot synthesize the essential molecule heme. Therefore, it has developed specialized pathways to scavenge it from its host. In recent years, some proteins involved in the import of heme, such as LHR1 and LFLVCRB, have been identified, but relevant aspects regarding the process remain unknown. Here, we characterized the kinetics of the uptake of the heme analogue Zn(II) Mesoporphyrin IX (ZnMP) in Leishmania major promastigotes as a model of a parasite causing cutaneous leishmaniasis with special focus on the force that drives the process. We found that ZnMP uptake is an active, inducible, and pH-dependent process that does not require a plasma membrane proton gradient but requires the presence of the monovalent cations Na+ and/or K+. In addition, we demonstrated that this parasite can efflux this porphyrin against a concentration gradient. We also found that ZnMP uptake differs among different dermotropic or viscerotropic Leishmania species and does not correlate with LHR1 or LFLVCRB expression levels. Finally, we showed that these transporters have only partially overlapping functions. Altogether, these findings contribute to a deeper understanding of an important process in the biology of this parasite.
Collapse
|
13
|
Bekkar A, Isorce N, Snäkä T, Claudinot S, Desponds C, Kopelyanskiy D, Prével F, Reverte M, Xenarios I, Fasel N, Teixeira F. Dissection of the macrophage response towards infection by the Leishmania-viral endosymbiont duo and dynamics of the type I interferon response. Front Cell Infect Microbiol 2022; 12:941888. [PMID: 35992159 PMCID: PMC9386148 DOI: 10.3389/fcimb.2022.941888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022] Open
Abstract
Leishmania RNA virus 1 (LRV1) is a double-stranded RNA virus found in some strains of the human protozoan parasite Leishmania, the causative agent of leishmaniasis, a neglected tropical disease. Interestingly, the presence of LRV1 inside Leishmania constitutes an important virulence factor that worsens the leishmaniasis outcome in a type I interferon (IFN)–dependent manner and contributes to treatment failure. Understanding how macrophages respond toward Leishmania alone or in combination with LRV1 as well as the role that type I IFNs may play during infection is fundamental to oversee new therapeutic strategies. To dissect the macrophage response toward infection, RNA sequencing was performed on murine wild-type and Ifnar-deficient bone marrow–derived macrophages infected with Leishmania guyanensis (Lgy) devoid or not of LRV1. Additionally, macrophages were treated with poly I:C (mimetic virus) or with type I IFNs. By implementing a weighted gene correlation network analysis, the groups of genes (modules) with similar expression patterns, for example, functionally related, coregulated, or the members of the same functional pathway, were identified. These modules followed patterns dependent on Leishmania, LRV1, or Leishmania exacerbated by the presence of LRV1. Not only the visualization of how individual genes were embedded to form modules but also how different modules were related to each other were observed. Thus, in the context of the observed hyperinflammatory phenotype associated to the presence of LRV1, it was noted that the biomarkers tumor-necrosis factor α (TNF-α) and the interleukin 6 (IL-6) belonged to different modules and that their regulating specific Src-family kinases were segregated oppositely. In addition, this network approach revealed the strong and sustained effect of LRV1 on the macrophage response and genes that had an early, late, or sustained impact during infection, uncovering the dynamics of the IFN response. Overall, this study contributed to shed light and dissect the intricate macrophage response toward infection by the Leishmania-LRV1 duo and revealed the crosstalk between modules made of coregulated genes and provided a new resource that can be further explored to study the impact of Leishmania on the macrophage response.
Collapse
Affiliation(s)
- Amel Bekkar
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Nathalie Isorce
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Tiia Snäkä
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Florence Prével
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Marta Reverte
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Ioannis Xenarios
- Agora Center, Center Hospitalier Universitaire (CHUV), Lausanne, Switzerland
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Fasel
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- *Correspondence: Nicolas Fasel, ; Filipa Teixeira,
| | - Filipa Teixeira
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- *Correspondence: Nicolas Fasel, ; Filipa Teixeira,
| |
Collapse
|
14
|
Venugopal G, Bird JT, Washam CL, Roys H, Bowlin A, Byrum SD, Weinkopff T. In vivo transcriptional analysis of mice infected with Leishmania major unveils cellular heterogeneity and altered transcriptomic profiling at single-cell resolution. PLoS Negl Trop Dis 2022; 16:e0010518. [PMID: 35789215 PMCID: PMC9286232 DOI: 10.1371/journal.pntd.0010518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 07/15/2022] [Accepted: 05/18/2022] [Indexed: 01/02/2023] Open
Abstract
Leishmania parasites cause cutaneous leishmaniasis (CL), a disease characterized by disfiguring, ulcerative skin lesions. Both parasite and host gene expression following infection with various Leishmania species has been investigated in vitro, but global transcriptional analysis following L. major infection in vivo is lacking. Thus, we conducted a comprehensive transcriptomic profiling study combining bulk RNA sequencing (RNA-Seq) and single-cell RNA sequencing (scRNA-Seq) to identify global changes in gene expression in vivo following L. major infection. Bulk RNA-Seq analysis revealed that host immune response pathways like the antigen processing and presentation pathway were significantly enriched amongst differentially expressed genes (DEGs) upon infection, while ribosomal pathways were significantly downregulated in infected mice compared to naive controls. scRNA-Seq analyses revealed cellular heterogeneity including distinct resident and recruited cell types in the skin following murine L. major infection. Within the individual immune cell types, several DEGs indicative of many interferon induced GTPases and antigen presentation molecules were significantly enhanced in the infected ears including macrophages, resident macrophages, and inflammatory monocytes. Ingenuity Pathway Analysis of scRNA-Seq data indicated the antigen presentation pathway was increased with infection, while EIF2 signaling is the top downregulated pathway followed by eIF4/p70S6k and mTOR signaling in multiple cell types including macrophages, blood and lymphatic endothelial cells. Altogether, this transcriptomic profile highlights known recruitment of myeloid cells to lesions and recognizes a potential role for EIF2 signaling in murine L. major infection in vivo.
Collapse
Affiliation(s)
- Gopinath Venugopal
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jordan T. Bird
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Research Institute, Little Rock, Arkansas, United States of America
| | - Charity L. Washam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Research Institute, Little Rock, Arkansas, United States of America
| | - Hayden Roys
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Anne Bowlin
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Research Institute, Little Rock, Arkansas, United States of America
- * E-mail: (SDB); (TW)
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail: (SDB); (TW)
| |
Collapse
|
15
|
Transcriptome Analysis of Intracellular Amastigotes of Clinical Leishmania infantum Lines from Therapeutic Failure Patients after Infection of Human Macrophages. Microorganisms 2022; 10:microorganisms10071304. [PMID: 35889023 PMCID: PMC9324091 DOI: 10.3390/microorganisms10071304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
Leishmaniasis is considered to be one of the most neglected tropical diseases affecting humans and animals around the world. Due to the absence of an effective vaccine, current treatment is based on chemotherapy. However, the continuous appearance of drug resistance and therapeutic failure (TF) lead to an early obsolescence of treatments. Identification of the factors that contribute to TF and drug resistance in leishmaniasis will constitute a useful tool for establishing future strategies to control this disease. In this manuscript, we evaluated the transcriptomic changes in the intracellular amastigotes of the Leishmania infantum parasites isolated from patients with leishmaniasis and TF at 96 h post-infection of THP-1 cells. The adaptation of the parasites to their new environment leads to expression alterations in the genes involved mainly in the transport through cell membranes, energy and redox metabolism, and detoxification. Specifically, the gene that codes for the prostaglandin f2α synthase seems to be relevant in the pathogenicity and TF since it appears substantially upregulated in all the L. infantum lines. Overall, our results show that at the late infection timepoint, the transcriptome of the parasites undergoes significant changes that probably improve the survival of the Leishmania lines in the host cells, contributing to the TF phenotype as well as drug therapy evasion.
Collapse
|
16
|
Margaroni M, Agallou M, Vasilakaki A, Karagkouni D, Skoufos G, Hatzigeorgiou AG, Karagouni E. Transcriptional Profiling of Leishmania infantum Infected Dendritic Cells: Insights into the Role of Immunometabolism in Host-Parasite Interaction. Microorganisms 2022; 10:microorganisms10071271. [PMID: 35888991 PMCID: PMC9322131 DOI: 10.3390/microorganisms10071271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
Leishmania parasites are capable of effectively invading dendritic cells (DCs), a cell population orchestrating immune responses against several diseases, including leishmaniasis, by bridging innate and adaptive immunity. Leishmania on the other hand has evolved various mechanisms to subvert DCs activation and establish infection. Thus, the transcriptional profile of DCs derived from bone marrow (BMDCs) that have been infected with Leishmania infantum parasite or of DCs exposed to chemically inactivated parasites was investigated via RNA sequencing, aiming to better understand the host–pathogen interplay. Flow cytometry analysis revealed that L. infantum actively inhibits maturation of not only infected but also bystander BMDCs. Analysis of double-sorted L. infantum infected BMDCs revealed significantly increased expression of genes mainly associated with metabolism and particularly glycolysis. Moreover, differentially expressed genes (DEGs) related to DC-T cell interactions were also found to be upregulated exclusively in infected BMDCs. On the contrary, transcriptome analysis of fixed parasites containing BMDCs indicated that energy production was mediated through TCA cycle and oxidative phosphorylation. In addition, DEGs related to differentiation of DCs leading to activation and differentiation of Th17 subpopulations were detected. These findings suggest an important role of metabolism on DCs-Leishmania interplay and eventually disease establishment.
Collapse
Affiliation(s)
- Maritsa Margaroni
- Immunology of Infection Laboratory, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece; (M.M.); (M.A.); (A.V.)
| | - Maria Agallou
- Immunology of Infection Laboratory, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece; (M.M.); (M.A.); (A.V.)
| | - Athina Vasilakaki
- Immunology of Infection Laboratory, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece; (M.M.); (M.A.); (A.V.)
| | - Dimitra Karagkouni
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece; (D.K.); (G.S.); (A.G.H.)
- Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Giorgos Skoufos
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece; (D.K.); (G.S.); (A.G.H.)
- Hellenic Pasteur Institute, 11521 Athens, Greece
- Department of Electrical & Computer Engineering, University of Thessaly, 38221 Volos, Greece
| | - Artemis G. Hatzigeorgiou
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece; (D.K.); (G.S.); (A.G.H.)
- Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Evdokia Karagouni
- Immunology of Infection Laboratory, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece; (M.M.); (M.A.); (A.V.)
- Correspondence: ; Tel.: +30-21-0647-8826
| |
Collapse
|
17
|
Gonçalves LO, Pulido AFV, Mathias FAS, Enes AES, Carvalho MGR, de Melo Resende D, Polak ME, Ruiz JC. Expression Profile of Genes Related to the Th17 Pathway in Macrophages Infected by Leishmania major and Leishmania amazonensis: The Use of Gene Regulatory Networks in Modeling This Pathway. Front Cell Infect Microbiol 2022; 12:826523. [PMID: 35774406 PMCID: PMC9239034 DOI: 10.3389/fcimb.2022.826523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Leishmania amazonensis and Leishmania major are the causative agents of cutaneous and mucocutaneous diseases. The infections‘ outcome depends on host–parasite interactions and Th1/Th2 response, and in cutaneous form, regulation of Th17 cytokines has been reported to maintain inflammation in lesions. Despite that, the Th17 regulatory scenario remains unclear. With the aim to gain a better understanding of the transcription factors (TFs) and genes involved in Th17 induction, in this study, the role of inducing factors of the Th17 pathway in Leishmania–macrophage infection was addressed through computational modeling of gene regulatory networks (GRNs). The Th17 GRN modeling integrated experimentally validated data available in the literature and gene expression data from a time-series RNA-seq experiment (4, 24, 48, and 72 h post-infection). The generated model comprises a total of 10 TFs, 22 coding genes, and 16 cytokines related to the Th17 immune modulation. Addressing the Th17 induction in infected and uninfected macrophages, an increase of 2- to 3-fold in 4–24 h was observed in the former. However, there was a decrease in basal levels at 48–72 h for both groups. In order to evaluate the possible outcomes triggered by GRN component modulation in the Th17 pathway. The generated GRN models promoted an integrative and dynamic view of Leishmania–macrophage interaction over time that extends beyond the analysis of single-gene expression.
Collapse
Affiliation(s)
- Leilane Oliveira Gonçalves
- Programa de Pós-graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- Grupo Informática de Biossistemas, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Brazil
| | - Andrés F. Vallejo Pulido
- Systems Immunology Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | | - Alexandre Estevão Silvério Enes
- Programa de Pós-graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- Grupo Informática de Biossistemas, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Brazil
| | | | - Daniela de Melo Resende
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Brazil
| | - Marta E. Polak
- Systems Immunology Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- *Correspondence: Jeronimo C. Ruiz, ; Marta E. Polak,
| | - Jeronimo C. Ruiz
- Grupo Informática de Biossistemas, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Brazil
- *Correspondence: Jeronimo C. Ruiz, ; Marta E. Polak,
| |
Collapse
|
18
|
Kent RS, Briggs EM, Colon BL, Alvarez C, Silva Pereira S, De Niz M. Paving the Way: Contributions of Big Data to Apicomplexan and Kinetoplastid Research. Front Cell Infect Microbiol 2022; 12:900878. [PMID: 35734575 PMCID: PMC9207352 DOI: 10.3389/fcimb.2022.900878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
In the age of big data an important question is how to ensure we make the most out of the resources we generate. In this review, we discuss the major methods used in Apicomplexan and Kinetoplastid research to produce big datasets and advance our understanding of Plasmodium, Toxoplasma, Cryptosporidium, Trypanosoma and Leishmania biology. We debate the benefits and limitations of the current technologies, and propose future advancements that may be key to improving our use of these techniques. Finally, we consider the difficulties the field faces when trying to make the most of the abundance of data that has already been, and will continue to be, generated.
Collapse
Affiliation(s)
- Robyn S. Kent
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, United States
| | - Emma M. Briggs
- Institute for Immunology and Infection Research, School of Biological Sciences, University Edinburgh, Edinburgh, United Kingdom
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Beatrice L. Colon
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Catalina Alvarez
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Sara Silva Pereira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Mariana De Niz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Institut Pasteur, Paris, France
| |
Collapse
|
19
|
Gupta AK, Das S, Kamran M, Ejazi SA, Ali N. The Pathogenicity and Virulence of Leishmania - interplay of virulence factors with host defenses. Virulence 2022; 13:903-935. [PMID: 35531875 PMCID: PMC9154802 DOI: 10.1080/21505594.2022.2074130] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Leishmaniasis is a group of disease caused by the intracellular protozoan parasite of the genus Leishmania. Infection by different species of Leishmania results in various host immune responses, which usually lead to parasite clearance and may also contribute to pathogenesis and, hence, increasing the complexity of the disease. Interestingly, the parasite tends to reside within the unfriendly environment of the macrophages and has evolved various survival strategies to evade or modulate host immune defense. This can be attributed to the array of virulence factors of the vicious parasite, which target important host functioning and machineries. This review encompasses a holistic overview of leishmanial virulence factors, their role in assisting parasite-mediated evasion of host defense weaponries, and modulating epigenetic landscapes of host immune regulatory genes. Furthermore, the review also discusses the diagnostic potential of various leishmanial virulence factors and the advent of immunomodulators as futuristic antileishmanial drug therapy.
Collapse
Affiliation(s)
- Anand Kumar Gupta
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sonali Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Mohd Kamran
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sarfaraz Ahmad Ejazi
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| |
Collapse
|
20
|
Forrester S, Goundry A, Dias BT, Leal-Calvo T, Moraes MO, Kaye PM, Mottram JC, Lima APCA. Tissue Specific Dual RNA-Seq Defines Host-Parasite Interplay in Murine Visceral Leishmaniasis Caused by Leishmania donovani and Leishmania infantum. Microbiol Spectr 2022; 10:e0067922. [PMID: 35384718 PMCID: PMC9045295 DOI: 10.1128/spectrum.00679-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 01/22/2023] Open
Abstract
Visceral leishmaniasis is associated with hepato-splenomegaly and altered immune and hematological parameters in both preclinical animal models and humans. We studied mouse experimental visceral leishmaniasis caused by Leishmania infantum and Leishmania donovani in BALB/c mice using dual RNA-seq to investigate the transcriptional response of host and parasite in liver and spleen. We identified only 4 species-specific parasite expressed genes (SSPEGs; log2FC >1, FDR <0.05) in the infected spleen, and none in the infected liver. For the host transcriptome, we found 789 differentially expressed genes (DEGs; log2FC >1, FDR <0.05) in the spleen that were common to both infections, with IFNγ signaling and complement and coagulation cascade pathways highly enriched, and an additional 286 and 186 DEGs that were selective to L. donovani and L. infantum infection, respectively. Among those, there were network interactions between genes of amino acid metabolism and PPAR signaling in L. donovani infection and increased IL1β and positive regulation of fatty acid transport in L. infantum infection, although no pathway enrichment was observed. In the liver, there were 1,939 DEGs in mice infected with either L. infantum or L. donovani in comparison to uninfected mice, and the most enriched pathways were IFNγ signaling, neutrophil mediated immunity, complement and coagulation, cytokine-chemokine responses, and hemostasis. Additionally, 221 DEGs were selective in L. donovani and 429 DEGs in L. infantum infections. These data show that the host response for these two visceral leishmaniasis infection models is broadly similar, and ∼10% of host DEGs vary in infections with either parasite species. IMPORTANCE Visceral leishmaniasis (VL) is caused by two species of Leishmania parasites, L. donovani in the Old World and L. infantum in the New World and countries bordering the Mediterranean. Although cardinal features such as hepato-splenomegaly and alterations in blood and immune function are evident, clinical presentation may vary by geography, with for example severe bleeding often associated with VL in Brazil. Although animal models of both L. donovani and L. infantum have been widely used to study disease pathogenesis, a direct side-by-side comparison of how these parasites species impact the infected host and/or how they might respond to the stresses of mammalian infection has not been previously reported. Identifying common and distinct pathways to pathogenesis will be important to ensure that new therapeutic or prophylactic approaches will be applicable across all forms of VL.
Collapse
Affiliation(s)
- Sarah Forrester
- York Biomedical Research Institute, Department of Biology, University of York, York, England, United Kingdom
| | - Amy Goundry
- York Biomedical Research Institute, Department of Biology, University of York, York, England, United Kingdom
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna Torres Dias
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, England, United Kingdom
| | - Jeremy C. Mottram
- York Biomedical Research Institute, Department of Biology, University of York, York, England, United Kingdom
| | - Ana Paula C. A. Lima
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Chaparro V, Graber TE, Alain T, Jaramillo M. Transcriptional profiling of macrophages reveals distinct parasite stage-driven signatures during early infection by Leishmania donovani. Sci Rep 2022; 12:6369. [PMID: 35430587 PMCID: PMC9013368 DOI: 10.1038/s41598-022-10317-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/05/2022] [Indexed: 12/22/2022] Open
Abstract
Macrophages undergo swift changes in mRNA abundance upon pathogen invasion. Herein we describe early remodelling of the macrophage transcriptome during infection by amastigotes or promastigotes of Leishmania donovani. Approximately 10–16% of host mRNAs were differentially modulated in L. donovani-infected macrophages when compared to uninfected controls. This response was partially stage-specific as a third of changes in mRNA abundance were either exclusively driven by one of the parasite forms or significantly different between them. Gene ontology analyses identified categories associated with immune functions (e.g. antigen presentation and leukocyte activation) among significantly downregulated mRNAs during amastigote infection while cytoprotective-related categories (e.g. DNA repair and apoptosis inhibition) were enriched in upregulated transcripts. Interestingly a combination of upregulated (e.g. cellular response to IFNβ) and repressed (e.g. leukocyte activation, chemotaxis) immune-related transcripts were overrepresented in the promastigote-infected dataset. In addition, Ingenuity Pathway Analysis (IPA) associated specific mRNA subsets with a number of upstream transcriptional regulators predicted to be modulated in macrophages infected with L. donovani amastigotes (e.g. STAT1 inhibition) or promastigotes (e.g. NRF2, IRF3, and IRF7 activation). Overall, our results indicate that early parasite stage-driven transcriptional remodelling in macrophages contributes to orchestrate both protective and deleterious host cell responses during L. donovani infection.
Collapse
|
22
|
Perea-Martínez A, García-Hernández R, Manzano JI, Gamarro F. Transcriptomic Analysis in Human Macrophages Infected with Therapeutic Failure Clinical Isolates of Leishmania infantum. ACS Infect Dis 2022; 8:800-810. [PMID: 35352952 PMCID: PMC9003231 DOI: 10.1021/acsinfecdis.1c00513] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Leishmaniasis is one of the neglected tropical diseases with a worldwide distribution, affecting humans and animals. In the absence of an effective vaccine, current treatment is through the use of chemotherapy; however, existing treatments have frequent appearance of drug resistance and therapeutic failure (TF). The identification of factors that contribute to TF in leishmaniasis will provide the basis for a future therapeutic strategy more efficient for the control of this disease. In this article, we have evaluated the transcriptomic changes in the host cells THP-1 after infection with clinical Leishmania infantum isolates from leishmaniasis patients with TF. Our results show that distinct L. infantum isolates differentially modulate host cell response, inducing phenotypic changes that probably may account for parasite survival and TF of patients. Analysis of differential expression genes (DEGs), with a statistical significance threshold of a fold change ≥ 2 and a false discovery rate value ≤ 0.05, revealed a different number of DEGs according to the Leishmanialine. Globally, there was a similar number of genes up- and downregulated in all the infected host THP-1 cells, with exception of Hi-L2221, which showed a higher number of downregulated DEGs. We observed a total of 58 DEGs commonly modulated in all infected host cells, including upregulated (log2FC ≥ 1) and downregulated (log2FC ≤ -1) genes. Based on the results obtained from the analysis of RNA-seq, volcano plot, and GO enrichment analysis, we identified the most significant transcripts of relevance for their possible contribution to the TF observed in patients with leishmaniasis.
Collapse
Affiliation(s)
- Ana Perea-Martínez
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Avda del Conocimiento 17, 18016 Armilla, Granada, Spain
| | - Raquel García-Hernández
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Avda del Conocimiento 17, 18016 Armilla, Granada, Spain
| | - José Ignacio Manzano
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Avda del Conocimiento 17, 18016 Armilla, Granada, Spain
| | - Francisco Gamarro
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Avda del Conocimiento 17, 18016 Armilla, Granada, Spain
| |
Collapse
|
23
|
Taslimi Y, Masoudzadeh N, Bahrami F, Rafati S. Cutaneous leishmaniasis: multiomics approaches to unravel the role of immune cells checkpoints. Expert Rev Proteomics 2022; 19:213-225. [PMID: 36191333 DOI: 10.1080/14789450.2022.2131545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Cutaneous leishmaniasis (CL) is the most frequent form of leishmaniases, associated with skin inflammation and ulceration. Understanding the interaction of different phagocytic cells in the recognition and uptake of different Leishmania species is critical for controlling the infection. Phagocytic cells have a pivotal role as professional antigen-presenting cells that bridge the innate and adaptive immunity and shape the outcome of the disease. AREAS COVERED Here we reviewed new technologies with high-throughput data collection capabilities along with systems biology approaches which are recently being used to decode the paradox of CL immunology. EXPERT OPINION We emphasized on the crosstalk between DC and T-cells while focusing on the immune checkpoints interactions between the human immune system and the Leishmania species. Further, we discussed omics technologies including bulk RNA sequencing, reverse transcriptase-multiplex ligation dependent probe amplification (RT-MLPA), and proximity extension assay (PEA) in studies on human blood or tissue-driven samples from CL patients in which we have so far been involved.
Collapse
Affiliation(s)
- Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran Iran
| | - Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran Iran
| | - Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran Iran
| |
Collapse
|
24
|
García-Hernández R, Manzano JI, Perea-Martínez A, Gamarro F. New Insights on Drug-Resistant Clinical Isolates of Leishmania infantum-Infected Human Macrophages as Determined by Comparative Transcriptome Analyses. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:165-177. [PMID: 35172107 DOI: 10.1089/omi.2021.0185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Leishmaniasis is the second most important neglected tropical parasitic disease after malaria. This disease is distributed worldwide and can be present in a variety of clinical forms, depending on the parasite species and host's genetic background. As chemotherapy is the only effective weapon whose effectiveness is limited by the frequent appearance of drug resistance and therapeutic failure, new therapeutic strategies are required. To better understand the factors that contribute to therapeutic failure and drug resistance in leishmaniasis, we studied the transcriptomic changes in host THP-1 cells after infection with clinical Leishmania infantum isolates with different susceptibilities to antileishmanial drugs by RNA-seq. Analysis of the differentially expressed genes (DEGs) in infected host cells revealed variations in DEG numbers in the THP-1-infected cells depending on the Leishmania line. A key conclusion of this study is that the modulation of host cells is Leishmania line dependent. Gene ontology enrichment analyses of DEGs indicated that certain biological processes were modulated in the infected host cells, specifically related to cellular metabolism, immune response, defense response, signaling pathways, and cell proliferation and apoptosis. Furthermore, this study provides new potential therapeutic markers and insights into the THP-1 host transcriptomic changes that occur after late infection with drug-resistant L. infantum clinical isolates.
Collapse
Affiliation(s)
| | - José Ignacio Manzano
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), Granada, Spain
| | - Ana Perea-Martínez
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), Granada, Spain
| | - Francisco Gamarro
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), Granada, Spain
| |
Collapse
|
25
|
Mas A, Martínez-Rodrigo A, Carrión J, Orden JA, Alzate JF, Domínguez-Bernal G, Horcajo P. Transcriptomic Profile of Canine DH82 Macrophages Infected by Leishmania infantum Promastigotes with Different Virulence Behavior. Int J Mol Sci 2022; 23:ijms23031466. [PMID: 35163386 PMCID: PMC8835757 DOI: 10.3390/ijms23031466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 02/07/2023] Open
Abstract
Zoonotic visceral leishmaniosis caused by Leishmania infantum is an endemic disease in the Mediterranean Basin affecting mainly humans and dogs, the main reservoir. The leishmaniosis outbreak declared in the Community of Madrid (Spain) led to a significant increase in human disease incidence without enhancing canine leishmaniosis prevalence, suggesting a better adaptation of the outbreak's isolates by other host species. One of the isolates obtained in the focus, IPER/ES/2012/BOS1FL1 (BOS1FL1), has previously demonstrated a different phenotype than the reference strain MCAN/ES/1996/BCN150 (BCN150), characterized by a lower infectivity when interacting with canine macrophages. Nevertheless, not enough changes in the cell defensive response were found to support their different behavior. Thus, we decided to investigate the molecular mechanisms involved in the interaction of both parasites with DH82 canine macrophages by studying their transcriptomic profiles developed after infection using RNA sequencing. The results showed a common regulation induced by both parasites in the phosphoinositide-3-kinase-protein kinase B/Akt and NOD-like receptor signaling pathways. However, other pathways, such as phagocytosis and signal transduction, including tumor necrosis factor, mitogen-activated kinases and nuclear factor-κB, were only regulated after infection with BOS1FL1. These differences could contribute to the reduced infection ability of the outbreak isolates in canine cells. Our results open a new avenue to investigate the true role of adaptation of L. infantum isolates in their interaction with their different hosts.
Collapse
Affiliation(s)
- Alicia Mas
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
| | - Abel Martínez-Rodrigo
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
| | - Javier Carrión
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
| | - José Antonio Orden
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
| | - Juan F. Alzate
- Centro Nacional de Secuenciación Genómica-CNSG, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia, Medellín 050010, Colombia;
| | - Gustavo Domínguez-Bernal
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.); (A.M.-R.); (J.C.); (J.A.O.)
- Correspondence: ; Tel.: +34-913943814
| | - Pilar Horcajo
- Animal Health and Zoonoses (SALUVET) Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, 28040 Madrid, Spain;
| |
Collapse
|
26
|
Smirlis D, Dingli F, Sabatet V, Roth A, Knippschild U, Loew D, Späth GF, Rachidi N. Identification of the Host Substratome of Leishmania-Secreted Casein Kinase 1 Using a SILAC-Based Quantitative Mass Spectrometry Assay. Front Cell Dev Biol 2022; 9:800098. [PMID: 35047509 PMCID: PMC8762337 DOI: 10.3389/fcell.2021.800098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/13/2021] [Indexed: 12/27/2022] Open
Abstract
Leishmaniasis is a severe public health problem, caused by the protozoan Leishmania. This parasite has two developmental forms, extracellular promastigote in the insect vector and intracellular amastigote in the mammalian host where it resides inside the phagolysosome of macrophages. Little is known about the virulence factors that regulate host-pathogen interactions and particularly host signalling subversion. All the proteomes of Leishmania extracellular vesicles identified the presence of Leishmania casein kinase 1 (L-CK1.2), a signalling kinase. L-CK1.2 is essential for parasite survival and thus might be essential for host subversion. To get insights into the functions of L-CK1.2 in the macrophage, the systematic identification of its host substrates is crucial, we thus developed an easy method to identify substrates, combining phosphatase treatment, in vitro kinase assay and Stable Isotope Labelling with Amino acids in Cell (SILAC) culture-based mass spectrometry. Implementing this approach, we identified 225 host substrates as well as a potential novel phosphorylation motif for CK1. We confirmed experimentally the enrichment of our substratome in bona fide L-CK1.2 substrates and showed they were also phosphorylated by human CK1δ. L-CK1.2 substratome is enriched in biological processes such as "viral and symbiotic interaction," "actin cytoskeleton organisation" and "apoptosis," which are consistent with the host pathways modified by Leishmania upon infection, suggesting that L-CK1.2 might be the missing link. Overall, our results generate important mechanistic insights into the signalling of host subversion by these parasites and other microbial pathogens adapted for intracellular survival.
Collapse
Affiliation(s)
- Despina Smirlis
- Institut Pasteur, Université de Paris, Institut National de Santé et Recherche Médicale INSERM U1201, Unité de parasitologie Moléculaire et Signalisation, Paris, France.,Hellenic Pasteur Institute, Athens, Greece
| | - Florent Dingli
- Laboratoire de Spectrométrie de Masse Protéomique (LSMP), Centre de Recherche, Institut Curie, PSL Research University, Paris, France
| | - Valentin Sabatet
- Laboratoire de Spectrométrie de Masse Protéomique (LSMP), Centre de Recherche, Institut Curie, PSL Research University, Paris, France
| | - Aileen Roth
- Department of General and Visceral Surgery, Centre of Surgery, University Hospital Ulm, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Centre of Surgery, University Hospital Ulm, Ulm, Germany
| | - Damarys Loew
- Laboratoire de Spectrométrie de Masse Protéomique (LSMP), Centre de Recherche, Institut Curie, PSL Research University, Paris, France
| | - Gerald F Späth
- Institut Pasteur, Université de Paris, Institut National de Santé et Recherche Médicale INSERM U1201, Unité de parasitologie Moléculaire et Signalisation, Paris, France
| | - Najma Rachidi
- Institut Pasteur, Université de Paris, Institut National de Santé et Recherche Médicale INSERM U1201, Unité de parasitologie Moléculaire et Signalisation, Paris, France
| |
Collapse
|
27
|
Ibeagha-Awemu EM, Bissonnette N, Bhattarai S, Wang M, Dudemaine PL, McKay S, Zhao X. Whole Genome Methylation Analysis Reveals Role of DNA Methylation in Cow's Ileal and Ileal Lymph Node Responses to Mycobacterium avium subsp. paratuberculosis Infection. Front Genet 2021; 12:797490. [PMID: 34992636 PMCID: PMC8724574 DOI: 10.3389/fgene.2021.797490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Johne's Disease (JD), caused by Mycobacterium avium subsp paratuberculosis (MAP), is an incurable disease of ruminants and other animal species and is characterized by an imbalance of gut immunity. The role of MAP infection on the epigenetic modeling of gut immunity during the progression of JD is still unknown. This study investigated the DNA methylation patterns in ileal (IL) and ileal lymph node (ILLN) tissues from cows diagnosed with persistent subclinical MAP infection over a one to 4 years period. DNA samples from IL and ILLN tissues from cows negative (MAPneg) (n = 3) or positive for MAP infection (MAPinf) (n = 4) were subjected to whole genome bisulfite sequencing. A total of 11,263 and 62,459 differentially methylated cytosines (DMCs), and 1259 and 8086 differentially methylated regions (DMRs) (FDR<0.1) were found between MAPinf and MAPneg IL and ILLN tissues, respectively. The DMRs were found on 394 genes (denoted DMR genes) in the IL and on 1305 genes in the ILLN. DMR genes with hypermethylated promoters/5'UTR [3 (IL) and 88 (ILLN)] or hypomethylated promoters/5'UTR [10 (IL) and 25 (ILLN)] and having multiple functions including response to stimulus/immune response (BLK, BTC, CCL21, AVPR1A, CHRNG, GABRA4, TDGF1), cellular processes (H2AC20, TEX101, GLA, NCKAP5L, RBM27, SLC18A1, H2AC20BARHL2, NLGN3, SUV39H1, GABRA4, PPA1, UBE2D2) and metabolic processes (GSTO2, H2AC20, SUV39H1, PPA1, UBE2D2) are potential DNA methylation candidate genes of MAP infection. The ILLN DMR genes were enriched for more biological process (BP) gene ontology (GO) terms (n = 374), most of which were related to cellular processes (27.6%), biological regulation (16.6%), metabolic processes (15.4%) and response to stimulus/immune response (8.2%) compared to 75 BP GO terms (related to cellular processes, metabolic processes and transport, and system development) enriched for IL DMR genes. ILLN DMR genes were enriched for more pathways (n = 47) including 13 disease pathways compared with 36 enriched pathways, including 7 disease/immune pathways for IL DMR genes. In conclusion, the results show tissue specific responses to MAP infection with more epigenetic changes (DMCs and DMRs) in the ILLN than in the IL tissue, suggesting that the ILLN and immune processes were more responsive to regulation by methylation of DNA relative to IL tissue. Our data is the first to demonstrate a potential role for DNA methylation in the pathogenesis of MAP infection in dairy cattle.
Collapse
Affiliation(s)
- Eveline M. Ibeagha-Awemu
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada
| | - Nathalie Bissonnette
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada
| | - Suraj Bhattarai
- Department of Animal and Veterinary Sciences, University of Vermont, Burlington, VT, United States
| | - Mengqi Wang
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada
| | - Pier-Luc Dudemaine
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada
| | - Stephanie McKay
- Department of Animal and Veterinary Sciences, University of Vermont, Burlington, VT, United States
| | - Xin Zhao
- Department of Animal Science, McGill University, Ste-Anne-Be-Bellevue, QC, Canada
| |
Collapse
|
28
|
Network-Based Approaches Reveal Potential Therapeutic Targets for Host-Directed Antileishmanial Therapy Driving Drug Repurposing. Microbiol Spectr 2021; 9:e0101821. [PMID: 34668739 PMCID: PMC8528132 DOI: 10.1128/spectrum.01018-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Leishmania parasites are the causal agent of leishmaniasis, an endemic disease in more than 90 countries worldwide. Over the years, traditional approaches focused on the parasite when developing treatments against leishmaniasis. Despite numerous attempts, there is not yet a universal treatment, and those available have allowed for the appearance of resistance. Here, we propose and follow a host-directed approach that aims to overcome the current lack of treatment. Our approach identifies potential therapeutic targets in the host cell and proposes known drug interactions aiming to improve the immune response and to block the host machinery necessary for the survival of the parasite. We started analyzing transcription factor regulatory networks of macrophages infected with Leishmania major. Next, based on the regulatory dynamics of the infection and available gene expression profiles, we selected potential therapeutic target proteins. The function of these proteins was then analyzed following a multilayered network scheme in which we combined information on metabolic pathways with known drugs that have a direct connection with the activity carried out by these proteins. Using our approach, we were able to identify five host protein-coding gene products that are potential therapeutic targets for treating leishmaniasis. Moreover, from the 11 drugs known to interact with the function performed by these proteins, 3 have already been tested against this parasite, verifying in this way our novel methodology. More importantly, the remaining eight drugs previously employed to treat other diseases, remain as promising yet-untested antileishmanial therapies. IMPORTANCE This work opens a new path to fight parasites by targeting host molecular functions by repurposing available and approved drugs. We created a novel approach to identify key proteins involved in any biological process by combining gene regulatory networks and expression profiles. Once proteins have been selected, our approach employs a multilayered network methodology that relates proteins to functions to drugs that alter these functions. By applying our novel approach to macrophages during the Leishmania infection process, we both validated our work and found eight drugs already approved for use in humans that to the best of our knowledge were never employed to treat leishmaniasis, rendering our work as a new tool in the box available to the scientific community fighting parasites.
Collapse
|
29
|
Sandholt AKS, Wattrang E, Lilja T, Ahola H, Lundén A, Troell K, Svärd SG, Söderlund R. Dual RNA-seq transcriptome analysis of caecal tissue during primary Eimeria tenella infection in chickens. BMC Genomics 2021; 22:660. [PMID: 34521339 PMCID: PMC8438895 DOI: 10.1186/s12864-021-07959-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/29/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Coccidiosis is an infectious disease with large negative impact on the poultry industry worldwide. It is an enteric infection caused by unicellular Apicomplexan parasites of the genus Eimeria. The present study aimed to gain more knowledge about interactions between parasites and the host immune system during the early asexual replication phase of E. tenella in chicken caeca. For this purpose, chickens were experimentally infected with E. tenella oocysts, sacrificed on days 1-4 and 10 after infection and mRNA from caecal tissues was extracted and sequenced. RESULTS Dual RNA-seq analysis revealed time-dependent changes in both host and parasite gene expression during the course of the infection. Chicken immune activation was detected from day 3 and onwards with the highest number of differentially expressed immune genes recorded on day 10. Among early (days 3-4) responses up-regulation of genes for matrix metalloproteinases, several chemokines, interferon (IFN)-γ along with IFN-stimulated genes GBP, IRF1 and RSAD2 were noted. Increased expression of genes with immune suppressive/regulatory effects, e.g. IL10, SOCS1, SOCS3, was also observed among early responses. For E. tenella a general up-regulation of genes involved in protein expression and energy metabolism as well as a general down-regulation genes for DNA and RNA processing were observed during the infection. Specific E. tenella genes with altered expression during the experiment include those for proteins in rhoptry and microneme organelles. CONCLUSIONS The present study provides novel information on both the transcriptional activity of E. tenella during schizogony in ceacal tissue and of the local host responses to parasite invasion during this phase of infection. Results indicate a role for IFN-γ and IFN-stimulated genes in the innate defence against Eimeria replication.
Collapse
Affiliation(s)
- Arnar K S Sandholt
- Department of Microbiology, National Veterinary Institute, Uppsala, Sweden
| | - Eva Wattrang
- Department of Microbiology, National Veterinary Institute, Uppsala, Sweden.
| | - Tobias Lilja
- Department of Microbiology, National Veterinary Institute, Uppsala, Sweden
| | - Harri Ahola
- Department of Microbiology, National Veterinary Institute, Uppsala, Sweden
| | - Anna Lundén
- Department of Microbiology, National Veterinary Institute, Uppsala, Sweden
| | - Karin Troell
- Department of Microbiology, National Veterinary Institute, Uppsala, Sweden
| | - Staffan G Svärd
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Robert Söderlund
- Department of Microbiology, National Veterinary Institute, Uppsala, Sweden
| |
Collapse
|
30
|
Gomez MA, Belew AT, Navas A, Rosales-Chilama M, Murillo J, Dillon LAL, Alexander TA, Martinez-Valencia A, El-Sayed NM. Early Leukocyte Responses in Ex-Vivo Models of Healing and Non-Healing Human Leishmania (Viannia) panamensis Infections. Front Cell Infect Microbiol 2021; 11:687607. [PMID: 34557423 PMCID: PMC8453012 DOI: 10.3389/fcimb.2021.687607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/16/2021] [Indexed: 11/21/2022] Open
Abstract
Early host-pathogen interactions drive the host response and shape the outcome of natural infections caused by intracellular microorganisms. These interactions involve a number of immune and non-immune cells and tissues, along with an assortment of host and pathogen-derived molecules. Our current knowledge has been predominantly derived from research on the relationships between the pathogens and the invaded host cell(s), limiting our understanding of how microbes elicit and modulate immunological responses at the organismal level. In this study, we explored the early host determinants of healing and non-healing responses in human cutaneous leishmaniasis (CL) caused by Leishmania (Viannia) panamensis. We performed a comparative transcriptomic profiling of peripheral blood mononuclear cells from healthy donors (PBMCs, n=3) exposed to promastigotes isolated from patients with chronic (CHR, n=3) or self-healing (SH, n=3) CL, and compared these to human macrophage responses. Transcriptomes of L. V. panamensis-infected PBMCs showed enrichment of functional gene categories derived from innate as well as adaptive immune cells signatures, demonstrating that Leishmania modulates adaptive immune cell functions as early as after 24h post interaction with PBMCs from previously unexposed healthy individuals. Among differentially expressed PBMC genes, four broad categories were commonly modulated by SH and CHR strains: cell cycle/proliferation/differentiation, metabolism of macromolecules, immune signaling and vesicle trafficking/transport; the first two were predominantly downregulated, and the latter upregulated in SH and CHR as compared to uninfected samples. Type I IFN signaling genes were uniquely up-regulated in PBMCs infected with CHR strains, while genes involved in the immunological synapse were uniquely downregulated in SH infections. Similarly, pro-inflammatory response genes were upregulated in isolated macrophages infected with CHR strains. Our data demonstrate that early responses during Leishmania infection extend beyond innate cell and/or phagocytic host cell functions, opening new frontiers in our understanding of the triggers and drivers of human CL.
Collapse
Affiliation(s)
- Maria Adelaida Gomez
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad IcesiI, Cali, Colombia
| | - Ashton Trey Belew
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| | - Adriana Navas
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| | - Mariana Rosales-Chilama
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad IcesiI, Cali, Colombia
| | - Julieth Murillo
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Pontificia Universidad Javeriana, Cali, Colombia
| | - Laura A. L. Dillon
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| | - Theresa A. Alexander
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| | | | - Najib M. El-Sayed
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| |
Collapse
|
31
|
Salloum T, Tokajian S, Hirt RP. Advances in Understanding Leishmania Pathobiology: What Does RNA-Seq Tell Us? Front Cell Dev Biol 2021; 9:702240. [PMID: 34540827 PMCID: PMC8440825 DOI: 10.3389/fcell.2021.702240] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/30/2021] [Indexed: 11/23/2022] Open
Abstract
Leishmaniasis is a vector-borne disease caused by a protozoa parasite from over 20 Leishmania species. The clinical manifestations and the outcome of the disease vary greatly. Global RNA sequencing (RNA-Seq) analyses emerged as a powerful technique to profile the changes in the transcriptome that occur in the Leishmania parasites and their infected host cells as the parasites progresses through their life cycle. Following the bite of a sandfly vector, Leishmania are transmitted to a mammalian host where neutrophils and macrophages are key cells mediating the interactions with the parasites and result in either the elimination the infection or contributing to its proliferation. This review focuses on RNA-Seq based transcriptomics analyses and summarizes the main findings derived from this technology. In doing so, we will highlight caveats in our understanding of the parasite's pathobiology and suggest novel directions for research, including integrating more recent data highlighting the role of the bacterial members of the sandfly gut microbiota and the mammalian host skin microbiota in their potential role in influencing the quantitative and qualitative aspects of leishmaniasis pathology.
Collapse
Affiliation(s)
- Tamara Salloum
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos, Lebanon
| | - Sima Tokajian
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos, Lebanon
| | - Robert P. Hirt
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
32
|
Palacios G, Diaz-Solano R, Valladares B, Dorta-Guerra R, Carmelo E. Early Transcriptional Liver Signatures in Experimental Visceral Leishmaniasis. Int J Mol Sci 2021; 22:7161. [PMID: 34281214 PMCID: PMC8267970 DOI: 10.3390/ijms22137161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 01/01/2023] Open
Abstract
Transcriptional analysis of complex biological scenarios has been used extensively, even though sometimes the results of such analysis may prove imprecise or difficult to interpret due to an overwhelming amount of information. In this study, a large-scale real-time qPCR experiment was coupled to multivariate statistical analysis in order to describe the main immunological events underlying the early L. infantum infection in livers of BALB/c mice. High-throughput qPCR was used to evaluate the expression of 223 genes related to immunological response and metabolism 1, 3, 5, and 10 days post infection. This integrative analysis showed strikingly different gene signatures at 1 and 10 days post infection, revealing the progression of infection in the experimental model based on the upregulation of particular immunological response patterns and mediators. The gene signature 1 day post infection was not only characterized by the upregulation of mediators involved in interferon signaling and cell chemotaxis, but also the upregulation of some inhibitory markers. In contrast, at 10 days post infection, the upregulation of many inflammatory and Th1 markers characterized a more defined gene signature with the upregulation of mediators in the IL-12 signaling pathway. Our results reveal a significant connection between the expression of innate immune response and metabolic and inhibitory markers in early L. infantum infection of the liver.
Collapse
Affiliation(s)
- Génesis Palacios
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUESTPC), Universidad de la Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38200 La Laguna (Tenerife), Spain; (G.P.); (R.D.-S.); (B.V.); (R.D.-G.)
| | - Raquel Diaz-Solano
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUESTPC), Universidad de la Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38200 La Laguna (Tenerife), Spain; (G.P.); (R.D.-S.); (B.V.); (R.D.-G.)
| | - Basilio Valladares
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUESTPC), Universidad de la Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38200 La Laguna (Tenerife), Spain; (G.P.); (R.D.-S.); (B.V.); (R.D.-G.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Avda. Astrofísico F. Sánchez s/n, 38200 La Laguna (Tenerife), Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET)
| | - Roberto Dorta-Guerra
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUESTPC), Universidad de la Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38200 La Laguna (Tenerife), Spain; (G.P.); (R.D.-S.); (B.V.); (R.D.-G.)
- Departamento de Matemáticas, Estadística e Investigación Operativa, Facultad de Ciencias, Universidad de La Laguna, 38200 La Laguna (Tenerife), Spain
| | - Emma Carmelo
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUESTPC), Universidad de la Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38200 La Laguna (Tenerife), Spain; (G.P.); (R.D.-S.); (B.V.); (R.D.-G.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Avda. Astrofísico F. Sánchez s/n, 38200 La Laguna (Tenerife), Spain
- Departamento de Matemáticas, Estadística e Investigación Operativa, Facultad de Ciencias, Universidad de La Laguna, 38200 La Laguna (Tenerife), Spain
| |
Collapse
|
33
|
Gil-Jaramillo N, Rocha AP, Raiol T, Motta FN, Favali C, Brigido MM, Bastos IMD, Santana JM. The First Contact of Human Dendritic Cells With Trypanosoma cruzi Reveals Response to Virus as an Unexplored Central Pathway. Front Immunol 2021; 12:638020. [PMID: 33897690 PMCID: PMC8062726 DOI: 10.3389/fimmu.2021.638020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/11/2021] [Indexed: 12/21/2022] Open
Abstract
Chagas disease is a debilitating and neglected disease caused by the protozoan Trypanosoma cruzi. Soon after infection, interactions among T. cruzi and host innate immunity cells can drive/contribute to disease outcome. Dendritic cells (DCs), present in all tissues, are one of the first immune cells to interact with Trypanosoma cruzi metacyclic trypomastigotes. Elucidating the immunological events triggered immediately after parasite-human DCs encounter may aid in understanding the role of DCs in the establishment of infection and in the course of the disease. Therefore, we performed a transcriptomic analysis of a 12 h interaction between T. cruzi and MoDCs (monocyte-derived DCs) from three human donors. Enrichment analyses of the 468 differentially expressed genes (DEGs) revealed viral infection response as the most regulated pathway. Additionally, exogenous antigen processing and presentation through MHC-I, chemokine signaling, lymphocyte co-stimulation, metallothioneins, and inflammasome activation were found up-regulated. Notable, we were able to identify the increased gene expression of alternative inflammasome sensors such as AIM2, IFI16, and RIG-I for the first time in a T. cruzi infection. Both transcript and protein expression levels suggest proinflammatory cytokine production during early T. cruzi-DCs contact. Our transcriptome data unveil antiviral pathways as an unexplored process during T. cruzi-DC initial interaction, disclosing a new panorama for the study of Chagas disease outcomes.
Collapse
Affiliation(s)
- Natalia Gil-Jaramillo
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Amanda Pereira Rocha
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Tainá Raiol
- Fiocruz Brasília–Gerência Regional de Brasília (GEREB), Fundação Oswaldo Cruz (Fiocruz), Brasília, Brazil
| | - Flávia Nader Motta
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
- Faculdade de Ceilândia, Universidade de Brasília, Brasília, Brazil
| | - Cecília Favali
- Laboratório de Imunologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Marcelo M. Brigido
- Laboratório de Imunologia Molecular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Izabela M. D. Bastos
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Jaime M. Santana
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
34
|
Bartholomeu DC, Teixeira SMR, Cruz AK. Genomics and functional genomics in Leishmania and Trypanosoma cruzi: statuses, challenges and perspectives. Mem Inst Oswaldo Cruz 2021; 116:e200634. [PMID: 33787768 PMCID: PMC8011669 DOI: 10.1590/0074-02760200634] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/19/2021] [Indexed: 12/16/2022] Open
Abstract
The availability of Trypanosomatid genomic data in public databases has opened myriad experimental possibilities that have contributed to a more comprehensive understanding of the biology of these parasites and their interactions with hosts. In this review, after brief remarks on the history of the Trypanosoma cruzi and Leishmania genome initiatives, we present an overview of the relevant contributions of genomics, transcriptomics and functional genomics, discussing the primary obstacles, challenges, relevant achievements and future perspectives of these technologies.
Collapse
Affiliation(s)
- Daniella C Bartholomeu
- Universidade Federal de Minas Gerais, Departamento de Parasitologia, Belo Horizonte, MG, Brasil
| | | | - Angela Kaysel Cruz
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Biologia Celular e Molecular, Ribeirão Preto, SP, Brasil
| |
Collapse
|
35
|
Rachidi N, Knippschild U, Späth GF. Dangerous Duplicity: The Dual Functions of Casein Kinase 1 in Parasite Biology and Host Subversion. Front Cell Infect Microbiol 2021; 11:655700. [PMID: 33869086 PMCID: PMC8044801 DOI: 10.3389/fcimb.2021.655700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/05/2021] [Indexed: 02/01/2023] Open
Abstract
Casein Kinase 1 (CK1) family members are serine/threonine protein kinases that are involved in many biological processes and highly conserved in eukaryotes from protozoan to humans. Even though pathogens exploit host CK1 signaling pathways to survive, the role of CK1 in infectious diseases and host/pathogen interaction is less well characterized compared to other diseases, such as cancer or neurodegenerative diseases. Here we present the current knowledge on CK1 in protozoan parasites highlighting their essential role for parasite survival and their importance for host-pathogen interactions. We also discuss how the dual requirement of CK1 family members for parasite biological processes and host subversion could be exploited to identify novel antimicrobial interventions.
Collapse
Affiliation(s)
- Najma Rachidi
- Unité de Parasitologie moléculaire et Signalisation, Department of Parasites and Insect Vectors, Institut Pasteur and INSERM U1201, Paris, France
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Centre, Ulm University Hospital, Ulm, Germany
| | - Gerald F. Späth
- Unité de Parasitologie moléculaire et Signalisation, Department of Parasites and Insect Vectors, Institut Pasteur and INSERM U1201, Paris, France
| |
Collapse
|
36
|
Maran SR, de Lemos Padilha Pitta JL, Dos Santos Vasconcelos CR, McDermott SM, Rezende AM, Silvio Moretti N. Epitranscriptome machinery in Trypanosomatids: New players on the table? Mol Microbiol 2021; 115:942-958. [PMID: 33513291 DOI: 10.1111/mmi.14688] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
Trypanosoma and Leishmania parasites cause devastating tropical diseases resulting in serious global health consequences. These organisms have complex life cycles with mammalian hosts and insect vectors. The parasites must, therefore, survive in different environments, demanding rapid physiological and metabolic changes. These responses depend upon regulation of gene expression, which primarily occurs posttranscriptionally. Altering the composition or conformation of RNA through nucleotide modifications is one posttranscriptional mechanism of regulating RNA fate and function, and modifications including N6-methyladenosine (m6A), N1-methyladenosine (m1A), N5-methylcytidine (m5C), N4-acetylcytidine (ac4C), and pseudouridine (Ψ), dynamically regulate RNA stability and translation in diverse organisms. Little is known about RNA modifications and their machinery in Trypanosomatids, but we hypothesize that they regulate parasite gene expression and are vital for survival. Here, we identified Trypanosomatid homologs for writers of m1A, m5C, ac4C, and Ψ and analyze their evolutionary relationships. We systematically review the evidence for their functions and assess their potential use as therapeutic targets. This work provides new insights into the roles of these proteins in Trypanosomatid parasite biology and treatment of the diseases they cause and illustrates that Trypanosomatids provide an excellent model system to study RNA modifications, their molecular, cellular, and biological consequences, and their regulation and interplay.
Collapse
Affiliation(s)
- Suellen Rodrigues Maran
- Laboratory of Molecular Biology of Pathogens, Department of Microbiology, Immunology and Parasitology, Federal University of Sao Paulo, São Paulo, Brazil
| | | | | | - Suzanne M McDermott
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Nilmar Silvio Moretti
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| |
Collapse
|
37
|
Dual RNA-Seq transcriptome analysis of chicken macrophage-like cells (HD11) infected in vitro with Eimeria tenella. Parasitology 2021; 148:712-725. [PMID: 33536090 PMCID: PMC8056837 DOI: 10.1017/s0031182021000111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The study aimed to monitor parasite and host gene expression during the early stages of Eimeria tenella infection of chicken cells using dual RNA-Seq analysis. For this, we used chicken macrophage-like cell line HD11 cultures infected in vitro with purified E. tenella sporozoites. Cultures were harvested between 2 and 72 h post-infection and mRNA was extracted and sequenced. Dual RNA-Seq analysis showed clear patterns of altered expression for both parasite and host genes during infection. For example, genes in the chicken immune system showed upregulation early (2–4 h), a strong downregulation of genes across the immune system at 24 h and a repetition of early patterns at 72 h, indicating that invasion by a second generation of parasites was occurring. The observed downregulation may be due to immune self-regulation or to immune evasive mechanisms exerted by E. tenella. Results also suggested pathogen recognition receptors involved in E. tenella innate recognition, MRC2, TLR15 and NLRC5 and showed distinct chemokine and cytokine induction patterns. Moreover, the expression of several functional categories of Eimeria genes, such as rhoptry kinase genes and microneme genes, were also examined, showing distinctive differences which were expressed in sporozoites and merozoites.
Collapse
|
38
|
Restrepo CM, Llanes A, Herrera L, Ellis E, Lleonart R, Fernández PL. Gene expression patterns associated with Leishmania panamensis infection in macrophages from BALB/c and C57BL/6 mice. PLoS Negl Trop Dis 2021; 15:e0009225. [PMID: 33617537 PMCID: PMC7932533 DOI: 10.1371/journal.pntd.0009225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 03/04/2021] [Accepted: 02/09/2021] [Indexed: 01/22/2023] Open
Abstract
Leishmania parasites can trigger different host immune responses that result in varying levels of disease severity. The C57BL/6 and BALB/c mouse strains are among the host models commonly used for characterizing the immunopathogenesis of Leishmania species and the possible antileishmanial effect of novel drug candidates. C57BL/6 mice tend to be resistant to Leishmania infections, whereas BALB/c mice display a susceptible phenotype. Studying species-specific interactions between Leishmania parasites and different host systems is a key step to characterize and validate these models for in vivo studies. Here, we use RNA-Seq and differential expression analysis to characterize the transcriptomic profiles of C57BL/6 and BALB/c peritoneal-derived macrophages in response to Leishmania panamensis infection. We observed differences between BALB/c and C57BL/6 macrophages regarding pathways associated with lysosomal degradation, arginine metabolism and the regulation of cell cycle. We also observed differences in the expression of chemokine and cytokine genes associated with regulation of immune responses. In conclusion, infection with L. panamensis induced an inflammatory gene expression pattern in C57BL/6 macrophages that is more consistently associated with a classic macrophage M1 activation, whereas in BALB/c macrophages a gene expression pattern consistent with an intermediate inflammatory response was observed.
Collapse
Affiliation(s)
- Carlos M. Restrepo
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama, Republic of Panama
| | - Alejandro Llanes
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama, Republic of Panama
| | - Lizzi Herrera
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama, Republic of Panama
| | - Esteban Ellis
- Departamento de Biotecnología, Facultad de Ciencias de la Salud, Universidad Latina de Panamá, Panama City, Panama, Republic of Panama
| | - Ricardo Lleonart
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama, Republic of Panama
| | - Patricia L. Fernández
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama, Republic of Panama
| |
Collapse
|
39
|
Dual Host-Intracellular Parasite Transcriptome of Enucleated Cells Hosting Leishmania amazonensis: Control of Half-Life of Host Cell Transcripts by the Parasite. Infect Immun 2020; 88:IAI.00261-20. [PMID: 32817329 DOI: 10.1128/iai.00261-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
Enucleated cells or cytoplasts (cells whose nucleus is removed in vitro) represent an unexplored biological model for intracellular infection studies due to the abrupt interruption of nuclear processing and new RNA synthesis by the host cell in response to pathogen entry. Using enucleated fibroblasts hosting the protozoan parasite Leishmania amazonensis, we demonstrate that parasite multiplication and biogenesis of large parasitophorous vacuoles in which parasites multiply are independent of the host cell nucleus. Dual RNA sequencing of both host cytoplast and intracellular parasite transcripts identified host transcripts that are more preserved or degraded upon interaction with parasites and also parasite genes that are differentially expressed when hosted by nucleated or enucleated cells. Cytoplasts are suitable host cells, which persist in culture for more than 72 h and display functional enrichment of transcripts related to mitochondrial functions and mRNA translation. Crosstalk between nucleated host de novo gene expression in response to intracellular parasitism and the parasite gene expression to counteract or benefit from these host responses induces a parasite transcriptional profile favoring parasite multiplication and aerobic respiration, and a host-parasite transcriptional landscape enriched in host cell metabolic functions related to NAD, fatty acid, and glycolytic metabolism. Conversely, interruption of host nucleus-parasite cross talk by infection of enucleated cells generates a host-parasite transcriptional landscape in which cytoplast transcripts are enriched in phagolysosome-related pathway, prosurvival, and SerpinB-mediated immunomodulation. In addition, predictive in silico analyses indicated that parasite transcript products secreted within cytoplasts interact with host transcript products conserving the host V-ATPase proton translocation function and glutamine/proline metabolism. The collective evidence indicates parasite-mediated control of host cell transcripts half-life that is beneficial to parasite intracellular multiplication and escape from host immune responses. These findings will contribute to improved drug targeting and serve as database for L. amazonensis-host cell interactions.
Collapse
|
40
|
Masoudzadeh N, Östensson M, Persson J, Mashayekhi Goyonlo V, Agbajogu C, Taslimi Y, Erfanian Salim R, Zahedifard F, Mizbani A, Malekafzali Ardekani H, Gunn BM, Rafati S, Harandi AM. Molecular signatures of anthroponotic cutaneous leishmaniasis in the lesions of patients infected with Leishmania tropica. Sci Rep 2020; 10:16198. [PMID: 33004861 PMCID: PMC7529897 DOI: 10.1038/s41598-020-72671-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Anthroponotic cutaneous leishmaniasis (CL) caused by Leishmania tropica (L. tropica) represents a public health challenge in several resource poor settings. We herein employed a systems analysis approach to study molecular signatures of CL caused by L. tropica in the skin lesions of ulcerative CL (UCL) and non-ulcerative CL (NUCL) patients. Results from RNA-seq analysis determined shared and unique functional transcriptional pathways in the lesions of the UCL and NUCL patients. Several transcriptional pathways involved in inflammatory response were positively enriched in the CL lesions. A multiplexed inflammatory protein analysis showed differential profiles of inflammatory cytokines and chemokines in the UCL and NUCL lesions. Transcriptional pathways for Fcγ receptor dependent phagocytosis were among shared enriched pathways. Using L. tropica specific antibody (Ab)-mediated phagocytosis assays, we could substantiate Ab-dependent cellular phagocytosis (ADCP) and Ab-dependent neutrophil phagocytosis (ADNP) activities in the lesions of the UCL and NUCL patients, which correlated with L. tropica specific IgG Abs. Interestingly, a negative correlation was observed between parasite load and L. tropica specific IgG/ADCP/ADNP in the skin lesions of CL patients. These results enhance our understanding of human skin response to CL caused by L. tropica.
Collapse
Affiliation(s)
- Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Malin Östensson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Josefine Persson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Christopher Agbajogu
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | | | - Farnaz Zahedifard
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Bronwyn M Gunn
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA, 02139, USA
- Paul G. Allen School of Global Animal Health, Washington State University, Pullman, WA, 99164, USA
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran.
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, Canada.
| |
Collapse
|
41
|
Masoudzadeh N, Mizbani A, Rafati S. Transcriptomic profiling in Cutaneous Leishmaniasis patients. Expert Rev Proteomics 2020; 17:533-541. [PMID: 32886890 DOI: 10.1080/14789450.2020.1812390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cutaneous leishmaniasis (CL), caused by different Leishmania parasite species, is associated with parasite-induced immune-mediated skin inflammation and ulceration. Whereas many CL studies focus on gene expression signatures in mouse models, the transcriptional response driving human patients in the field is less characterized. Human studies in CL disease provide the opportunity to directly investigate the host-pathogen interaction in the cutaneous lesion site. AREAS COVERED Advances in high-throughput sequencing technologies, particularly their application for evaluation of the global gene expression changes, have made transcriptomics as a powerful tool to understand the pathogen-host molecular interactions. EXPERT COMMENTARY In this review, we focus on the transcriptomics studies that have been performed so far on human blood or tissue-driven samples to investigate Leishmania parasites interplay with the CL patients. Further, we summarize microarray and RNA-seq studies associated with lesion biopsies of CL patients to discuss how current whole genome analysis along with systems biology approaches have developed novel CL biomarkers for further applications, not only for research, but also for accelerating vaccine development.
Collapse
Affiliation(s)
- Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran , Tehran, Iran
| | - Amir Mizbani
- Department of Health Sciences and Technology, ETH Zurich , Switzerland
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran , Tehran, Iran
| |
Collapse
|
42
|
Aoki JI, Muxel SM, Laranjeira-Silva MF, Zampieri RA, Müller KE, Nerland AH, Floeter-Winter LM. Dual transcriptome analysis reveals differential gene expression modulation influenced by Leishmania arginase and host genetic background. Microb Genom 2020; 6:mgen000427. [PMID: 32886592 PMCID: PMC7643972 DOI: 10.1099/mgen.0.000427] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/13/2020] [Indexed: 01/04/2023] Open
Abstract
The outcome of Leishmania infection is strongly influenced by the host's genetic background. BALB/c mice are susceptible to Leishmania infection, while C57BL/6 mice show discrete resistance. Central to the fate of the infection is the availability of l-arginine and the related metabolic processes in the host and parasite. Depending on l-arginine availability, nitric oxide synthase 2 (NOS2) of the host cell produces nitric oxide (NO) controlling the parasite growth. On the other hand, Leishmania can also use host l-arginine for the production of polyamines through its own arginase activity, thus favouring parasite replication. Considering RNA-seq data, we analysed the dual modulation of host and parasite gene expression of BALB/c or C57BL/6 mouse bone marrow-derived macrophages (BMDMs) after 4 h of infection with Leishmania amazonensis wild-type (La-WT) or L. amazonensis arginase knockout (La-arg-). We identified 12 641 host transcripts and 8282 parasite transcripts by alignment analysis with the respective Mus musculus and L. mexicana genomes. The comparison of BALB/c_La-arg-versus BALB/c_La-WT revealed 233 modulated transcripts, with most related to the immune response and some related to the amino acid transporters and l-arginine metabolism. In contrast, the comparison of C57BL/6_La-arg-vs. C57BL/6_La-WT revealed only 30 modulated transcripts, including some related to the immune response but none related to amino acid transport or l-arginine metabolism. The transcriptome profiles of the intracellular amastigote revealed 94 modulated transcripts in the comparison of La-arg-_BALB/c vs. La-WT_BALB/c and 45 modulated transcripts in the comparison of La-arg-_C57BL/6 vs. La-WT_C57BL/6. Taken together, our data present new insights into the impact of parasite arginase activity on the orchestration of the host gene expression modulation, including in the immune response and amino acid transport and metabolism, mainly in susceptible BALB/c-infected macrophages. Moreover, we show how parasite arginase activity affects parasite gene expression modulation, including amino acid uptake and amastin expression.
Collapse
Affiliation(s)
- Juliana Ide Aoki
- Department of Physiology, Institute of Bioscience, University of São Paulo, São Paulo, Brazil
| | - Sandra Marcia Muxel
- Department of Physiology, Institute of Bioscience, University of São Paulo, São Paulo, Brazil
| | | | | | - Karl Erik Müller
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Drammen Hospital, Drammen, Norway
| | | | | |
Collapse
|
43
|
Pessenda G, da Silva JS. Arginase and its mechanisms in Leishmania persistence. Parasite Immunol 2020; 42:e12722. [PMID: 32294247 DOI: 10.1111/pim.12722] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/14/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Leishmaniasis is a neglected infectious disease with clinical presentations ranging from asymptomatic or mild symptoms to chronic infection and eventual death. The mechanisms of disease susceptibility and pathology have been extensively studied, but there are no steadfast rules regarding leishmaniasis. A Th1 response is usually associated with infection control, while a predominant Th2 response is detrimental to the patient. In this scenario, the enzymes arginase and inducible nitric oxide synthase represent two possible pathways of immune response. While the former contributes to parasite replication, the latter is crucial for its control. In the present review, we collected study results that associate arginase expression in patients and in experimental models with disease susceptibility/chronicity and show some proposed mechanisms that explain the role of arginase in maintaining Leishmania infection, including polyamine and thiol synthesis, tissue-resident macrophage (TRM) proliferation and activation and T-cell suppression and exhaustion.
Collapse
Affiliation(s)
- Gabriela Pessenda
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Santana da Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Fundação Oswaldo Cruz Bi-institucional, Ribeirão Preto, Brazil
| |
Collapse
|
44
|
Ulusan Ö, Mert U, Sadıqova A, Öztürk S, Caner A. Identification of gene expression profiles in Leishmania major infection by integrated bioinformatics analyses. Acta Trop 2020; 208:105517. [PMID: 32360239 DOI: 10.1016/j.actatropica.2020.105517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 10/24/2022]
Abstract
Gene expression profiling in mouse models of leishmaniasis has given useful information to understand the molecular pathways active in lesions and to discover new diagnostic/therapeutic targets. Although the host response plays a critical role in protection from leishmaniasis and promoting disease severity, there are still unexplained aspects in the mechanism of non-healing cutaneous lesions, which need biomarkers for both targeted- therapy and diagnosis. To address this, transcriptional profiling of the skin lesions obtained from BALB/c mice infected with Leishmania major and healthy skin from naïve mice were evaluated by bioinformatics analysis, and then the results were validated by Revers Transcriptase-PCR. Five genes among the up-regulated differentially expressed genes named FCGR4, CCL4, CXCL9, Arg1 and IL-1β were found to have relatively high diagnostic value for CL due to L. major. Pathway analysis revealed that Triggering Receptor Expressed On Myeloid Cells 1 (TREM1) signaling pathways are active in cutaneous lesions, providing new insights for the understanding and treatment of leishmaniasis.
Collapse
Affiliation(s)
- Özlem Ulusan
- Department of Parasitology, Ege University Medical School, Izmir, Turkey
| | - Ufuk Mert
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Aygül Sadıqova
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Sercan Öztürk
- Departments of Computer Science, Dokuz Eylül University, Izmir, Turkey
| | - Ayse Caner
- Department of Parasitology, Ege University Medical School, Izmir, Turkey; Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey; Department of Bioinformatics, Ege University, Institute of Health Sciences, Izmir, Turkey; Departments of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
45
|
Phylloseptin-1 is Leishmanicidal for Amastigotes of Leishmania amazonensis Inside Infected Macrophages. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17134856. [PMID: 32640562 PMCID: PMC7370015 DOI: 10.3390/ijerph17134856] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/25/2020] [Accepted: 07/01/2020] [Indexed: 12/26/2022]
Abstract
Leishmania protozoans are the causal agents of neglected diseases that represent an important public health issue worldwide. The growing occurrence of drug-resistant strains of Leishmania and severe side effects of available treatments represent an important challenge for the leishmaniases treatment. We have previously reported the leishmanicidal activity of phylloseptin-1 (PSN-1), a peptide found in the skin secretion of Phyllomedusa azurea (=Pithecopus azureus), against Leishmania amazonensis promastigotes. However, its impact on the amastigote form of L. amazonensis and its impact on infected macrophages are unknown. In this work, we evaluated the effects of PSN-1 on amastigotes of L. amazonensis inside macrophages infected in vitro. We assessed the production of hydrogen peroxide and nitric oxide, as well as the levels of inflammatory and immunomodulatory markers (TGF-β, TNF-α and IL-12), in infected and non-infected macrophages treated with PSN-1. Treatment with PSN-1 decreased the number of infected cells and the number of ingested amastigotes per cell when compared with the untreated cells. At 32 µM (64 µg/mL), PSN-1 reduced hydrogen peroxide levels in both infected and uninfected macrophages, whereas it had little effect on NO production or TGF-β release. The effect of PSN-1 on IL-12 and TNF-α secretion depended on its concentration, but, in general, their levels tended to increase as PSN-1 concentration increased. Further in vitro and in vivo studies are needed to clarify the mechanisms of action of PSN-1 and its interaction with the immune system aiming to develop pharmacological applications.
Collapse
|
46
|
Jha MK, Sarode AY, Bodhale N, Mukherjee D, Pandey SP, Srivastava N, Rub A, Silvestre R, Sarkar A, Saha B. Development and Characterization of an Avirulent Leishmania major Strain. THE JOURNAL OF IMMUNOLOGY 2020; 204:2734-2753. [PMID: 32245818 DOI: 10.4049/jimmunol.1901362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/05/2020] [Indexed: 01/12/2023]
Abstract
Leishmania major causes cutaneous leishmaniasis. An antileishmanial vaccine for humans is unavailable. In this study, we report development of two attenuated L. major strains-5ASKH-HP and LV39-HP-by continuous culture (high passage) of the corresponding virulent strains (low passage). Both avirulent strains showed similar changes in proteome profiles when analyzed by surface-enhanced laser desorption ionization mass spectrometry. Liquid chromatography-mass spectrometry and microarray characterization of 5ASKH strains revealed substantially altered gene and protein expression profiles, respectively. Both virulent and avirulent L. major strains grew comparably in culture, but the avirulent strain survived significantly less in BALB/c-derived peritoneal macrophages. Both attenuated strains failed to infect BALB/c mice and elicited IFN-γ, but not IL-4 and IL-10, responses. 5ASKH-HP parasites failed to induce significant infection even in severely immunocompromised- SCID or inducible NO synthase-, CD40-, or IL-12-deficient mice, indicating attenuation. The avirulent strain induced less IL-10, but higher IL-12, in macrophages. The avirulent strain failed to reduce CD40 relocation to the detergent-resistant membrane domain and to inhibit CD40-induced phosphorylation of the kinases Lyn and protein kinase C-β and MAPKs MKK-3/6 and p38MAPK or to upregulate MEK-1/2 and ERK-1/2 in BALB/c-derived peritoneal macrophages. The virulent and the avirulent strains reciprocally modulated CD40-induced Ras-mediated signaling through PI-3K and Raf-1. Avirulent 5ASKH-primed BALB/c mice were protected against virulent L. major challenge infection. The loss of virulence accompanied by substantially altered proteome profiles and the elicitation of host-protective immune responses indicate plausibly irreversible attenuation of the L. major strain and its potential use as a vaccine strain.
Collapse
Affiliation(s)
- Mukesh Kumar Jha
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Aditya Y Sarode
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Neelam Bodhale
- Jagadis Bose National Science Talent Search, Kolkata, West Bengal 700107, India
| | - Debasri Mukherjee
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Surya Prakash Pandey
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Neetu Srivastava
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Abdur Rub
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal; and
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, Odisha 751024, India
| | - Bhaskar Saha
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India; .,Trident Academy of Creative Technology, Bhubaneswar, Odisha 751024, India
| |
Collapse
|
47
|
Coutinho-Abreu IV, Serafim TD, Meneses C, Kamhawi S, Oliveira F, Valenzuela JG. Distinct gene expression patterns in vector-residing Leishmania infantum identify parasite stage-enriched markers. PLoS Negl Trop Dis 2020; 14:e0008014. [PMID: 32126078 PMCID: PMC7053709 DOI: 10.1371/journal.pntd.0008014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/24/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Leishmaniasis is a vector-borne neglected disease. Inside the natural sand fly vector, the promastigote forms of Leishmania undergo a series of extracellular developmental stages to reach the infectious stage, the metacyclic promastigote. There is limited information regarding the expression profile of L. infantum developmental stages inside the sand fly vector, and molecular markers that can distinguish the different parasite stages are lacking. METHODOLOGY/PRINCIPAL FINDINGS We performed RNAseq on unaltered midguts of the sand fly Lutzomyia longipalpis after infection with L. infantum parasites. RNAseq was carried out at various time points throughout parasite development. Principal component analysis separated the transcripts corresponding to the different Leishmania promastigote stages, the procyclic, nectomonad, leptomonad and metacyclics. Importantly, there were a significant number of differentially expressed genes when comparing the sequential development of the various Leishmania stages in the sand fly. There were 836 differentially expressed (DE) genes between procyclic and long nectomonad promastigotes; 113 DE genes between nectomonad and leptomonad promastigotes; and 302 DE genes between leptomonad and metacyclic promastigotes. Most of the DE genes do not overlap across stages, highlighting the uniqueness of each Leishmania stage. Furthermore, the different stages of Leishmania parasites exhibited specific transcriptional enrichment across chromosomes. Using the transcriptional signatures exhibited by distinct Leishmania stages during their development in the sand fly midgut, we determined the genes predominantly enriched in each stage, identifying multiple potential stage-specific markers for L. infantum. CONCLUSIONS Overall, these findings demonstrate the transcriptional plasticity of the Leishmania parasite inside the sand fly vector and provide a repertoire of potential stage-specific markers for further development as molecular tools for epidemiological studies.
Collapse
Affiliation(s)
- Iliano V. Coutinho-Abreu
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Tiago D. Serafim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| |
Collapse
|
48
|
Çakır T, Panagiotou G, Uddin R, Durmuş S. Novel Approaches for Systems Biology of Metabolism-Oriented Pathogen-Human Interactions: A Mini-Review. Front Cell Infect Microbiol 2020; 10:52. [PMID: 32117818 PMCID: PMC7031156 DOI: 10.3389/fcimb.2020.00052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/27/2020] [Indexed: 12/23/2022] Open
Abstract
Pathogenic microorganisms exploit host metabolism for sustained survival by rewiring its metabolic interactions. Therefore, several metabolic changes are induced in both pathogen and host cells in the course of infection. A systems-based approach to elucidate those changes includes the integrative use of genome-scale metabolic networks and molecular omics data, with the overall goal of better characterizing infection mechanisms for novel treatment strategies. This review focuses on novel aspects of metabolism-oriented systems-based investigation of pathogen-human interactions. The reviewed approaches are the generation of dual-omics data for the characterization of metabolic signatures of pathogen-host interactions, the reconstruction of pathogen-host integrated genome-scale metabolic networks, which has a high potential to be applied to pathogen-gut microbiota interactions, and the structure-based analysis of enzymes playing role in those interactions. The integrative use of those approaches will pave the way for the identification of novel biomarkers and drug targets for the prediction and prevention of infectious diseases.
Collapse
Affiliation(s)
- Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey
| | - Gianni Panagiotou
- Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoll Institute, Jena, Germany
| | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Saliha Durmuş
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey
| |
Collapse
|
49
|
Azevedo LG, de Queiroz ATL, Barral A, Santos LA, Ramos PIP. Proteins involved in the biosynthesis of lipophosphoglycan in Leishmania: a comparative genomic and evolutionary analysis. Parasit Vectors 2020; 13:44. [PMID: 32000835 PMCID: PMC6993435 DOI: 10.1186/s13071-020-3914-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/24/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Leishmania spp. are digenetic parasites capable of infecting humans and causing a range of diseases collectively known as leishmaniasis. The main mechanisms involved in the development and permanence of this pathology are linked to evasion of the immune response. Crosstalk between the immune system and particularities of each pathogenic species is associated with diverse disease manifestations. Lipophosphoglycan (LPG), one of the most important molecules present on the surface of Leishmania parasites, is divided into four regions with high molecular variability. Although LPG plays an important role in host-pathogen and vector-parasite interactions, the distribution and phylogenetic relatedness of the genes responsible for its synthesis remain poorly explored. The recent availability of full genomes and transcriptomes of Leishmania parasites offers an opportunity to leverage insight on how LPG-related genes are distributed and expressed by these pathogens. RESULTS Using a phylogenomics-based framework, we identified a catalog of genes involved in LPG biosynthesis across 22 species of Leishmania from the subgenera Viannia and Leishmania, as well as 5 non-Leishmania trypanosomatids. The evolutionary relationships of these genes across species were also evaluated. Nine genes related to the production of the glycosylphosphatidylinositol (GPI)-anchor were highly conserved among compared species, whereas 22 genes related to the synthesis of the repeat unit presented variable conservation. Extensive gain/loss events were verified, particularly in genes SCG1-4 and SCA1-2. These genes act, respectively, on the synthesis of the side chain attached to phosphoglycans and in the transfer of arabinose residues. Phylogenetic analyses disclosed evolutionary patterns reflective of differences in host specialization, geographic origin and disease manifestation. CONCLUSIONS The multiple gene gain/loss events identified by genomic data mining help to explain some of the observed intra- and interspecies variation in LPG structure. Collectively, our results provide a comprehensive catalog that details how LPG-related genes evolved in the Leishmania parasite specialization process.
Collapse
Affiliation(s)
- Lucas Gentil Azevedo
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia Brazil
- Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia Brazil
| | - Artur Trancoso Lopo de Queiroz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia Brazil
| | - Aldina Barral
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Universidade Federal da Bahia, Salvador, Bahia Brazil
- Instituto de Investigação em Imunologia (iii-INCT), São Paulo, São Paulo Brazil
| | - Luciane Amorim Santos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia Brazil
| | - Pablo Ivan Pereira Ramos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia Brazil
| |
Collapse
|
50
|
Gatto M, Borim PA, Wolf IR, Fukuta da Cruz T, Ferreira Mota GA, Marques Braz AM, Casella Amorim B, Targino Valente G, de Assis Golim M, Venturini J, Araújo Junior JP, Pontillo A, Sartori A. Transcriptional analysis of THP-1 cells infected with Leishmania infantum indicates no activation of the inflammasome platform. PLoS Negl Trop Dis 2020; 14:e0007949. [PMID: 31961876 PMCID: PMC6994165 DOI: 10.1371/journal.pntd.0007949] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 01/31/2020] [Accepted: 11/25/2019] [Indexed: 12/31/2022] Open
Abstract
Leishmaniasis is caused by intracellular parasites transmitted to vertebrates by sandfly bites. Clinical manifestations include cutaneous, mucosal or visceral involvement depending upon the host immune response and the parasite species. To assure their survival inside macrophages, these parasites developed a plethora of highly successful strategies to manipulate various immune system pathways. Considering that inflammasome activation is critical for the establishment of a protective immune response in many parasite infections, in this study we determined the transcriptome of THP-1 cells after infection with L. infantum, with a particular focus on the inflammasome components. To this end, the human cell line THP-1, previously differentiated into macrophages by PMA treatment, was infected with L. infantum promastigotes. Differentiated THP-1 cells were also stimulated with LPS to be used as a comparative parameter. The gene expression signature was determined 8 hours after by RNA-seq technique. Infected or uninfected THP-1 cells were stimulated with nigericin (NIG) to measure active caspase-1 and TNF-α, IL-6 and IL-1β levels in culture supernatants after 8, 24 and 48 hours. L. infantum triggered a gene expression pattern more similar to non-infected THP-1 cells and very distinct from LPS-stimulated cells. Some of the most up-regulated genes in L. infantum-infected cells were CDC20, CSF1, RPS6KA1, CD36, DUSP2, DUSP5, DUSP7 and TNFAIP3. Some up-regulated GO terms in infected cells included cell coagulation, regulation of MAPK cascade, response to peptide hormone stimulus, negative regulation of transcription from RNA polymerase II promoter and nerve growth factor receptor signaling pathway. Infection was not able to induce the expression of genes associated with the inflammasome signaling pathway. This finding was confirmed by the absence of caspase-1 activation and IL-1β production after 8, 24 and 48 hours of infection. Our results indicate that L. infantum was unable to activate the inflammasomes during the initial interaction with THP-1 cells. Visceral leishmaniasis, caused by Leishmania infantum, is a disease that affects millions of people worldwide. The entry of microorganisms into the host is commonly associated with activation of a multiprotein platform called inflammasome whose assembly culminates in caspase-1 activation and IL-1β production. ILβ activates other cells and effector mechanisms leading to clearance of pathogens. However, the involvement of inflammasomes in the human infection with L. infantum is poorly known. To investigate the parasite-host interaction is fundamental to understand the immunopathogenesis of visceral leishmaniasis and to allow the development of new therapeutic strategies. In this study, we used RNA-seq, a tool that allowed to investigate the global gene expression of THP-1 cells, which is a macrophage-like human cell line, infected with L. infantum. By using computational analysis, this approach allowed us to evaluate the expression of genes that compose the inflammasomes pathway and other gene networks and signaling pathways triggered after infection. This analysis indicated that, unlike species causing cutaneous leishmaniasis, L. infantum did not induce the expression of genes of inflammasome pathways, nor caspase-1 activation or IL-1β production, possibly reflecting a parasite strategy to manipulate immune system and therefore, to allow its survival inside the cells.
Collapse
Affiliation(s)
- Mariana Gatto
- Tropical Diseases Department, Botucatu Medical School – UNESP, Botucatu, Brazil
- * E-mail:
| | | | - Ivan Rodrigo Wolf
- Bioprocess and Biotechnology Department, Agronomic Sciences School – UNESP, Botucatu, Brazil
| | - Taís Fukuta da Cruz
- Microbiology and Immunology Department, Biosciences Institute - UNESP, Botucatu, Brazil
| | | | | | | | | | | | | | | | | | - Alexandrina Sartori
- Tropical Diseases Department, Botucatu Medical School – UNESP, Botucatu, Brazil
| |
Collapse
|