1
|
Bruno F, Castelli G, Li B, Reale S, Carra E, Vitale F, Scibetta S, Calzolari M, Varani S, Ortalli M, Franceschini E, Gennari W, Rugna G, Späth GF. Genomic and epidemiological evidence for the emergence of a L. infantum/L. donovani hybrid with unusual epidemiology in northern Italy. mBio 2024; 15:e0099524. [PMID: 38832792 PMCID: PMC11253594 DOI: 10.1128/mbio.00995-24] [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: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 06/05/2024] Open
Abstract
Leishmania (L.) infantum is one of the main causative agents of animal and human leishmaniasis across many endemic areas in South America, Europe, North Africa, and Asia. Despite its clinical significance, little is known about the genetic diversity of L. infantum circulating in a given endemic area. Here, we investigate this important open question by applying a comparative genomics approach to seven L. infantum isolates from different hosts and Italian regions, including the northern part of the country (Emilia-Romagna, RER), Sicily, and Sardinia, as an initial attempt to explore the breadth of parasite genetic heterogeneity in Italy. Additionally, microsatellite analysis was carried out to compare the isolates from RER with other 70 L. infantum strains from the same region as well as 65 strains belonging to the L. donovani complex from other countries. We revealed important karyotypic instability and identified strain-specific changes in gene dosage, which affected important virulence factors such as amastins and surface antigen-like proteins. Single nucleotide polymorphism-based clustering analysis of these genomes together with over 80 publicly available L. infantum and L. donovani genomes placed the Italian isolates into three geographically distinct clusters within the Mediterranean basin and uncovered three isolates clustering with putative L. infantum/L. donovani hybrids isolated in Cyprus. As judged by microsatellite profiling, these hybrid isolates are representative of a sub-population of parasites circulating in northern Italy that preferentially infect humans but not dogs. Our results place Italy at the crossroads of L. infantum infection in the Mediterranean and call attention to the public health risk represented by the introduction of non-European Leishmania species.IMPORTANCEThis study closes important knowledge gaps with respect to Leishmania (L.) infantum genetic heterogeneity in a given endemic country, as exemplified here for Italy, and reveals genetic hybridization as a main cause for re-emerging human leishmaniasis in northern Italy. The observed high diversity of Leishmania parasites on the Italian peninsula suggests different geographical origins, with genomic adaptation to various ecologies affecting both pathogenicity and transmission potential. This is documented by the discovery of a putative L. infantum/L. donovani hybrid strain, which has been shown to preferentially infect humans but not dogs. Our results provide important information to health authorities, which need to consider the public health risk represented by the introduction of new Leishmania species into EU countries due to population displacement or travel from countries where exotic/allochthonous parasite species are endemic.
Collapse
Affiliation(s)
- F. Bruno
- WOAH Leishmania Reference Laboratory, Istituto Zooprofilattico Sperimentale della Sicilia, Centro di Referenza Nazionale per le Leishmaniosi (C.Re.Na.L.), Palermo, Italy
| | - G. Castelli
- WOAH Leishmania Reference Laboratory, Istituto Zooprofilattico Sperimentale della Sicilia, Centro di Referenza Nazionale per le Leishmaniosi (C.Re.Na.L.), Palermo, Italy
| | - B. Li
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - S. Reale
- WOAH Leishmania Reference Laboratory, Istituto Zooprofilattico Sperimentale della Sicilia, Centro di Referenza Nazionale per le Leishmaniosi (C.Re.Na.L.), Palermo, Italy
| | - E. Carra
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "B. Ubertini", Brescia, Italy
| | - F. Vitale
- WOAH Leishmania Reference Laboratory, Istituto Zooprofilattico Sperimentale della Sicilia, Centro di Referenza Nazionale per le Leishmaniosi (C.Re.Na.L.), Palermo, Italy
| | - S. Scibetta
- WOAH Leishmania Reference Laboratory, Istituto Zooprofilattico Sperimentale della Sicilia, Centro di Referenza Nazionale per le Leishmaniosi (C.Re.Na.L.), Palermo, Italy
| | - M. Calzolari
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "B. Ubertini", Brescia, Italy
| | - S. Varani
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - M. Ortalli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - E. Franceschini
- Infectious Disease Unit, Azienda Ospedaliera Universitaria di Modena, Modena, Italy
| | - W. Gennari
- Virology and Molecular Microbiology Unit, University Hospital of Modena, Modena, Italy
| | - G. Rugna
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "B. Ubertini", Brescia, Italy
| | - G. F. Späth
- Unité de Parasitologie moléculaire et Signalisation, INSERM U1201, Institut Pasteur, Université Paris Cité, Paris, France
| |
Collapse
|
2
|
Vijayakumar S, Narayan PK, Kumari S, Ranjan R, Kumar V, Kumar A, Alti D. A review of non-invasive samples and tools in kala-azar diagnosis and test of cure. Exp Parasitol 2024; 259:108713. [PMID: 38350522 DOI: 10.1016/j.exppara.2024.108713] [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: 10/20/2023] [Revised: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
The recurrence of visceral leishmaniasis (VL), also called kala-azar (KA), in endemic regions of tropical countries like India, is primarily attributed to asymptomatic VL, post-kala azar dermal leishmaniasis (PKDL), and human immunodeficiency virus (HIV) co-infection. To effectively manage VL cases and elimination targets, an early and rapid diagnosis as well as accurate field surveillance is highly essential. The traditional sampling methods like bone marrow (BM), spleen, and lymph node (LN) tissue aspirations are invasive, painful, tedious, and prone to nosocomial infections, require skilled persons and hospital facilities, and are not feasible in rural areas. Therefore, there is an urgent requirement for the adoption of a patient-friendly, non-invasive, non-hospitalized sampling procedure that ensures an effective VL diagnosis. This review aims to meticulously evaluate the most recent scientific research that focuses on the precision, feasibility, and applicability of non-invasive sampling (NIS) and techniques for the diagnosis and test of cure of VL, particularly in resource-limited settings. Apart from that, the non-invasive techniques (NIT) that have shown promising results while monitoring VL treatment response and relapse are also reviewed. The limitations associated with NIT and possible improvements in this regard are discussed as well to improve the diagnosis and management of VL.
Collapse
Affiliation(s)
- Saravanan Vijayakumar
- National Centre for Disease Informatics and Research (ICMR-NCDIR), Bengaluru, 562110, India.
| | | | - Shobha Kumari
- ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India, 800007.
| | - Ravi Ranjan
- ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India, 800007.
| | - Vikash Kumar
- ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India, 800007.
| | - Ashish Kumar
- ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India, 800007.
| | - Dayakar Alti
- ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India, 800007.
| |
Collapse
|
3
|
Nemati Haravani T, Parvizi P, Hejazi SH, Sedaghat MM, Eskandarian A, Nateghi Rostami M. Evaluation of expression variations in virulence-related genes of Leishmania major after several culture passages compared with Phlebotomus papatasi isolated promastigotes. PLoS One 2023; 18:e0284240. [PMID: 37053214 PMCID: PMC10101501 DOI: 10.1371/journal.pone.0284240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
Cutaneous leishmaniasis (CL) is a prevalent infectious disease with considerable morbidity annually. Here, we aimed to investigate the likely variations in gene expression of glycoprotein63 (gp63), heat shock protein 70 (HSP70), histone, arginase, cysteine protease B (CPB), Leishmania homologue of receptors for activated C kinase (LACK), small hydrophilic endoplasmic reticulum-associated protein (SHERP) in metacyclic promastigotes of L. major isolated from Phlebotomus papatasi sand flies and promastigotes excessively cultured in culture medium. The parasites were collected from suspected CL cases in Pasteur Institute of Iran, cultured and inoculated into the female BALB/c mice (2×106 promastigotes). Sand flies were trapped in Qom province, fed with the blood of euthanized infected mice and subsequently dissected in order to isolate the midgut including stomodeal valve. The metacyclic promastigotes were isolated from Ph. papatasi (Pro-Ppap) using peanut agglutinin test (PNA), then continuously cultured in RPMI-1640 medium enriched with fetal bovine serum, penicillin (100 U/ml) and streptomycin (100 mg/ml) to reach stationary phase (Pro-Stat). The gene expression was evaluated in both parasitic stages (Pro-Ppap and Pro-Stat) using qRT-PCR. Out results showed a significant increased gene expression at Pro-Ppap stage for gp63 (P = 0.002), SHERP (P = 0.001) and histone (P = 0.026) genes, in comparison with Pro-Stat stage. Noticeably, significant changes were, also, demonstrated in 10th to 15th passages [gp63 (P = 0.041), arginase (P = 0.016), LACK (P = 0.025)] and in 5th to 20th passage (SHERP) (P = 0.029). In conclusion, the findings of the present study seem to be essential in designing Leishmania studies, in particular regarding host-parasite interaction, immunization and infectivity studies.
Collapse
Affiliation(s)
- Taher Nemati Haravani
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parviz Parvizi
- Molecular Systematics Laboratory, Parasitology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Hossein Hejazi
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Mehdi Sedaghat
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbasali Eskandarian
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
4
|
Vithayathil M, Modolell I, Ortiz-Fernandez-Sordo J, Oukrif D, Pappas A, Januszewicz W, O'Donovan M, Hadjinicolaou A, Bianchi M, Blasko A, White J, Kaye P, Novelli M, Wernisch L, Ragunath K, di Pietro M. Image-Enhanced Endoscopy and Molecular Biomarkers Vs Seattle Protocol to Diagnose Dysplasia in Barrett's Esophagus. Clin Gastroenterol Hepatol 2022; 20:2514-2523.e3. [PMID: 35183768 DOI: 10.1016/j.cgh.2022.01.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/21/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Dysplasia in Barrett's esophagus often is invisible on high-resolution white-light endoscopy (HRWLE). We compared the diagnostic accuracy for inconspicuous dysplasia of the combination of autofluorescence imaging (AFI)-guided probe-based confocal laser endomicroscopy (pCLE) and molecular biomarkers vs HRWLE with Seattle protocol biopsies. METHODS Barrett's esophagus patients with no dysplastic lesions were block-randomized to standard endoscopy (HRWLE with the Seattle protocol) or AFI-guided pCLE with targeted biopsies for molecular biomarkers (p53 and cyclin A by immunohistochemistry; aneuploidy by image cytometry), with crossover to the other arm after 6 to 12 weeks. The primary end point was the histologic diagnosis from all study biopsies (trial histology). A sensitivity analysis was performed for overall histology, which included diagnoses within 12 months from the first study endoscopy. Endoscopists were blinded to the referral endoscopy and histology results. The primary outcome was diagnostic accuracy for dysplasia by real-time pCLE vs HRWLE biopsies. RESULTS Of 154 patients recruited, 134 completed both arms. In the primary outcome analysis (trial histology analysis), AFI-guided pCLE had similar sensitivity for dysplasia compared with standard endoscopy (74.3%; 95% CI, 56.7-87.5 vs 80.0%; 95% CI, 63.1-91.6; P = .48). Multivariate logistic regression showed pCLE optical dysplasia, aberrant p53, and aneuploidy had the strongest correlation with dysplasia (secondary outcome). This 3-biomarker panel had higher sensitivity for any grade of dysplasia than the Seattle protocol (81.5% vs 51.9%; P < .001) in the overall histology analysis, but not in the trial histology analysis (91.4% vs 80.0%; P = .16), with an area under the receiver operating curve of 0.83. CONCLUSIONS Seattle protocol biopsies miss dysplasia in approximately half of patients with inconspicuous neoplasia. AFI-guided pCLE has similar accuracy to the current gold standard. The addition of molecular biomarkers could improve diagnostic accuracy.
Collapse
Affiliation(s)
- Mathew Vithayathil
- Medical Research Council Cancer Unit, University of Cambridge, United Kingdom
| | - Ines Modolell
- Department of Gastroenterology, University Hospital National Health Service Foundation Trust, United Kingdom
| | - Jacobo Ortiz-Fernandez-Sordo
- Nottingham Digestive Diseases Centre, National Institute of Health Research Nottingham Biomedical Research Centre, United Kingdom
| | - Dahmane Oukrif
- Department of Histopathology, University College London Hospital, Longdon, United Kingdom
| | - Apostolos Pappas
- Medical Research Council Cancer Unit, University of Cambridge, United Kingdom
| | - Wladyslaw Januszewicz
- Medical Research Council Cancer Unit, University of Cambridge, United Kingdom; Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Centre for Postgraduate Education, Warsaw, Poland
| | - Maria O'Donovan
- Department of Histopathology, Cambridge University Hospital National Health Service Foundation Trust, United Kingdom
| | - Andreas Hadjinicolaou
- Medical Research Council Cancer Unit, University of Cambridge, United Kingdom; Department of Gastroenterology, University Hospital National Health Service Foundation Trust, United Kingdom
| | - Michele Bianchi
- Medical Research Council Cancer Unit, University of Cambridge, United Kingdom
| | - Adrienn Blasko
- Medical Research Council Cancer Unit, University of Cambridge, United Kingdom
| | - Jonathan White
- Nottingham Digestive Diseases Centre, National Institute of Health Research Nottingham Biomedical Research Centre, United Kingdom
| | - Philip Kaye
- Department of Histopathology, Nottingham University Hospitals National Health Service Trust, University of Nottingham, United Kingdom
| | - Marco Novelli
- Department of Histopathology, University College London Hospital, Longdon, United Kingdom
| | - Lorenz Wernisch
- BIOS Health, Ltd, Cambridge, United Kingdom; Medical Research Council Biostatistics Unit, University of Cambridge, United Kingdom
| | - Krish Ragunath
- Nottingham Digestive Diseases Centre, National Institute of Health Research Nottingham Biomedical Research Centre, United Kingdom
| | - Massimiliano di Pietro
- Medical Research Council Cancer Unit, University of Cambridge, United Kingdom; Department of Gastroenterology, University Hospital National Health Service Foundation Trust, United Kingdom.
| |
Collapse
|
5
|
Karim M, Singh G, Thakur S, Rana A, Rub A, Akhter Y. Evaluating complete surface-associated and secretory proteome of Leishmania donovani for discovering novel vaccines and diagnostic targets. Arch Microbiol 2022; 204:604. [PMID: 36069945 DOI: 10.1007/s00203-022-03219-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
Abstract
The protozoa Leishmania donovani causes visceral leishmaniasis (kala-azar), the third most common vector-borne disease. The visceral organs, particularly the spleen, liver, and bone marrow, are affected by the disease. The lack of effective treatment regimens makes curing and eradicating the disease difficult. The availability of complete L. donovani genome/proteome data allows for the development of specific and efficient vaccine candidates using the reverse vaccinology method, while utilizing the unique sequential and structural features of potential antigenic proteins to induce protective T cell and B cell responses. Such shortlisted candidates may then be tested quickly for their efficacy in the laboratory and later in clinical settings. These antigens will also be useful for designing antigen-based next-generation sero-diagnostic assays. L. donovani's cell surface-associated proteins and secretory proteins are among the first interacting entities to be exposed to the host immune machinery. As a result, potential antigenic epitope peptides derived from these proteins could serve as competent vaccine components. We used a stepwise filtering-based in silico approach to identify the entire surface-associated and secretory proteome of L. donovani, which may provide rationally selected most exposed antigenic proteins. Our study identified 12 glycosylphosphatidylinositol-anchored proteins, 45 transmembrane helix-containing proteins, and 73 secretory proteins as potent antigens unique to L. donovani. In addition, we used immunoinformatics to identify B and T cell epitopes in them. Out of the shortlisted surface-associated and secretory proteome, 66 protein targets were found to have the most potential overlapping B cell and T cell epitopes (linear and conformational; MHC class I and MHC class II).
Collapse
Affiliation(s)
- Munawwar Karim
- School of Life Sciences, Central University of Himachal Pradesh, District-Kangra, Shahpur, Himachal Pradesh, 176206, India
| | - Garima Singh
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, 226025, India
| | - Shweta Thakur
- School of Life Sciences, Central University of Himachal Pradesh, District-Kangra, Shahpur, Himachal Pradesh, 176206, India
| | - Aarti Rana
- School of Life Sciences, Central University of Himachal Pradesh, District-Kangra, Shahpur, Himachal Pradesh, 176206, India
| | - Abdur Rub
- Infection and Immunity Lab, Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, 226025, India.
| |
Collapse
|
6
|
Tandon R, Reyaz E, Roshanara, Jadhav M, Gandhi M, Dey R, Salotra P, Nakhasi HL, Selvapandiyan A. Identification of protein biomarkers of attenuation and immunogenicity of centrin or p27 gene deleted live vaccine candidates of Leishmania against visceral leishmaniasis. Parasitol Int 2022; 92:102661. [PMID: 36049661 DOI: 10.1016/j.parint.2022.102661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 07/08/2022] [Accepted: 08/23/2022] [Indexed: 10/15/2022]
Abstract
Currently, no licensed vaccine is available for human visceral leishmaniasis (VL), a fatal disease caused by the protozoan parasite Leishmania donovani. Two of our live attenuated L. donovani vaccine candidates, either deleted for Centrin1 (LdCen1-/-) or p27 gene (Ldp27-/-), that display reduced growth in macrophages were studied to be safe, immunogenic and protective against VL in various animal models. This report involves the identification of differentially expressed proteins, their related pathways and its underlying mechanism in the intracellular stage of these parasites, using Isobaric Tags for Relative and Absolute Quantitation (iTRAQ) methods. Out of 50-60 proteins, found to be differentially expressed in these mutant parasites, 36 were found to be common in both the parasites. Such proteins mainly belong to the functional categories viz. metabolic enzymes, chaperones and stress proteins, proteins involved in translation, processing and transport and proteins involved in nucleic acid processing. Proteins known to be host protective, like Glyceraldehyde-3-phosphate dehydrogenase (GAPDH), cytochrome c, calreticulin and those responsible for inducing immune response, namely tubulins, DEAD box RNA helicases, HSP70 and tryparedoxin, have been detected to be modulated in these parasites. Such proteins could be predicted as biomarkers, with further scope of study for their role in growth attenuation. SIGNIFICANCE: This study aims at predicting proteomic biomarkers of Leishmania parasite growth attenuation, that have immunomodulatory role in the disease leishmaniasis. Advanced studies could be helpful in establishing the role of these identified proteins in parasitic virulence and to predict the host interaction at molecular level. Also, these proteins could be exploited as attenuation markers during the development of genetically modified live attenuated parasites as vaccine candidates. These could be cross validated in varied species of Leishmania and other tyrpanosomatids for similar response towards identifying them as universal biomarkers of attenuation.
Collapse
Affiliation(s)
- Rati Tandon
- JH-Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Enam Reyaz
- JH-Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Roshanara
- JH-Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Manali Jadhav
- Centre for Research in Nanotechnology & Science, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Mayuri Gandhi
- Centre for Research in Nanotechnology & Science, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Poonam Salotra
- National Institute of Pathology (ICMR), Safdarjung Hospital Campus, New Delhi 110029, India
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Angamuthu Selvapandiyan
- JH-Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
7
|
Alonso A, Larraga J, Loayza FJ, Martínez E, Valladares B, Larraga V, Alcolea PJ. Stable Episomal Transfectant Leishmania infantum Promastigotes Over-Expressing the DEVH1 RNA Helicase Gene Down-Regulate Parasite Survival Genes. Pathogens 2022; 11:pathogens11070761. [PMID: 35890006 PMCID: PMC9323391 DOI: 10.3390/pathogens11070761] [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: 05/19/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 12/10/2022] Open
Abstract
The compartmentalization of untranslated mRNA molecules in granules occurring in many eukaryotic organisms including trypanosomatids involves the formation of complexes between mRNA molecules and RNA-binding proteins (RBPs). The putative ATP-dependent DEAD/H RNA helicase (DEVH1) from Leishmania infantum (Kinetoplastida: Trypanosomatidae) is one such proteins. The objective of this research is finding differentially expressed genes in a stable episomal transfectant L. infantum promastigote line over-expressing DEVH1 in the stationary phase of growth in axenic culture to get insight into the biological roles of this RNA helicase in the parasite. Interestingly, genes related to parasite survival and virulence factors, such as the hydrophilic surface protein/small hydrophilic endoplasmic reticulum protein (HASP/SHERP) gene cluster, an amastin, and genes related to reactive oxygen species detoxification are down-regulated in DEVH1 transfectant promastigotes.
Collapse
Affiliation(s)
- Ana Alonso
- Laboratory of Molecular Parasitology and Vaccines, Biological, Immunological, and Chemical Drug Development for Global Health Unit (BICS), Department of Cellular and Molecular Biology, Center for Biological Research Margarita Salas, Spanish Research Council (CIBMS-CSIC), Calle Ramiro de Maeztu 9, 28040 Madrid, Spain; (A.A.); (J.L.); (F.J.L.); (V.L.)
| | - Jaime Larraga
- Laboratory of Molecular Parasitology and Vaccines, Biological, Immunological, and Chemical Drug Development for Global Health Unit (BICS), Department of Cellular and Molecular Biology, Center for Biological Research Margarita Salas, Spanish Research Council (CIBMS-CSIC), Calle Ramiro de Maeztu 9, 28040 Madrid, Spain; (A.A.); (J.L.); (F.J.L.); (V.L.)
| | - Francisco Javier Loayza
- Laboratory of Molecular Parasitology and Vaccines, Biological, Immunological, and Chemical Drug Development for Global Health Unit (BICS), Department of Cellular and Molecular Biology, Center for Biological Research Margarita Salas, Spanish Research Council (CIBMS-CSIC), Calle Ramiro de Maeztu 9, 28040 Madrid, Spain; (A.A.); (J.L.); (F.J.L.); (V.L.)
| | - Enrique Martínez
- Department of Obstetrics and Gynecology, Pediatrics, Preventive Medicine and Public Health, Toxicology, Legal and Forensic Medicine and Parasitology, Faculty of Pharmacy, University Institute of Public Health of the Canary Islands (IUETSPC), University of La Laguna (ULL), Avda, Astrofísico Francisco, Sánchez s/n, Campus de Anchieta, 38207 La Laguna, Spain; (E.M.); (B.V.)
| | - Basilio Valladares
- Department of Obstetrics and Gynecology, Pediatrics, Preventive Medicine and Public Health, Toxicology, Legal and Forensic Medicine and Parasitology, Faculty of Pharmacy, University Institute of Public Health of the Canary Islands (IUETSPC), University of La Laguna (ULL), Avda, Astrofísico Francisco, Sánchez s/n, Campus de Anchieta, 38207 La Laguna, Spain; (E.M.); (B.V.)
| | - Vicente Larraga
- Laboratory of Molecular Parasitology and Vaccines, Biological, Immunological, and Chemical Drug Development for Global Health Unit (BICS), Department of Cellular and Molecular Biology, Center for Biological Research Margarita Salas, Spanish Research Council (CIBMS-CSIC), Calle Ramiro de Maeztu 9, 28040 Madrid, Spain; (A.A.); (J.L.); (F.J.L.); (V.L.)
| | - Pedro José Alcolea
- Laboratory of Molecular Parasitology and Vaccines, Biological, Immunological, and Chemical Drug Development for Global Health Unit (BICS), Department of Cellular and Molecular Biology, Center for Biological Research Margarita Salas, Spanish Research Council (CIBMS-CSIC), Calle Ramiro de Maeztu 9, 28040 Madrid, Spain; (A.A.); (J.L.); (F.J.L.); (V.L.)
- Correspondence: ; Tel.: +34-9-1837-3112; Fax: +34-9-1536-0432
| |
Collapse
|
8
|
Mukkala AN, Kariyawasam R, Lau R, Valencia BM, Llanos-Cuentas A, Boggild AK. Elevated baseline expression of seven virulence factor RNA transcripts in visceralizing species of Leishmania: a preliminary quantitative PCR study. Ther Adv Infect Dis 2022; 9:20499361221102665. [PMID: 35663431 PMCID: PMC9158425 DOI: 10.1177/20499361221102665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 05/04/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction: Leishmaniasis is a neglected tropical disease that manifests as three major disease phenotypes: cutaneous, mucocutaneous, and visceral. In this preliminary study, we quantified virulence factor (VF) RNA transcript expression in Leishmania species, stratified by geographic origin and propensity for specific disease phenotypes. Methods: Cultured promastigotes of 19 Leishmania clinical and ATCC isolates were extracted for total cellular RNA, cDNA was reverse transcribed, and qPCR assays were performed to quantify VF RNA transcript expression for hsp23, hsp70, hsp83, hsp100, mpi, cpb, and gp63. Results: Comparison of visceralizing species (Leishmania donovani, Leishmania chagasi, and Leishmania infantum) versus non-visceralizing species [Leishmania (Viannia) spp., Leishmania tropica, Leishmania major, Leishmania mexicana, and Leishmania amazonensis] revealed a significantly greater pooled transcript expression for visceralizing species (p = 0.0032). Similarly, Old World species demonstrated significantly higher VF RNA transcript expression than New World species (p = 0.0015). On a per-gene basis, species with a propensity to visceralize ubiquitously expressed higher levels of gp63 (p = 0.005), cpb (p = 0.0032), mpi (p = 0.0032), hsp23 (p = 0.0039), hsp70 (p = 0.0032), hsp83 (p = 0.0032), and hsp100 (p = 0.0032). Conclusion: Here, we provide quantitative, preliminary evidence of elevated VF RNA transcript expression driven largely by the visceralizing causative species of Leishmania. This work highlights the extensive heterogeneity in pathogenicity mechanisms between Leishmania species, which may partly underpin the fatal progression of visceral leishmaniasis.
Collapse
Affiliation(s)
| | - Ruwandi Kariyawasam
- Division of Diagnostic and Applied Microbiology, University of Alberta, Edmonton, AB, Canada
- Alberta Precision Laboratories-Public Health Laboratory (ProvLab), Edmonton, AB, Canada
| | - Rachel Lau
- Public Health Ontario Laboratory, Toronto, ON, Canada
| | - Braulio M. Valencia
- Viral Immunology Systems Program, Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Alejandro Llanos-Cuentas
- Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Andrea K. Boggild
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Tropical Disease Unit, Toronto General Hospital, Room 13EN-218, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
9
|
Zabala-Peñafiel A, Cysne-Finkelstein L, Conceição-Silva F, Fagundes A, Miranda LDFC, Souza-Silva F, Brandt AAML, Dias-Lopes G, Alves CR. Novel Insights Into Leishmania (Viannia) braziliensis In Vitro Fitness Guided by Temperature Changes Along With Its Subtilisins and Oligopeptidase B. Front Cell Infect Microbiol 2022; 12:805106. [PMID: 35531337 PMCID: PMC9069558 DOI: 10.3389/fcimb.2022.805106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Proteases are virulence factors with a recognized impact on the Leishmania spp. life cycle. This study considers a set of analyses measuring phenotypic factors of L. (V.) braziliensis clinical isolates as promastigotes growth curves, murine peritoneal macrophages infection, inflammatory mediators production, and serine proteases gene expression (subtilisin 13: S13, subtilisin 28: S28, oligopeptidase B: OPB) assessing these isolates’ fitness on in vitro conditions. Parasites had different behavior during the early growth phase from day zero to day three, and all isolates reached the stationary growth phase between days four and seven. Macrophages infection showed two tendencies, one of decreased infection rate and number of parasites per macrophage (Infection Index <1000) and another with a constant infection index (≥1400). TNF-α (≥10 pg/mL) detected in infections by 75% of isolates, IL-6 (≥80 pg/mL) by 30% of isolates and low levels of NO (≥0.01µM) in almost all infections. Gene expression showed higher values of S13 (≥2RQ) in the intracellular amastigotes of all the isolates evaluated. On the contrary, S28 expression was low (≤1RQ) in all isolates. OPB expression was different between promastigotes and intracellular amastigotes, being significantly higher (≥2RQ) in the latter form of 58% of the isolates. Predictive structural assays of S13 and OPB were performed to explore temperature influence on gene expression and the encoded proteases. Gene expression data is discussed based on in silico predictions of regulatory regions that show plasticity in the linearity index of secondary structures of S13 and OPB 3’-untranslated regions of mRNA, dependent on temperature changes. While hairpin structures suggest an active region of mRNA for both genes above 26°C, pseudoknot structure found in S13 is an indication of a particular profile of this gene at mammalian host temperatures (37°C). Furthermore, the predicted 3D structures are in accordance with the influence of these temperatures on the catalytic site stability of both enzymes, favoring their action over peptide substrates. Data gathered here suggest that L. (V.) braziliensis serine proteases can be influenced by the temperature conditions affecting parasite fitness throughout its life cycle.
Collapse
Affiliation(s)
- Anabel Zabala-Peñafiel
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Lea Cysne-Finkelstein
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Fatima Conceição-Silva
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Aline Fagundes
- Laboratório de Pesquisa Clínica e Vigilância em Leishmanioses, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Luciana de Freitas Campos Miranda
- Laboratório de Pesquisa Clínica e Vigilância em Leishmanioses, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Franklin Souza-Silva
- Centro de Desenvolvimento Tecnológico em Saúde, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Universidade Iguaçu, Dom Rodrigo, Nova Iguaçu, Rio de Janeiro, Brazil
| | - Artur A. M. L. Brandt
- Departamento de Computação e Sistemas, Faculdade de Educação Tecnológica do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Ciência da Computação, Univeritas-Rio, Rio de Janeiro, Brazil
| | - Geovane Dias-Lopes
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- *Correspondence: Geovane Dias-Lopes, ; Carlos Roberto Alves,
| | - Carlos Roberto Alves
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- *Correspondence: Geovane Dias-Lopes, ; Carlos Roberto Alves,
| |
Collapse
|
10
|
Medina J, Cruz-Saavedra L, Patiño LH, Muñoz M, Ramírez JD. Comparative analysis of the transcriptional responses of five Leishmania species to trivalent antimony. Parasit Vectors 2021; 14:419. [PMID: 34419127 PMCID: PMC8380399 DOI: 10.1186/s13071-021-04915-y] [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: 04/29/2021] [Accepted: 08/02/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Leishmaniasis is a neglected tropical disease caused by several species of Leishmania. The resistance phenotype of these parasites depends on the characteristics of each species, which contributes to increased therapeutic failures. Understanding the mechanism used by the parasite to survive under treatment pressure in order to identify potential common and specific therapeutic targets is essential for the control of leishmaniasis. The aim of this study was to investigate the expression profiles and potential shared and specific resistance markers of the main Leishmania species of medical importance [subgenus L. (Leishmania): L. donovani, L. infantum and L. amazonensis; subgenus L. (Viannia): L. panamensis and L. braziliensis)] resistant and sensitive to trivalent stibogluconate (SbIII). METHODS We conducted comparative analysis of the transcriptomic profiles (only coding sequences) of lines with experimentally induced resistance to SbIII from biological replicates of five Leishmania species available in the databases of four articles based on ortholog attribution. Simultaneously, we carried out functional analysis of ontology and reconstruction of metabolic pathways of the resulting differentially expressed genes (DEGs). RESULTS Resistant lines for each species had differential responses in metabolic processes, compound binding, and membrane components concerning their sensitive counterpart. One hundred and thirty-nine metabolic pathways were found, with the three main pathways comprising cysteine and methionine metabolism, glycolysis, and the ribosome. Differentially expressed orthologous genes assigned to species-specific responses predominated, with 899 self-genes. No differentially expressed genes were found in common among the five species. Two common upregulated orthologous genes were found among four species (L. donovani, L. braziliensis, L. amazonensis, and L. panamensis) related to an RNA-binding protein and the NAD(P)H cytochrome-B5-oxidoreductase complex, associated with transcriptional control and de novo synthesis of linoleic acid, critical mechanisms in resistance to antimonials. CONCLUSION Herein, we identified potential species-specific genes related to resistance to SbIII. Therefore, we suggest that future studies consider a treatment scheme that is species-specific. Despite the limitations of our study, this is the first approach toward unraveling the pan-genus genetic mechanisms of resistance in leishmaniasis.
Collapse
Affiliation(s)
- Julián Medina
- Centro de Investigaciones en Microbiología y Biotecnología- UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Lissa Cruz-Saavedra
- Centro de Investigaciones en Microbiología y Biotecnología- UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Luz Helena Patiño
- Centro de Investigaciones en Microbiología y Biotecnología- UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología- UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología- UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
11
|
Papadaki A, Tziouvara O, Kotopouli A, Koumarianou P, Doukas A, Rios P, Tardieux I, Köhn M, Boleti H. The Leishmania donovani LDBPK_220120.1 Gene Encodes for an Atypical Dual Specificity Lipid-Like Phosphatase Expressed in Promastigotes and Amastigotes; Substrate Specificity, Intracellular Localizations, and Putative Role(s). Front Cell Infect Microbiol 2021; 11:591868. [PMID: 33842381 PMCID: PMC8027504 DOI: 10.3389/fcimb.2021.591868] [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: 08/05/2020] [Accepted: 01/25/2021] [Indexed: 11/13/2022] Open
Abstract
The intracellular protozoan parasites of the Leishmania genus are responsible for Leishmaniases, vector borne diseases with a wide range of clinical manifestations. Leishmania (L.) donovani causes visceral leishmaniasis (kala azar), the most severe of these diseases. Along their biological cycle, Leishmania parasites undergo distinct developmental transitions including metacyclogenesis and differentiation of metacyclic promastigotes (MPs) to amastigotes. Metacyclogenesis inside the phlebotomine sandfly host's midgut converts the procyclic dividing promastigotes to non-dividing infective MPs eventually injected into the skin of mammalian hosts and phagocytosed by macrophages where the MPs are converted inside modified phagolysosomes to the intracellular amastigotes. These developmental transitions involve dramatic changes in cell size and shape and reformatting of the flagellum requiring thus membrane and cytoskeleton remodeling in which phosphoinositide (PI) signaling and metabolism must play central roles. This study reports on the LDBPK_220120.1 gene, the L. donovani ortholog of LmjF.22.0250 from L. major that encodes a phosphatase from the "Atypical Lipid Phosphatases" (ALPs) enzyme family. We confirmed the expression of the LDBPK_220120.1 gene product in both L. donovani promastigotes and axenic amastigotes and showed that it behaves in vitro as a Dual Specificity P-Tyr and monophosphorylated [PI(3)P and PI(4)P] PI phosphatase and therefore named it LdTyrPIP_22 (Leishmaniad onovani Tyrosine PI Phosphatase, gene locus at chromosome 22). By immunofluorescence confocal microscopy we localized the LdTyrPIP_22 in several intracellular sites in the cell body of L. donovani promastigotes and amastigotes and in the flagellum. A temperature and pH shift from 25°C to 37°C and from pH 7 to 5.5, induced a pronounced recruitment of LdTyrPIP_22 epitopes to the flagellar pocket and a redistribution around the nucleus. These results suggest possible role(s) for this P-Tyr/PI phosphatase in the regulation of processes initiated or upregulated by this temperature/pH shift that contribute to the developmental transition from MPs to amastigotes inside the mammalian host macrophages.
Collapse
Affiliation(s)
- Amalia Papadaki
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Olympia Tziouvara
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Anastasia Kotopouli
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Petrina Koumarianou
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece.,Light Microscopy Unit, Hellenic Pasteur Institute, Athens, Greece
| | - Anargyros Doukas
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Pablo Rios
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Isabelle Tardieux
- Team «Biomechanics of Host Parasite Interactions», Institut for Advanced BioSciences, Univ. Grenoble Alpes, Inserm U1209 - CNRS UMR 5309, 38700 La Tronche, France
| | - Maja Köhn
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Haralabia Boleti
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece.,Light Microscopy Unit, Hellenic Pasteur Institute, Athens, Greece
| |
Collapse
|
12
|
Lima BSDS, Esteves BB, Fialho-Júnior LC, Mendes TADO, Pires SDF, Chapeourouge A, Perales J, de Andrade HM. Study of the differentially abundant proteins among Leishmania amazonensis, L. braziliensis, and L. infantum. PLoS One 2020; 15:e0240612. [PMID: 33057350 PMCID: PMC7561129 DOI: 10.1371/journal.pone.0240612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/29/2020] [Indexed: 01/05/2023] Open
Abstract
Leishmaniasis has been considered as emerging and re-emerging disease, and its increasing global incidence has raised concerns. The great clinical diversity of the disease is mainly determined by the species. In several American countries, tegumentary leishmaniasis (TL) is associated with both Leishmania amazonensis and L. braziliensis, while visceral leishmaniasis (VL) is associated with L. (L.) infantum. The major molecules that determine the most diverse biological variations are proteins. In the present study, through a DIGE approach, we identified differentially abundant proteins among the species mentioned above. We observed a variety of proteins with differential abundance among the studied species; and the biological networks predicted for each species showed that many of these proteins interacted with each other. The prominent proteins included the heat shock proteins (HSPs) and the protein network involved in oxide reduction process in L. amazonensis, the protein network of ribosomes in L. braziliensis, and the proteins involved in energy metabolism in L. infantum. The important proteins, as revealed by the PPI network results, enrichment categories, and exclusive proteins analysis, were arginase, HSPs, and trypanothione reductase in L. amazonensis; enolase, peroxidoxin, and tryparedoxin1 in L. braziliensis; and succinyl-CoA ligase [GDP -forming] beta-chain and transaldolase in L. infantum.
Collapse
Affiliation(s)
- Bruna Soares de Souza Lima
- Departamento de Medicina, Faculdade Dinâmica do Vale do Piranga (FADIP), Ponte Nova, Minas Gerais, Brazil
- Departamento de Parasitologia, Laboratório de Leishmanioses, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Barbara Beiral Esteves
- Departamento de Parasitologia, Laboratório de Leishmanioses, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz Carlos Fialho-Júnior
- Departamento de Parasitologia, Laboratório de Leishmanioses, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Simone da Fonseca Pires
- Departamento de Parasitologia, Laboratório de Leishmanioses, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Jonas Perales
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Helida Monteiro de Andrade
- Departamento de Parasitologia, Laboratório de Leishmanioses, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
13
|
García-Sánchez M, Jiménez-Pelayo L, Horcajo P, Regidor-Cerrillo J, Collantes-Fernández E, Ortega-Mora LM. Gene Expression Profiling of Neospora caninum in Bovine Macrophages Reveals Differences Between Isolates Associated With Key Parasite Functions. Front Cell Infect Microbiol 2019; 9:354. [PMID: 31681630 PMCID: PMC6803445 DOI: 10.3389/fcimb.2019.00354] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/01/2019] [Indexed: 12/19/2022] Open
Abstract
Intraspecific differences in biological traits between Neospora caninum isolates have been widely described and associated with variations in virulence. However, the molecular basis underlying these differences has been poorly studied. We demonstrated previously that Nc-Spain7 and Nc-Spain1H, high- and low-virulence isolates, respectively, show different invasion, proliferation and survival capabilities in bovine macrophages (boMØs), a key cell in the immune response against Neospora, and modulate the cell immune response in different ways. Here, we demonstrate that these differences are related to specific tachyzoite gene expression profiles. Specifically, the low-virulence Nc-Spain1H isolate showed enhanced expression of genes encoding for surface antigens and genes related to the bradyzoite stage. Among the primary up-regulated genes in Nc-Spain7, genes involved in parasite growth and redox homeostasis are particularly noteworthy because of their correlation with the enhanced proliferation and survival rates of Nc-Spain7 in boMØs relative to Nc-Spain1H. Genes potentially implicated in induction of proinflammatory immune responses were found to be up-regulated in the low-virulence isolate, whereas the high-virulence isolate showed enhanced expression of genes that may be involved in immune evasion. These results represent a further step in understanding the parasite effector molecules that may be associated to virulence and thus to disease traits as abortion and transmission.
Collapse
Affiliation(s)
- Marta García-Sánchez
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - Laura Jiménez-Pelayo
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - Pilar Horcajo
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - Javier Regidor-Cerrillo
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain.,Saluvet-Innova, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - Esther Collantes-Fernández
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - Luis Miguel Ortega-Mora
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
14
|
Major changes in chromosomal somy, gene expression and gene dosage driven by Sb III in Leishmania braziliensis and Leishmania panamensis. Sci Rep 2019; 9:9485. [PMID: 31263131 PMCID: PMC6603004 DOI: 10.1038/s41598-019-45538-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/06/2019] [Indexed: 12/17/2022] Open
Abstract
Leishmania braziliensis and Leishmania panamensis are two species clinically and epidemiologically important, among others because of their relative resistance to first-line drugs (antimonials). The precise mechanism underlying the ability of these species to survive antimony treatment remains unknown. Therefore, elucidating the pathways mediating drug resistance is essential. We herein experimentally selected resistance to trivalent antimony (SbIII) in the reference strains of L. braziliensis (MHOM/BR75/M2904) and L. panamensis (MHOM/COL/81L13) and compared whole genome and transcriptome alterations in the culture promastigote stage. The results allowed us to identify differences in somy, copy number variations in some genes related to antimony resistance and large-scale copy number variations (deletions and duplications) in chromosomes with no somy changes. We found mainly in L. braziliensis, a direct relation between the chromosomal/local copy number variation and the gene expression. We identified differentially expressed genes in the resistant lines that are involved in antimony resistance, virulence, and vital biological processes in parasites. The results of this study may be useful for characterizing the genetic mechanisms of these Leishmania species under antimonial pressure, and for clarifying why the parasites are resistant to first-line drug treatments.
Collapse
|
15
|
de Pablos LM, Ferreira TR, Dowle AA, Forrester S, Parry E, Newling K, Walrad PB. The mRNA-bound Proteome of Leishmania mexicana: Novel Genetic Insight into an Ancient Parasite. Mol Cell Proteomics 2019; 18:1271-1284. [PMID: 30948621 PMCID: PMC6601212 DOI: 10.1074/mcp.ra118.001307] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/23/2019] [Indexed: 01/08/2023] Open
Abstract
Leishmania parasite infections, termed the leishmaniases, cause significant global infectious disease burden. The lifecycle of the parasite embodies three main stages that require precise coordination of gene regulation to survive environmental shifts between sandfly and mammalian hosts. Constitutive transcription in kinetoplastid parasites means that gene regulation is overwhelmingly reliant on post-transcriptional mechanisms, yet strikingly few Leishmania trans-regulators are known. Using optimized crosslinking and deep, quantified mass spectrometry, we present a comprehensive analysis of 1400 mRNA binding proteins (mRBPs) and whole cell proteomes from the three main Leishmania lifecycle stages. Supporting the validity, although the crosslinked RBPome is magnitudes more enriched, the protein identities of the crosslinked and non-crosslinked RBPomes were nearly identical. Moreover, multiple candidate RBPs were endogenously tagged and found to associate with discrete mRNA target pools in a stage-specific manner. Results indicate that in L. mexicana parasites, mRNA levels are not a strong predictor of the whole cell expression or RNA binding potential of encoded proteins. Evidence includes a low correlation between transcript and corresponding protein expression and stage-specific variation in protein expression versus RNA binding potential. Unsurprisingly, RNA binding protein enrichment correlates strongly with relative replication efficiency of the specific lifecycle stage. Our study is the first to quantitatively define and compare the mRBPome of multiple stages in kinetoplastid parasites. It provides novel, in-depth insight into the trans-regulatory mRNA:Protein (mRNP) complexes that drive Leishmania parasite lifecycle progression.
Collapse
Affiliation(s)
| | | | - Adam A Dowle
- §Metabolomics and Proteomics Lab, Bioscience Technology Facility, and
| | | | - Ewan Parry
- From the ‡Centre for Immunology and Infection
| | - Katherine Newling
- ¶Genomics and Bioinformatics Lab, Bioscience Technology Facility, Department of Biology, University of York, UK
| | | |
Collapse
|
16
|
Abstract
Background Leishmania development in the sand fly gut leads to highly infective forms called metacyclic promastigotes. This process can be routinely mimicked in culture. Gene expression–profiling studies by transcriptome analysis have been performed with the aim of studying promastigote forms in the sand fly gut, as well as differences between sand fly–and culture-derived promastigotes. Findings Transcriptome analysis has revealed the crucial role of the microenvironment in parasite development within the sand fly gut because substantial differences and moderate correlation between the transcriptomes of cultured and sand fly–derived promastigotes have been found. Sand fly–derived metacyclics are more infective than metacyclics in culture. Therefore, some caution should be exercised when using cultured promastigotes, depending on the experimental design. The most remarkable examples are the hydrophilic acidic surface protein/small endoplasmic reticulum protein (HASP/SHERP) cluster, the glycoprotein 63 (gp63), and autophagy genes, which are up-regulated in sand fly–derived promastigotes compared with cultured promastigotes. Because HASP/SHERP genes are up-regulated in nectomonad and metacyclic promastigotes in the sand fly, the encoded proteins are not metacyclic specific. Metacyclic promastigotes are distinguished by morphology and high infectivity. Isolating them from the sand fly gut is not exempt from technical difficulty, because other promastigote forms remain in the gut even 15 days after infection. Leishmania major procyclic promastigotes within the sand fly gut up-regulate genes involved in cell cycle regulation and glucose catabolism, whereas metacyclics increase transcript levels of fatty acid biosynthesis and ATP-coupled proton transport genes. Most parasite's signal transduction pathways remain uncharacterized. Future elucidation may improve understanding of parasite development, particularly signaling molecule-encoding genes in sand fly versus culture and between promastigote forms in the sand fly gut. Conclusions Transcriptome analysis has been demonstrated to be technically efficacious to study differential gene expression in sand fly gut promastigote forms. Transcript and protein levels are not well correlated in these organisms (approximately 25% quantitative coincidences), especially under stress situations and at differentiation processes. However, transcript and protein levels behave similarly in approximately 60% of cases from a qualitative point of view (increase, decrease, or no variation). Changes in translational efficiency observed in other trypanosomatids strongly suggest that the differences are due to translational regulation and regulation of the steady-state protein levels. The lack of low-input sample strategies does not allow translatome and proteome analysis of sand fly–derived promastigotes so far.
Collapse
|
17
|
Rastrojo A, Corvo L, Lombraña R, Solana JC, Aguado B, Requena JM. Analysis by RNA-seq of transcriptomic changes elicited by heat shock in Leishmania major. Sci Rep 2019; 9:6919. [PMID: 31061406 PMCID: PMC6502937 DOI: 10.1038/s41598-019-43354-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 04/23/2019] [Indexed: 12/03/2022] Open
Abstract
Besides their medical relevance, Leishmania is an adequate model for studying post-transcriptional mechanisms of gene expression. In this microorganism, mRNA degradation/stabilization mechanisms together with translational control and post-translational modifications of proteins are the major drivers of gene expression. Leishmania parasites develop as promastigotes in sandflies and as amastigotes in mammalians, and during host transmission, the parasite experiences a sudden temperature increase. Here, changes in the transcriptome of Leishmania major promastigotes after a moderate heat shock were analysed by RNA-seq. Several of the up-regulated transcripts code for heat shock proteins, other for proteins previously reported to be amastigote-specific and many for hypothetical proteins. Many of the transcripts experiencing a decrease in their steady-state levels code for transporters, proteins involved in RNA metabolism or translational factors. In addition, putative long noncoding RNAs were identified among the differentially expressed transcripts. Finally, temperature-dependent changes in the selection of the spliced leader addition sites were inferred from the RNA-seq data, and particular cases were further validated by RT-PCR and Northern blotting. This study provides new insights into the post-transcriptional mechanisms by which Leishmania modulate gene expression.
Collapse
Affiliation(s)
- Alberto Rastrojo
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Campus de Excelencia Internacional (CEI) UAM+CSIC, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Corvo
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Campus de Excelencia Internacional (CEI) UAM+CSIC, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rodrigo Lombraña
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Campus de Excelencia Internacional (CEI) UAM+CSIC, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jose C Solana
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Campus de Excelencia Internacional (CEI) UAM+CSIC, Universidad Autónoma de Madrid, Madrid, Spain
| | - Begoña Aguado
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Campus de Excelencia Internacional (CEI) UAM+CSIC, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Jose M Requena
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Campus de Excelencia Internacional (CEI) UAM+CSIC, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
18
|
Kariyawasam R, Mukkala AN, Lau R, Valencia BM, Llanos-Cuentas A, Boggild AK. Virulence factor RNA transcript expression in the Leishmania Viannia subgenus: influence of species, isolate source, and Leishmania RNA virus-1. Trop Med Health 2019; 47:25. [PMID: 31007536 PMCID: PMC6458769 DOI: 10.1186/s41182-019-0153-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Leishmania RNA virus-1 (LRV1) is a double-stranded RNA virus identified in 20-25% of Viannia-species endemic to Latin America, and is believed to accelerate cutaneous to mucosal leishmaniasis over time. Our objective was to quantify known virulence factor (VF) RNA transcript expression according to LRV1 status, causative species, and isolate source. METHODS Eight cultured isolates of Leishmania were used, four of which were LRV1-positive (Leishmania Viannia braziliensis [n = 1], L. (V.) guyanensis [n = 1], L. (V.) panamensis [n = 2]), and four were LRV1-negative (L. (V.) panamensis [n = 3], L. (V.) braziliensis [n = 1]). Promastigotes were inoculated into macrophage cultures, and harvested at 24 and 48 h. RNA transcript expression of hsp23, hsp70, hsp90, hsp100, mpi, cpb, and gp63 were quantified by qPCR. RESULTS RNA transcript expression of hsp100 (p = 0.012), cpb (p = 0.016), and mpi (p = 0.022) showed significant increases from baseline pure culture expression to 24- and 48-h post-macrophage infection, whereas hsp70 (p = 0.004) was significantly decreased. A trend toward increased transcript expression of hsp100 at baseline in isolates of L. (V.) panamensis was noted. Pooled VF RNA transcript expression by L. (V.) panamensis isolates was lower than that of L. (V.) braziliensis and L. (V.) guyananesis at 24 h (p = 0.03). VF RNA transcript expression did not differ by LRV1 status, or source of cultured isolate at baseline, 24, or 48 h; however, a trend toward increased VF RNA transcript expression of 2.71- and 1.93-fold change of mpi (p = 0.11) and hsp90 (p = 0.11), respectively, in LRV1 negative isolates was noted. Similarly, a trend toward lower levels of overall VF RNA transcript expression in clinical isolates (1.15-fold change) compared to ATCC® strains at 24 h was noted (p = 0.07). CONCLUSIONS Our findings suggest that known VF RNA transcript expression may be affected by the process of macrophage infection. We were unable to demonstrate definitively that LRV-1 presence affected VF RNA transcript expression in the species and isolates studied. L. (V.) guyanensis and L. (V.) braziliensis demonstrated higher pooled VF RNA transcript expression than L. (V.) panamensis; however, further analyses of protein expression to corroborate this finding are warranted.
Collapse
Affiliation(s)
| | - Avinash N. Mukkala
- Institute of Medical Sciences, University of Toronto, Toronto, ON Canada
| | - Rachel Lau
- Public Health Ontario Laboratory, Toronto, ON Canada
| | - Braulio M. Valencia
- Instituto de Medicina Tropical “Alejandro von Humboldt”, Lima, Peru
- Viral Immunology Systems Program, Kirby Institute, University of New South Wales, Sydney, Australia
| | - Alejandro Llanos-Cuentas
- Instituto de Medicina Tropical “Alejandro von Humboldt”, Lima, Peru
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Andrea K. Boggild
- Institute of Medical Sciences, University of Toronto, Toronto, ON Canada
- Public Health Ontario Laboratory, Toronto, ON Canada
- Department of Medicine, University of Toronto, Toronto, ON Canada
- Tropical Disease Unit, Toronto General Hospital, 200 Elizabeth Street, 13EN-218, Toronto, ON M5G 2C4 Canada
| |
Collapse
|
19
|
Samarasinghe SR, Samaranayake N, Kariyawasam UL, Siriwardana YD, Imamura H, Karunaweera ND. Genomic insights into virulence mechanisms of Leishmania donovani: evidence from an atypical strain. BMC Genomics 2018; 19:843. [PMID: 30486770 PMCID: PMC6262978 DOI: 10.1186/s12864-018-5271-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 11/19/2018] [Indexed: 01/20/2023] Open
Abstract
Background Leishmaniasis is a neglected tropical disease with diverse clinical phenotypes, determined by parasite, host and vector interactions. Despite the advances in molecular biology and the availability of more Leishmania genome references in recent years, the association between parasite species and distinct clinical phenotypes remains poorly understood. We present a genomic comparison of an atypical variant of Leishmania donovani from a South Asian focus, where it mostly causes cutaneous form of leishmaniasis. Results Clinical isolates from six cutaneous leishmaniasis patients (CL-SL); 2 of whom were poor responders to antimony (CL-PR), and two visceral leishmaniasis patients (VL-SL) were sequenced on an Illumina MiSeq platform. Chromosome aneuploidy was observed in both groups but was more frequent in CL-SL. 248 genes differed by 2 fold or more in copy number among the two groups. Genes involved in amino acid use (LdBPK_271940) and energy metabolism (LdBPK_271950), predominated the VL-SL group with the same distribution pattern reflected in gene tandem arrays. Genes encoding amastins were present in higher copy numbers in VL-SL and CL-PR as well as being among predicted pseudogenes in CL-SL. Both chromosome and SNP profiles showed CL-SL and VL-SL to form two distinct groups. While expected heterozygosity was much higher in VL-SL, SNP allele frequency patterns did not suggest potential recent recombination breakpoints. The SNP/indel profile obtained using the more recently generated PacBio sequence did not vary markedly from that based on the standard LdBPK282A1 reference. Several genes previously associated with resistance to antimonials were observed in higher copy numbers in the analysis of CL-PR. H-locus amplification was seen in one cutaneous isolate which however did not belong to the CL-PR group. Conclusions The data presented suggests that intra species variations at chromosome and gene level are more likely to influence differences in tropism as well as response to treatment, and contributes to greater understanding of parasite molecular mechanisms underpinning these differences. These findings should be substantiated with a larger sample number and expression/functional studies. Electronic supplementary material The online version of this article (10.1186/s12864-018-5271-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sumudu R Samarasinghe
- Department of Parasitology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Nilakshi Samaranayake
- Department of Parasitology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Udeshika L Kariyawasam
- Department of Parasitology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Yamuna D Siriwardana
- Department of Parasitology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Hideo Imamura
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Nadira D Karunaweera
- Department of Parasitology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.
| |
Collapse
|
20
|
An Insight into the Constitutive Proteome Throughout Leishmania donovani Promastigote Growth and Differentiation. Int Microbiol 2018; 22:143-154. [DOI: 10.1007/s10123-018-00036-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 10/08/2018] [Accepted: 10/10/2018] [Indexed: 01/28/2023]
|
21
|
ROS regulate differentiation of visceralizing Leishmania species into the virulent amastigote form. ACTA ACUST UNITED AC 2018; 4. [PMID: 31093331 DOI: 10.1017/pao.2018.15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Leishmania virulence and disease development critically depends on the ability of Leishmania promastigotes to infect, differentiate into amastigote forms and replicate inside mammalian host macrophages. Understanding changes associated with amastigote differentiation in axenic culture conditions is key to identifying virulence factors. Here we compared efficiency of the conventional pH-temperature-dependent shift method to induce amastigote differentiation with the recently identified trigger for differentiation mediated by mitochondrial reactive oxygen species (ROS). Using two different visceral leishmaniasis species, L. infantum and. L. donovani, we show that ROS-generating methods such as iron deprivation or exposure to sub-lethal concentrations of H2O2 or menadione are significantly more effective in promoting promastigoteamastigote differentiation than the low pH-high temperature shift, leading to higher survival rates, morphological changes and gene expression patterns characteristic of the amastigote stage. Notably, both H2O2 and menadione-mediated differentiation did not require up-regulation of the mitochondrial electron transport chain (ETC)-associated protein p27, suggesting that treatment with oxidants bypasses the necessity to upregulate mitochondrial activity, a precondition for mROS generation. Our findings confirm that ROS-induced differentiation occurs in multiple Leishmania species, including the medically important visceralizing species, and provide mechanistic rationale for earlier reports demonstrating markedly increased virulence of L. infantum promastigotes pre-treated with oxidative reagents.
Collapse
|
22
|
Alonso A, Larraga V, Alcolea PJ. The contribution of DNA microarray technology to gene expression profiling in Leishmania spp.: A retrospective view. Acta Trop 2018; 187:129-139. [PMID: 29746872 DOI: 10.1016/j.actatropica.2018.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/04/2018] [Accepted: 05/06/2018] [Indexed: 01/15/2023]
Abstract
The first completed genome project of any living organism, excluding viruses, was of the gammaproteobacteria Haemophilus influenzae in 1995. Until the last decade, genome sequencing was very tedious because genome survey sequences (GSS) and/or expressed sequence tags (ESTs) belonging to plasmid, cosmid, and artificial chromosome genome libraries had to be sequenced and assembled in silico. No genome is completely assembled because gaps and unassembled contigs are always remaining. However, most represent an organism's whole genome from a practical point of view. The first genome sequencing projects of trypanosomatid parasites Leishmania major, Trypanosoma cruzi, and T. brucei were completed in 2005 following those strategies. The functional genomics era developed on the basis of microarray technology and has been continuously evolving. In the case of the genus Leishmania, substantial information about differentiation in the digenetic life cycle of the parasite has been obtained. More recently, next generation sequencing has revolutionized genome sequencing and functional genomics, leading to more sensitive and accurate results by using much fewer resources. Though this new technology is more advantageous, it does not invalidate microarray results. In fact, promising vaccine candidates and drug targets have been found by means of microarray-based screening and preliminary proof-of-concept tests.
Collapse
|
23
|
Singh N, Goel R, Jain E. Differential Metabolic Pathway Analysis of the Proteomes of Leishmania donovani and Leptomonas seymouri. Proteomics Clin Appl 2018; 12:e1600087. [PMID: 29469990 DOI: 10.1002/prca.201600087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 12/14/2017] [Indexed: 12/26/2022]
Abstract
PURPOSE Although in trypanosomatids, monoxeny (Leptomonas) is ancestral to dixeny (Leishmania), however clinical cases of visceral leishmanisis with Leptomonas co-infection are increasingly being reported from India. Using a proteogenomic approach, a detailed proteome analysis of these two kinetoplastid parasites viz., Leishmania and its sister Leptomonas, to catalog the key proteins associated with and therefore possibly responsible for phenotype changes in Leptomonas evolution and domestication as co-infection with Leishmania is carried out. EXPERIMENTAL DESIGN LC-MS/MS is utilized for this proteomic purpose. One Leishmania donovani WHO reference strain and two Leptomonas seymouri isolates, which are originally isolated from clinical cases of kala azar patients with different inherent drug sensitivity viz., responsive and unresponsive, are used in this study. RESULTS A network analysis, leveraging protein-protein interaction data helped to find the roles of the proteins in carbon metabolism and biosynthesis of secondary metabolites which is seen to be altered under stress conditions like drug resistance. CONCLUSIONS AND CLINICAL RELEVANCE The information provided about the metabolic pathways modulated when contrasting these two phenotypes may lead to the development of new strategies to block parasite differentiation within the host and to also circumvent the problem of drug resistance. This proteomic study also offers new grounds for the investigation of novel hypothetical proteins potentially playing a role in evolutionary biology the knowledge of which is essential for treatment of patients co-infected with these two kinetoplastid parasites.
Collapse
Affiliation(s)
- Neeloo Singh
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Renu Goel
- Drug Discovery Research Centre (DDRC), Translational Health Science and Technology Institute (THSTI), Haryana, India
| | - Ekta Jain
- Consulting Bioinformatician, M.res Bioinformatics, University of Newcastle Upon Tyne, UK, Affiliated with Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| |
Collapse
|
24
|
Soulat D, Bogdan C. Function of Macrophage and Parasite Phosphatases in Leishmaniasis. Front Immunol 2017; 8:1838. [PMID: 29312331 PMCID: PMC5743797 DOI: 10.3389/fimmu.2017.01838] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023] Open
Abstract
The kinetoplastid protozoan parasites belonging to the genus Leishmania are the causative agents of different clinical forms of leishmaniasis, a vector-borne infectious disease with worldwide prevalence. The protective host immune response against Leishmania parasites relies on myeloid cells such as dendritic cells and macrophages in which upon stimulation by cytokines (e.g., interferon-γ) a complex network of signaling pathways is switched on leading to strong antimicrobial activities directed against the intracellular parasite stage. The regulation of these pathways classically depends on post-translational modifications of proteins, with phosphorylation events playing a cardinal role. Leishmania parasites deactivate their phagocytic host cells by inducing specific mammalian phosphatases that are capable to impede signaling. On the other hand, there is now also evidence that Leishmania spp. themselves express phosphatases that might target host cell molecules and thereby facilitate the intracellular survival of the parasite. This review will present an overview on the modulation of host phosphatases by Leishmania parasites as well as on the known families of Leishmania phosphatases and their possible function as virulence factors. A more detailed understanding of the role of phosphatases in Leishmania–host cell interactions might open new avenues for the treatment of non-healing, progressive forms of leishmaniasis.
Collapse
Affiliation(s)
- Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Interdisciplinary Center of the FAU, Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Interdisciplinary Center of the FAU, Erlangen, Germany
| |
Collapse
|
25
|
de Vasconcelos TCB, Furtado MC, Belo VS, Morgado FN, Figueiredo FB. Canine susceptibility to visceral leishmaniasis: A systematic review upon genetic aspects, considering breed factors and immunological concepts. INFECTION GENETICS AND EVOLUTION 2017; 74:103293. [PMID: 28987807 DOI: 10.1016/j.meegid.2017.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/30/2017] [Accepted: 10/03/2017] [Indexed: 01/01/2023]
Abstract
Dogs have different susceptibility degrees to leishmaniasis; however, genetic research on this theme is scarce, manly on visceral form. The aims of this systematic review were to describe and discuss the existing scientific findings on genetic susceptibility to canine leishmaniasis, as well as to show the gaps of the existing knowledge. Twelve articles were selected, including breed immunological studies, genome wide associations or other gene polymorphism or gene sequencing studies, and transcription approaches. As main results of literature, there was a suggestion of genetic clinical resistance background for Ibizan Hound dogs, and alleles associated with protection or susceptibility to visceral leishmaniasis in Boxer dogs. Genetic markers can explain phenotypic variance in both pro- and anti-inflammatory cytokines and in cellular immune responses, including antigen presentation. Many gene segments are involved in canine visceral leishmaniasis phenotype, with Natural Resistance Associated Macrophage Protein 1 (NRAMP1) as the most studied. This was related to both protection and susceptibility. In comparison with murine and human genetic approaches, lack of knowledge in dogs is notorious, with many possibilities for new studies, revealing a wide field to be assessed on canine leishmaniasis susceptibility research.
Collapse
Affiliation(s)
- Tassia Cristina Bello de Vasconcelos
- Centro de Controle de Zoonoses, Vigilância em Saúde, Secretaria Municipal de Saúde, Prefeitura Municipal de Resende, Rua Euridices Paulina de Almeida, 300, Vicentina II, Resende, RJ 27500-000, Brazil.
| | - Marina Carvalho Furtado
- Fiocruz Mata Atlântica, Fundação Oswaldo Cruz, Estrada Rodrigues Caldas, 3400, Taquara, Rio de Janeiro, RJ 22713-375, Brazil
| | - Vinícius Silva Belo
- Universidade Federal de São João del-Rei, campus Centro Oeste Dona Lindu, Rua Sebastião Gonçalves Coelho, 400, Chanadour, Divinópolis, MG 35.501-296, Brazil
| | - Fernanda Nazaré Morgado
- Laboratório de Pesquisa em Leishmaniose, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, 4365, Manguinhos, Rio de Janeiro, RJ 21040-360, Brazil
| | - Fabiano Borges Figueiredo
- Laboratório de Biologia Celular, Instituto Carlos Chagas, Rua Professor Algacyr Munhoz Mader, 3.775, CIC, campus do Tecpar, bloco C, Curitiba, PR 81.350-010 Brazil
| |
Collapse
|
26
|
Characterization of the Protein Tyrosine Phosphatase LmPRL-1 Secreted by Leishmania major via the Exosome Pathway. Infect Immun 2017; 85:IAI.00084-17. [PMID: 28507071 DOI: 10.1128/iai.00084-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/11/2017] [Indexed: 01/02/2023] Open
Abstract
Similar to other intracellular pathogens, Leishmania parasites are known to evade the antimicrobial effector functions of host immune cells. To date, however, only a few virulence factors have been described for Leishmania major, one of the causative agents of cutaneous leishmaniasis. Here, we have characterized the expression and function of an L. major phosphatase, which we termed LmPRL-1. This enzyme shows a strong structural similarity to the human phosphatases of regenerating liver (PRL-1, -2, and -3) that regulate the proliferation, differentiation, and motility of cells. The biochemical characterization of the L. major phosphatase revealed that the enzyme is redox sensitive. When analyzing the subcellular localization of LmPRL-1 in promastigotes, amastigotes, and infected macrophages, we found that the phosphatase was predominantly expressed and secreted by promastigotes via the exosome route. Finally, we observed that ectopic expression of LmPRL-1 in L. major led to an increased number of parasites in macrophages. From these data, we conclude that the L. major phosphatase LmPRL-1 contributes to the intracellular survival of the parasites in macrophages.
Collapse
|
27
|
Immuno-informatics based approaches to identify CD8+ T cell epitopes within the Leishmania donovani 3-ectonucleotidase in cured visceral leishmaniasis subjects. Microbes Infect 2017; 19:358-369. [DOI: 10.1016/j.micinf.2017.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 03/10/2017] [Accepted: 03/24/2017] [Indexed: 01/22/2023]
|
28
|
Doehl JSP, Sádlová J, Aslan H, Pružinová K, Metangmo S, Votýpka J, Kamhawi S, Volf P, Smith DF. Leishmania HASP and SHERP Genes Are Required for In Vivo Differentiation, Parasite Transmission and Virulence Attenuation in the Host. PLoS Pathog 2017; 13:e1006130. [PMID: 28095465 PMCID: PMC5271408 DOI: 10.1371/journal.ppat.1006130] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/27/2017] [Accepted: 12/15/2016] [Indexed: 12/15/2022] Open
Abstract
Differentiation of extracellular Leishmania promastigotes within their sand fly vector, termed metacyclogenesis, is considered to be essential for parasites to regain mammalian host infectivity. Metacyclogenesis is accompanied by changes in the local parasite environment, including secretion of complex glycoconjugates within the promastigote secretory gel and colonization and degradation of the sand fly stomodeal valve. Deletion of the stage-regulated HASP and SHERP genes on chromosome 23 of Leishmania major is known to stall metacyclogenesis in the sand fly but not in in vitro culture. Here, parasite mutants deficient in specific genes within the HASP/SHERP chromosomal region have been used to investigate their role in metacyclogenesis, parasite transmission and establishment of infection. Metacyclogenesis was stalled in HASP/SHERP mutants in vivo and, although still capable of osmotaxis, these mutants failed to secrete promastigote secretory gel, correlating with a lack of parasite accumulation in the thoracic midgut and failure to colonise the stomodeal valve. These defects prevented parasite transmission to a new mammalian host. Sand fly midgut homogenates modulated parasite behaviour in vitro, suggesting a role for molecular interactions between parasite and vector in Leishmania development within the sand fly. For the first time, stage-regulated expression of the small HASPA proteins in Leishmania (Leishmania) has been demonstrated: HASPA2 is expressed only in extracellular promastigotes and HASPA1 only in intracellular amastigotes. Despite its lack of expression in amastigotes, replacement of HASPA2 into the null locus background delays onset of pathology in BALB/c mice. This HASPA2-dependent effect is reversed by HASPA1 gene addition, suggesting that the HASPAs may have a role in host immunomodulation.
Collapse
Affiliation(s)
- Johannes S. P. Doehl
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| | - Jovana Sádlová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Hamide Aslan
- 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
| | - Kateřina Pružinová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Sonia Metangmo
- 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
| | - Jan Votýpka
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - 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
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Deborah F. Smith
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| |
Collapse
|
29
|
Increased Abundance of Proteins Involved in Resistance to Oxidative and Nitrosative Stress at the Last Stages of Growth and Development of Leishmania amazonensis Promastigotes Revealed by Proteome Analysis. PLoS One 2016; 11:e0164344. [PMID: 27776144 PMCID: PMC5077082 DOI: 10.1371/journal.pone.0164344] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 09/23/2016] [Indexed: 12/23/2022] Open
Abstract
Leishmania amazonensis is one of the major etiological agents of the neglected, stigmatizing disease termed american cutaneous leishmaniasis (ACL). ACL is a zoonosis and rodents are the main reservoirs. Most cases of ACL are reported in Brazil, Bolivia, Colombia and Peru. The biological cycle of the parasite is digenetic because sand fly vectors transmit the motile promastigote stage to the mammalian host dermis during blood meal intakes. The amastigote stage survives within phagocytes of the mammalian host. The purpose of this study is detection and identification of changes in protein abundance by 2DE/MALDI-TOF/TOF at the main growth phases of L. amazonensis promastigotes in axenic culture and the differentiation process that takes place simultaneously. The average number of proteins detected per gel is 202 and the non-redundant cumulative number is 339. Of those, 63 are differentially abundant throughout growth and simultaneous differentiation of L. amazonensis promastigotes. The main finding is that certain proteins involved in resistance to nitrosative and oxidative stress are more abundant at the last stages of growth and differentiation of cultured L. amazonensis promastigotes. These proteins are the arginase, a light variant of the tryparedoxin peroxidase, the iron superoxide dismutase, the regulatory subunit of the protein kinase A and a light HSP70 variant. These data taken together with the decrease of the stress-inducible protein 1 levels are additional evidence supporting the previously described pre-adaptative hypothesis, which consists of preparation in advance towards the amastigote stage.
Collapse
|
30
|
Alcolea PJ, Tuñón GIL, Alonso A, García-Tabares F, Ciordia S, Mena MC, Campos RNS, Almeida RP, Larraga V. Differential protein abundance in promastigotes of nitric oxide-sensitive and resistantLeishmania chagasistrains. Proteomics Clin Appl 2016; 10:1132-1146. [DOI: 10.1002/prca.201600054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 08/24/2016] [Accepted: 08/31/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Pedro J. Alcolea
- Department of Molecular Microbiology and Biology of Infections and Service of Proteomics and Genomics; Centro de Investigaciones Biológicas (Consejo Superior de Investigaciones Científicas); Madrid Spain
| | - Gabriel I. L. Tuñón
- Department of Morphology; Universidade Federal de Sergipe; São Cristóvão Sergipe Brazil
| | - Ana Alonso
- Department of Molecular Microbiology and Biology of Infections and Service of Proteomics and Genomics; Centro de Investigaciones Biológicas (Consejo Superior de Investigaciones Científicas); Madrid Spain
| | - Francisco García-Tabares
- Department of Molecular Microbiology and Biology of Infections and Service of Proteomics and Genomics; Centro de Investigaciones Biológicas (Consejo Superior de Investigaciones Científicas); Madrid Spain
| | - Sergio Ciordia
- Proteomics Unit; Centro Nacional de Biotecnología (Consejo Superior de Investigaciones Científicas); Madrid Spain
| | - María C. Mena
- Proteomics Unit; Centro Nacional de Biotecnología (Consejo Superior de Investigaciones Científicas); Madrid Spain
| | - Roseane N. S. Campos
- Department of Morphology; Universidade Federal de Sergipe; São Cristóvão Sergipe Brazil
| | - Roque P. Almeida
- Department of Medicine; Universidade Federal de Sergipe; Aracaju Sergipe Brazil
| | - Vicente Larraga
- Department of Molecular Microbiology and Biology of Infections and Service of Proteomics and Genomics; Centro de Investigaciones Biológicas (Consejo Superior de Investigaciones Científicas); Madrid Spain
| |
Collapse
|
31
|
Alcolea PJ, Alonso A, Degayón MA, Moreno-Paz M, Jiménez M, Molina R, Larraga V. In vitro infectivity and differential gene expression of Leishmania infantum metacyclic promastigotes: negative selection with peanut agglutinin in culture versus isolation from the stomodeal valve of Phlebotomus perniciosus. BMC Genomics 2016; 17:375. [PMID: 27206922 PMCID: PMC4874012 DOI: 10.1186/s12864-016-2672-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 04/26/2016] [Indexed: 11/10/2022] Open
Abstract
Background Leishmania infantum is the protozoan parasite responsible for zoonotic visceral leishmaniasis in the Mediterranean basin. A recent outbreak in humans has been reported in this area. The life cycle of the parasite is digenetic. The promastigote stage develops within the gut of phlebotomine sand flies, whereas amastigotes survive and multiply within phagolysosomes of mammalian host phagocytes. The major vector of L. infantum in Spain is Phlebotomus perniciosus. The axenic culture model of promastigotes is generally used because it is able to mimic the conditions of the natural environment (i.e. the sand fly vector gut). However, infectivity decreases with culture passages and infection of laboratory animals is frequently required. Enrichment of the stationary phase population in highly infective metacyclic promastigotes is achieved by negative selection with peanut agglutinin (PNA), which is possible only in certain Leishmania species such as L. major and L. infantum. In this study, in vitro infectivity and differential gene expression of cultured PNA-negative promastigotes (Pro-PNA−) and metacyclic promastigotes isolated from the sand fly anterior thoracic midgut (Pro-Pper) have been compared. Results In vitro infectivity is about 30 % higher in terms of rate of infected cells and number of amastigotes per infected cell in Pro-Pper than in Pro-PNA−. This finding is in agreement with up-regulation of a leishmanolysin gene (gp63) and genes involved in biosynthesis of glycosylinositolphospholipids (GIPL), lipophosphoglycan (LPG) and proteophosphoglycan (PPG) in Pro-Pper. In addition, differences between Pro-Pper and Pro-PNA− in genes involved in important cellular processes (e.g. signaling and regulation of gene expression) have been found. Conclusions Pro-Pper are significantly more infective than peanut lectin non-agglutinating ones. Therefore, negative selection with PNA is an appropriate method for isolating metacyclic promastigotes in stationary phase of axenic culture but it does not allow reaching the in vitro infectivity levels of Pro-Pper. Indeed, GIPL, LPG and PPG biosynthetic genes together with a gp63 gene are up-regulated in Pro-Pper and interestingly, the correlation coefficient between both transcriptomes in terms of transcript abundance is R2 = 0.68. This means that the correlation is sufficiently high to consider that both samples are physiologically comparable (i.e. the experiment was correctly designed and performed) and sufficiently low to conclude that important differences in transcript abundance have been found. Therefore, the implications of axenic culture should be evaluated case-by-case in each experimental design even when the stationary phase population in culture is enriched in metacyclic promastigotes by negative selection with PNA. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2672-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pedro J Alcolea
- Laboratorio de Parasitología Molecular, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas (Consejo Superior de Investigaciones Científicas), calle Ramiro de Maeztu, 9, 28040, Madrid, Spain.
| | - Ana Alonso
- Laboratorio de Parasitología Molecular, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas (Consejo Superior de Investigaciones Científicas), calle Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - María A Degayón
- Laboratorio de Parasitología Molecular, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas (Consejo Superior de Investigaciones Científicas), calle Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - Mercedes Moreno-Paz
- Laboratorio de Ecología Molecular, Centro de Astrobiología, (Instituto Nacional de Técnica Aeroespacial "Esteban Terradas"-Consejo Superior de Investigaciones Científicas), ctra. de Ajalvir Km 4, 28850, Torrejón de Ardoz, Madrid, Spain
| | - Maribel Jiménez
- Unidad de Entomología Médica, Servicio de Parasitología, Centro Nacional de Microbiología, Virología e Inmunología Sanitarias (Instituto de Salud Carlos III), ctra. Majadahonda-Pozuelo s/n, 28220, Majadahonda, Madrid, Spain
| | - Ricardo Molina
- Unidad de Entomología Médica, Servicio de Parasitología, Centro Nacional de Microbiología, Virología e Inmunología Sanitarias (Instituto de Salud Carlos III), ctra. Majadahonda-Pozuelo s/n, 28220, Majadahonda, Madrid, Spain
| | - Vicente Larraga
- Laboratorio de Parasitología Molecular, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas (Consejo Superior de Investigaciones Científicas), calle Ramiro de Maeztu, 9, 28040, Madrid, Spain
| |
Collapse
|
32
|
Rasid O, Mériaux V, Khan EM, Borde C, Ciulean IS, Fitting C, Manoury B, Cavaillon JM, Doyen N. Cathepsin B-Deficient Mice Resolve Leishmania major Inflammation Faster in a T Cell-Dependent Manner. PLoS Negl Trop Dis 2016; 10:e0004716. [PMID: 27182703 PMCID: PMC4868322 DOI: 10.1371/journal.pntd.0004716] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/27/2016] [Indexed: 11/18/2022] Open
Abstract
A critical role for intracellular TLR9 has been described in recognition and host resistance to Leishmania parasites. As TLR9 requires endolysosomal proteolytic cleavage to achieve signaling functionality, we investigated the contribution of different proteases like asparagine endopeptidase (AEP) or cysteine protease cathepsins B (CatB), L (CatL) and S (CatS) to host resistance during Leishmania major (L. major) infection in C57BL/6 (WT) mice and whether they would impact on TLR9 signaling. Unlike TLR9-/-, which are more susceptible to infection, AEP-/-, CatL-/- and CatS-/- mice are as resistant to L. major infection as WT mice, suggesting that these proteases are not individually involved in TLR9 processing. Interestingly, we observed that CatB-/- mice resolve L. major lesions significantly faster than WT mice, however we did not find evidence for an involvement of CatB on either TLR9-dependent or independent cytokine responses of dendritic cells and macrophages or in the innate immune response to L. major infection. We also found no difference in antigen presenting capacity. We observed a more precocious development of T helper 1 responses accompanied by a faster decline of inflammation, resulting in resolution of footpad inflammation, reduced IFNγ levels and decreased parasite burden. Adoptive transfer experiments into alymphoid RAG2-/-γc-/- mice allowed us to identify CD3+ T cells as responsible for the immune advantage of CatB-/- mice towards L. major. In vitro data confirmed the T cell intrinsic differences between CatB-/- mice and WT. Our study brings forth a yet unappreciated role for CatB in regulating T cell responses during L. major infection.
Collapse
Affiliation(s)
- Orhan Rasid
- Institut Pasteur, Département Infection et Epidémiologie, Unité Cytokines & Inflammation, Paris, France
- * E-mail: (OR); (ND)
| | - Véronique Mériaux
- Institut Pasteur, Département Infection et Epidémiologie, Unité Cytokines & Inflammation, Paris, France
| | - Erin M. Khan
- Institut Pasteur, Département Infection et Epidémiologie, Unité Cytokines & Inflammation, Paris, France
| | - Chloé Borde
- Institut Pasteur, Département Infection et Epidémiologie, Unité Cytokines & Inflammation, Paris, France
| | - Ioana S. Ciulean
- Institut Pasteur, Département Infection et Epidémiologie, Unité Cytokines & Inflammation, Paris, France
- Cantacuzino National Research Institute, Bucharest, Romania
| | - Catherine Fitting
- Institut Pasteur, Département Infection et Epidémiologie, Unité Cytokines & Inflammation, Paris, France
| | - Bénédicte Manoury
- Institut Necker Enfants Malades, INSERM U1151-CNRS UMR 8253, Hôpital Necker-Enfants Malades, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Jean-Marc Cavaillon
- Institut Pasteur, Département Infection et Epidémiologie, Unité Cytokines & Inflammation, Paris, France
| | - Noëlle Doyen
- Institut Pasteur, Département Infection et Epidémiologie, Unité Cytokines & Inflammation, Paris, France
- * E-mail: (OR); (ND)
| |
Collapse
|
33
|
Alcolea PJ, Alonso A, Domínguez M, Parro V, Jiménez M, Molina R, Larraga V. Influence of the Microenvironment in the Transcriptome of Leishmania infantum Promastigotes: Sand Fly versus Culture. PLoS Negl Trop Dis 2016; 10:e0004693. [PMID: 27163123 PMCID: PMC4862625 DOI: 10.1371/journal.pntd.0004693] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 04/14/2016] [Indexed: 01/04/2023] Open
Abstract
Zoonotic visceral leishmaniasis is a vector-borne disease caused by Leishmania infantum in the Mediterranean Basin, where domestic dogs and wild canids are the main reservoirs. The promastigote stage replicates and develops within the gut of blood-sucking phlebotomine sand flies. Mature promastigotes are injected in the dermis of the mammalian host and differentiate into the amastigote stage within parasitophorous vacuoles of phagocytic cells. The major vector of L. infantum in Spain is Phlebotomus perniciosus. Promastigotes are routinely axenized and cultured to mimic in vitro the conditions inside the insect gut, which allows for most molecular, cellular, immunological and therapeutical studies otherwise inviable. Culture passages are known to decrease infectivity, which is restored by passage through laboratory animals. The most appropriate source of promastigotes is the gut of the vector host but isolation of the parasite is technically challenging. In fact, this option is not viable unless small samples are sufficient for downstream applications like promastigote cultures and nucleic acid amplification. In this study, in vitro infectivity and differential gene expression have been studied in cultured promastigotes at the stationary phase and in promastigotes isolated from the stomodeal valve of the sand fly P. perniciosus. About 20 ng RNA per sample could be isolated. Each sample contained L. infantum promastigotes from 20 sand flies. RNA was successfully amplified and processed for shotgun genome microarray hybridization analysis. Most differentially regulated genes are involved in regulation of gene expression, intracellular signaling, amino acid metabolism and biosynthesis of surface molecules. Interestingly, meta-analysis by hierarchical clustering supports that up-regulation of 22.4% of the differentially regulated genes is specifically enhanced by the microenvironment (i.e. sand fly gut or culture). The correlation between cultured and naturally developed promastigotes is strong but not very high (Pearson coefficient R2 = 0.727). Therefore, the influence of promastigote culturing should be evaluated case-by-case in experimentation. The protozoan parasite Leishmania infantum causes visceral leishmaniasis in humans and is responsible for a recent outbreak reported in central Spain. Domestic dogs and wild canids are the main reservoirs. The life cycle of the parasite involves two stages and two hosts. The motile promastigote stage differentiates within the gut of the sand fly vector host and develops into non-motile amastigotes within phagocytes of the mammalian host. Promastigotes are routinely cultured in liquid media because it is assumed that they mimic the conditions within the gut of the insect. Therefore, the culture model is used in most studies about the biology of the parasite, pathogenesis and development of vaccines and new compounds for treatment. Isolating promastigotes from the natural microenvironment (i.e. the vector host) is desirable but technically challenging. We were able to perform a high-throughput analysis of gene expression thanks to mRNA amplification. The over-expressed genes detected may influence life cycle progression depending on the promastigote microenvironment (i.e. culture or vector host). Upcoming studies based on these results may reveal new therapeutic targets or vaccine candidates. Our results suggest that evaluating the influence of cultures in experimentation is convenient.
Collapse
Affiliation(s)
- Pedro J. Alcolea
- Laboratorio de Parasitología Molecular, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- * E-mail:
| | - Ana Alonso
- Laboratorio de Parasitología Molecular, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Mercedes Domínguez
- Unidad de Inmunología Microbiana, Centro Nacional de Microbiología, Virología e Inmunología Sanitarias, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Víctor Parro
- Laboratorio de Ecología Molecular, Centro de Astrobiología, Instituto Nacional de Técnica Aeroespacial “Esteban Terradas”—Consejo Superior de Investigaciones Científicas, Torrejón de Ardoz, Madrid, Spain
| | - Maribel Jiménez
- Unidad de Entomología Médica, Servicio de Parasitología, Centro Nacional de Microbiología, Virología e Inmunología Sanitarias, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Ricardo Molina
- Unidad de Entomología Médica, Servicio de Parasitología, Centro Nacional de Microbiología, Virología e Inmunología Sanitarias, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Vicente Larraga
- Laboratorio de Parasitología Molecular, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
34
|
Alcolea PJ, Alonso A, Moreno-Izquierdo MA, Degayón MA, Moreno I, Larraga V. Serum Removal from Culture Induces Growth Arrest, Ploidy Alteration, Decrease in Infectivity and Differential Expression of Crucial Genes in Leishmania infantum Promastigotes. PLoS One 2016; 11:e0150172. [PMID: 26959417 PMCID: PMC4784933 DOI: 10.1371/journal.pone.0150172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/10/2016] [Indexed: 11/19/2022] Open
Abstract
Leishmania infantum is one of the species responsible for visceral leishmaniasis. This species is distributed basically in the Mediterranean basin. A recent outbreak in humans has been reported in Spain. Axenic cultures are performed for most procedures with Leishmania spp. promastigotes. This model is stable and reproducible and mimics the conditions of the gut of the sand fly host, which is the natural environment of promastigote development. Culture media are undefined because they contain mammalian serum, which is a rich source of complex lipids and proteins. Serum deprivation slows down the growth kinetics and therefore, yield in biomass. In fact, we have confirmed that the growth rate decreases, as well as infectivity. Ploidy is also affected. Regarding the transcriptome, a high-throughput approach has revealed a low differential expression rate but important differentially regulated genes. The most remarkable profiles are: up-regulation of the GINS Psf3, the fatty acyl-CoA synthase (FAS1), the glyoxylase I (GLO1), the hydrophilic surface protein B (HASPB), the methylmalonyl-CoA epimerase (MMCE) and an amastin gene; and down-regulation of the gPEPCK and the arginase. Implications for metabolic adaptations, differentiation and infectivity are discussed herein.
Collapse
Affiliation(s)
- Pedro J. Alcolea
- Department of Molecular Microbiology and Biology of Infections, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB-CSIC), Madrid. Spain
- * E-mail:
| | - Ana Alonso
- Department of Molecular Microbiology and Biology of Infections, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB-CSIC), Madrid. Spain
| | - Miguel A. Moreno-Izquierdo
- Department of Molecular Microbiology and Biology of Infections, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB-CSIC), Madrid. Spain
| | - María A. Degayón
- Department of Molecular Microbiology and Biology of Infections, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB-CSIC), Madrid. Spain
| | - Inmaculada Moreno
- Unit of Microbial Immunology, Centro Nacional de Microbiología, Virología e Inmunología Sanitarias, Instituto de Salud Carlos III (CNM-ISCIII), Majadahonda, Spain
| | - Vicente Larraga
- Department of Molecular Microbiology and Biology of Infections, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB-CSIC), Madrid. Spain
| |
Collapse
|
35
|
Dillon LAL, Suresh R, Okrah K, Corrada Bravo H, Mosser DM, El-Sayed NM. Simultaneous transcriptional profiling of Leishmania major and its murine macrophage host cell reveals insights into host-pathogen interactions. BMC Genomics 2015; 16:1108. [PMID: 26715493 PMCID: PMC4696162 DOI: 10.1186/s12864-015-2237-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/24/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Parasites of the genus Leishmania are the causative agents of leishmaniasis, a group of diseases that range in manifestations from skin lesions to fatal visceral disease. The life cycle of Leishmania parasites is split between its insect vector and its mammalian host, where it resides primarily inside of macrophages. Once intracellular, Leishmania parasites must evade or deactivate the host's innate and adaptive immune responses in order to survive and replicate. RESULTS We performed transcriptome profiling using RNA-seq to simultaneously identify global changes in murine macrophage and L. major gene expression as the parasite entered and persisted within murine macrophages during the first 72 h of an infection. Differential gene expression, pathway, and gene ontology analyses enabled us to identify modulations in host and parasite responses during an infection. The most substantial and dynamic gene expression responses by both macrophage and parasite were observed during early infection. Murine genes related to both pro- and anti-inflammatory immune responses and glycolysis were substantially upregulated and genes related to lipid metabolism, biogenesis, and Fc gamma receptor-mediated phagocytosis were downregulated. Upregulated parasite genes included those aimed at mitigating the effects of an oxidative response by the host immune system while downregulated genes were related to translation, cell signaling, fatty acid biosynthesis, and flagellum structure. CONCLUSIONS The gene expression patterns identified in this work yield signatures that characterize multiple developmental stages of L. major parasites and the coordinated response of Leishmania-infected macrophages in the real-time setting of a dual biological system. This comprehensive dataset offers a clearer and more sensitive picture of the interplay between host and parasite during intracellular infection, providing additional insights into how pathogens are able to evade host defenses and modulate the biological functions of the cell in order to survive in the mammalian environment.
Collapse
Affiliation(s)
- Laura A L Dillon
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, 20742, USA. .,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, 20742, USA.
| | - Rahul Suresh
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, 20742, USA.
| | - Kwame Okrah
- Department of Mathematics, University of Maryland, College Park, MD, 20742, USA.
| | - Hector Corrada Bravo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, 20742, USA. .,Department of Computer Science, University of Maryland, College Park, MD, 20742, USA.
| | - David M Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, 20742, USA.
| | - Najib M El-Sayed
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, 20742, USA. .,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, 20742, USA. .,Present Address: 3128 Bioscience Research Bldg., University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
36
|
di Pietro M, Bird-Lieberman EL, Liu X, Nuckcheddy-Grant T, Bertani H, O'Donovan M, Fitzgerald RC. Autofluorescence-Directed Confocal Endomicroscopy in Combination With a Three-Biomarker Panel Can Inform Management Decisions in Barrett's Esophagus. Am J Gastroenterol 2015; 110:1549-58. [PMID: 26416188 DOI: 10.1038/ajg.2015.295] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 07/16/2015] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Barrett's esophagus (BE) surveillance with white-light endoscopy and quadrantic biopsies (Seattle protocol) is resource intensive and limited by sampling error. Previous work suggests that autofluorescence imaging (AFI) in combination with a molecular panel might reduce the number of biopsies, but this was not sufficiently sensitive for low-grade dysplasia, now a point for endoscopic intervention. Here we used AFI to direct narrow-field imaging tools for real-time optical assessment of dysplasia and biopsies for a biomarker panel. We compared the new diagnostic algorithm with the current standard. METHODS A total of 55 patients with BE were recruited at a single tertiary referral center. Patients underwent high-resolution endoscopy followed by AFI. AFI-targeted areas (n=194) were examined in turn by narrow-band imaging with magnification (NBIz) and probe-based confocal laser endomicroscopy (pCLE). Biopsies were taken from AFI-targeted areas and tested using an established molecular panel comprising aneuploidy plus cyclin A and p53 immunohistochemistry. RESULTS In the per-patient analysis the overall sensitivity and specificity of AFI-targeted pCLE were 100% and 53.6% for high-grade dysplasia/intramucosal cancer and 96.4% and 74.1% for any grade of dysplasia, respectively. NBIz had equal specificity for dysplasia detection (74.1%), but significantly lower sensitivity (57.1%) than pCLE. The time required to perform AFI-targeted pCLE was shorter that that taken by the Seattle protocol (P=0.0004). We found enrichment of molecular abnormalities in areas with optical dysplasia by pCLE (P<0.001), regardless of histologic dysplasia. The addition of the 3-biomarker panel reduced the false positive rate of pCLE by 50%, leading to sensitivity and specificity for any grade of dysplasia of 89.2% and 88.9%, respectively. CONCLUSIONS The combination of pCLE on AFI-targeted areas and a 3-biomarker panel identifies patients with dysplasia.
Collapse
Affiliation(s)
| | - Elizabeth L Bird-Lieberman
- MRC Cancer Unit, University of Cambridge, Cambridge, UK.,Translational Gastroenterology Unit, Experimental Medicine Division, John Radcliffe Hospital, Oxford, UK
| | - Xinxue Liu
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
| | | | - Helga Bertani
- Department of Digestive Endoscopy, Nuovo Ospedale Civile S. Agostino, Modena, Italy
| | - Maria O'Donovan
- Department of Histopathology, Cambridge University Hospitals, Cambridge, UK
| | | |
Collapse
|
37
|
Westrop GD, Williams RAM, Wang L, Zhang T, Watson DG, Silva AM, Coombs GH. Metabolomic Analyses of Leishmania Reveal Multiple Species Differences and Large Differences in Amino Acid Metabolism. PLoS One 2015; 10:e0136891. [PMID: 26368322 PMCID: PMC4569581 DOI: 10.1371/journal.pone.0136891] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/09/2015] [Indexed: 01/09/2023] Open
Abstract
Comparative genomic analyses of Leishmania species have revealed relatively minor heterogeneity amongst recognised housekeeping genes and yet the species cause distinct infections and pathogenesis in their mammalian hosts. To gain greater information on the biochemical variation between species, and insights into possible metabolic mechanisms underpinning visceral and cutaneous leishmaniasis, we have undertaken in this study a comparative analysis of the metabolomes of promastigotes of L. donovani, L. major and L. mexicana. The analysis revealed 64 metabolites with confirmed identity differing 3-fold or more between the cell extracts of species, with 161 putatively identified metabolites differing similarly. Analysis of the media from cultures revealed an at least 3-fold difference in use or excretion of 43 metabolites of confirmed identity and 87 putatively identified metabolites that differed to a similar extent. Strikingly large differences were detected in their extent of amino acid use and metabolism, especially for tryptophan, aspartate, arginine and proline. Major pathways of tryptophan and arginine catabolism were shown to be to indole-3-lactate and arginic acid, respectively, which were excreted. The data presented provide clear evidence on the value of global metabolomic analyses in detecting species-specific metabolic features, thus application of this technology should be a major contributor to gaining greater understanding of how pathogens are adapted to infecting their hosts.
Collapse
Affiliation(s)
- Gareth D. Westrop
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Roderick A. M. Williams
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
- Institute of Biomedical and Environmental Health Research, University of the West of Scotland, Paisley, United Kingdom
| | - Lijie Wang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Tong Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - David G. Watson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Ana Marta Silva
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Graham H. Coombs
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
Dillon LAL, Okrah K, Hughitt VK, Suresh R, Li Y, Fernandes MC, Belew AT, Corrada Bravo H, Mosser DM, El-Sayed NM. Transcriptomic profiling of gene expression and RNA processing during Leishmania major differentiation. Nucleic Acids Res 2015; 43:6799-813. [PMID: 26150419 PMCID: PMC4538839 DOI: 10.1093/nar/gkv656] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Protozoan parasites of the genus Leishmania are the etiological agents of leishmaniasis, a group of diseases with a worldwide incidence of 0.9–1.6 million cases per year. We used RNA-seq to conduct a high-resolution transcriptomic analysis of the global changes in gene expression and RNA processing events that occur as L. major transforms from non-infective procyclic promastigotes to infective metacyclic promastigotes. Careful statistical analysis across multiple biological replicates and the removal of batch effects provided a high quality framework for comprehensively analyzing differential gene expression and transcriptome remodeling in this pathogen as it acquires its infectivity. We also identified precise 5′ and 3′ UTR boundaries for a majority of Leishmania genes and detected widespread alternative trans-splicing and polyadenylation. An investigation of possible correlations between stage-specific preferential trans-splicing or polyadenylation sites and differentially expressed genes revealed a lack of systematic association, establishing that differences in expression levels cannot be attributed to stage-regulated alternative RNA processing. Our findings build on and improve existing expression datasets and provide a substantially more detailed view of L. major biology that will inform the field and potentially provide a stronger basis for drug discovery and vaccine development efforts.
Collapse
Affiliation(s)
- Laura A L Dillon
- Department of Cell Biology and Molecular Genetics, 3128 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Kwame Okrah
- Department of Mathematics, University of Maryland, College Park, MD 20742, USA
| | - V Keith Hughitt
- Department of Cell Biology and Molecular Genetics, 3128 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Rahul Suresh
- Department of Cell Biology and Molecular Genetics, 3128 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA
| | - Yuan Li
- Department of Cell Biology and Molecular Genetics, 3128 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Maria Cecilia Fernandes
- Department of Cell Biology and Molecular Genetics, 3128 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - A Trey Belew
- Department of Cell Biology and Molecular Genetics, 3128 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Hector Corrada Bravo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - David M Mosser
- Department of Cell Biology and Molecular Genetics, 3128 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA
| | - Najib M El-Sayed
- Department of Cell Biology and Molecular Genetics, 3128 Bioscience Research Building, University of Maryland, College Park, MD 20742, USA Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
39
|
Daviel C, Carter PM, Nation CS, Pizarro JC, Guidry J, Aiyar A, Kelly BL. LACK, a RACK1 ortholog, facilitates cytochrome c oxidase subunit expression to promote Leishmania major fitness. Mol Microbiol 2015; 96:95-109. [PMID: 25582232 PMCID: PMC6055511 DOI: 10.1111/mmi.12924] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2015] [Indexed: 12/22/2022]
Abstract
Leishmania are kinetoplastid parasites that cause the sandfly-transmitted disease leishmaniasis. To maintain fitness throughout their infectious life cycle, Leishmania must undergo rapid metabolic adaptations to the dramatically distinct environments encountered during transition between sandfly and vertebrate hosts. We performed proteomic and immunoblot analyses of attenuated L. major strains deficient for LACK, the Leishmania ortholog of the mammalian receptor for activated c kinase (RACK1), that is important for parasite thermotolerance and virulence. This approach identified cytochrome c oxidase (LmCOX) subunit IV as a LACK-dependent fitness protein. Consistent with decreased levels of LmCOX subunit IV at mammalian temperature, and in amastigotes, LmCOX activity and mitochondrial function were also impaired in LACK-deficient L. major under these conditions. Importantly, overexpression of LmCOX subunit IV in LACK-deficient L. major restored thermotolerance and macrophage infectivity. Interestingly, overexpression of LmCOX subunit IV enhanced LmCOX subunit VI expression at mammalian temperature. Collectively, our data suggest LACK promotes Leishmania adaptation to the mammalian host environment by sustaining LmCOX subunit IV expression and hence energy metabolism in response to stress stimuli such as heat. These findings extend the repertoire of RACK1 protein utility to include a role in mitochondrial function.
Collapse
Affiliation(s)
- Cardenas Daviel
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Pamela M. Carter
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Catherine S. Nation
- Department of Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Juan C. Pizarro
- Department of Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Jessie Guidry
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ashok Aiyar
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ben L. Kelly
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
40
|
An Insight into the proteome of Crithidia fasciculata choanomastigotes as a comparative approach to axenic growth, peanut lectin agglutination and differentiation of Leishmania spp. promastigotes. PLoS One 2014; 9:e113837. [PMID: 25503511 PMCID: PMC4263474 DOI: 10.1371/journal.pone.0113837] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/31/2014] [Indexed: 01/31/2023] Open
Abstract
The life cycle of the trypanosomatid Crithidia fasciculata is monogenetic, as the unique hosts of these parasites are different species of culicids. The comparison of these non-pathogenic microorganisms evolutionary close to other species of trypanosomatids that develop digenetic life cycles and cause chronic severe sickness to millions of people worldwide is of outstanding interest. A ground-breaking analysis of differential protein abundance in Crithidia fasciculata is reported herein. The comparison of the outcome with previous gene expression profiling studies developed in the related human pathogens of the genus Leishmania has revealed substantial differences between the motile stages of these closely related organisms in abundance of proteins involved in catabolism, redox homeostasis, intracellular signalling, and gene expression regulation. As L. major and L. infantum agglutinate with peanut lectin and non-agglutinating parasites are more infective, the agglutination properties were evaluated in C. fasciculata. The result is that choanomastigotes are able to agglutinate with peanut lectin and a non-agglutinating subpopulation can be also isolated. As a difference with L. infantum, the non-agglutinating subpopulation over-expresses the whole machinery for maintenance of redox homeostasis and the translation factors eIF5a, EF1α and EF2, what suggests a relationship between the lack of agglutination and a differentiation process.
Collapse
|
41
|
Alcolea PJ, Alonso A, Gómez MJ, Postigo M, Molina R, Jiménez M, Larraga V. Stage-specific differential gene expression in Leishmania infantum: from the foregut of Phlebotomus perniciosus to the human phagocyte. BMC Genomics 2014; 15:849. [PMID: 25281593 PMCID: PMC4203910 DOI: 10.1186/1471-2164-15-849] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 09/19/2014] [Indexed: 12/16/2022] Open
Abstract
Background Leishmania infantum is the etiological agent of zoonotical visceral leishmaniasis in the Mediterranean basin. A recent outbreak in humans has been recently reported in central Spain. Leishmania spp. parasites are transmitted to the mammalian host by the bite of sand flies. The primary vector of L. infantum in Spain is Phlebotomus perniciosus. For decades, research on these parasites has involved the axenic culture model of the promastigote stage including gene expression profiling studies performed in the post-genome era. Unlike the controversial axenic culturing of amastigotes, promastigote cultures are generally accepted and used, although with the precaution of avoiding excessive culture passage. The primary objective of this differentiation study is to compare the gene expression profiles of promastigotes isolated from the foregut of the sand fly and amastigotes. For this purpose, P. perniciosus sand flies were infected with L. infantum and differentiated promastigotes were extracted by dissection of the foreguts. Shotgun DNA microarray hybridization analyses allowed for transcriptome comparison of these promastigotes with amastigotes obtained by infection of the U937 cell line. The results have been compared with those described in published expression analyses using axenic promastigotes. Results A total of 277 up-regulated genes were found through this hybridization experiment. The comparison of these particular results with published gene expression profile analyses performed using the same experimental procedure to study cultured promastigotes in stationary phase versus amastigotes revealed considerable differences (approximately 95% of the up-regulated genes were different). We found that the up-regulation rate is lower in amastigotes than in sand fly-derived promastigotes, which is in agreement with the over-expression of genes involved in gene expression regulation and signaling in those promastigote populations. Conclusions The up-regulation rate is lower in intracellular amastigotes than in promastigotes obtained from the sand fly gut. This was also reported by us using the promastigote culture model and is an evidence for the hypothesis of promastigote preadaptation towards life in the intracellular environment. Regarding transcript abundance, the set of differentially regulated genes is notably different when using promastigotes from the sand fly foregut instead of axenic cultures. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-849) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pedro J Alcolea
- Laboratorio de Parasitología Molecular, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Científicas, Calle Ramiro de Maeztu, 9, 28040 Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
42
|
Pawar H, Renuse S, Khobragade SN, Chavan S, Sathe G, Kumar P, Mahale KN, Gore K, Kulkarni A, Dixit T, Raju R, Prasad TSK, Harsha HC, Patole MS, Pandey A. Neglected Tropical Diseases and Omics Science: Proteogenomics Analysis of the Promastigote Stage ofLeishmania majorParasite. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2014; 18:499-512. [DOI: 10.1089/omi.2013.0159] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Harsh Pawar
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Rajiv Gandhi University of Health Sciences, Bangalore, India
| | - Santosh Renuse
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Department of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
| | | | - Sandip Chavan
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal University, Madhav Nagar, Manipal, India
| | - Gajanan Sathe
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal University, Madhav Nagar, Manipal, India
| | - Praveen Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | | | | | | | - Tanwi Dixit
- National Centre for Cell Sciences, Pune, India
| | - Rajesh Raju
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | | | - H. C. Harsha
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | | | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
43
|
Abstract
A decade of genome sequencing has transformed our understanding of how
trypanosomatid parasites have evolved and provided fresh impetus to explaining
the origins of parasitism in the Kinetoplastida. In this review, I will consider
the many ways in which genome sequences have influenced our view of genomic
reduction in trypanosomatids; how species-specific genes, and the genomic
domains they occupy, have illuminated the innovations in trypanosomatid genomes;
and how comparative genomics has exposed the molecular mechanisms responsible
for innovation and adaptation to a parasitic lifestyle.
Collapse
|
44
|
Hartley MA, Drexler S, Ronet C, Beverley SM, Fasel N. The immunological, environmental, and phylogenetic perpetrators of metastatic leishmaniasis. Trends Parasitol 2014; 30:412-22. [PMID: 24954794 DOI: 10.1016/j.pt.2014.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 05/23/2014] [Accepted: 05/24/2014] [Indexed: 02/07/2023]
Abstract
Cutaneous leishmaniases have persisted for centuries as chronically disfiguring parasitic infections affecting millions of people across the subtropics. Symptoms range from the more prevalent single, self-healing cutaneous lesion to a persistent, metastatic disease, where ulcerations and granulomatous nodules can affect multiple secondary sites of the skin and delicate facial mucosa, even sometimes diffusing throughout the cutaneous system as a papular rash. The basis for such diverse pathologies is multifactorial, ranging from parasite phylogeny to host immunocompetence and various environmental factors. Although complex, these pathologies often prey on weaknesses in the innate immune system and its pattern recognition receptors. This review explores the observed and potential associations among the multifactorial perpetrators of infectious metastasis and components of the innate immune system.
Collapse
Affiliation(s)
- Mary-Anne Hartley
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Stefan Drexler
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Catherine Ronet
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Stephen M Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.
| |
Collapse
|
45
|
de Jesus JB, Mesquita-Rodrigues C, Cuervo P. Proteomics advances in the study of Leishmania parasites and leishmaniasis. Subcell Biochem 2014; 74:323-349. [PMID: 24264252 DOI: 10.1007/978-94-007-7305-9_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Leishmania spp. are digenetic parasites which cause a broad spectrum of fatal diseases in humans. These parasites, as well as the other trypanosomatid, regulate gene expression at the post-transcriptional and post-translational levels, so that a poor correlation is observed between mRNA content and translated proteins. The completion of the genomic sequencing of several Leishmania species has enormous relevance to the study of the leishmaniasis pathogenesis. The combination of the available genomic resources of these parasites with powerful high-throughput proteomic analysis has shed light on various aspects of Leishmania biology as well as on the mechanisms underlying the disease. Diverse proteomic approaches have been used to describe and catalogue global protein profiles of Leishmania spp., reveal changes in protein expression during development, determine the subcellular localization of gene products, evaluate host-parasite interactions and elucidate drug resistance mechanisms. The characterization of these proteins has advanced, although many fundamental questions remain unanswered. Here, we present a historic review summarizing the different proteomic technologies applied to the study of Leishmania parasites during the last decades and we discuss the proteomic discoveries that have contributed to the understanding of Leishmania parasites biology and leishmaniasis.
Collapse
Affiliation(s)
- Jose Batista de Jesus
- Departamento de Engenharia de Biossistemas, Universidade Federal de São João Del Rei, São João Del Rei, MG, Brazil,
| | | | | |
Collapse
|
46
|
Mittra B, Andrews NW. IRONy OF FATE: role of iron-mediated ROS in Leishmania differentiation. Trends Parasitol 2013; 29:489-96. [PMID: 23948431 DOI: 10.1016/j.pt.2013.07.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 07/16/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023]
Abstract
The protozoan parasite Leishmania experiences extreme environmental changes as it alternates between insect and mammalian hosts. In some species, differentiation of insect promastigotes into mammalian-infective amastigotes is induced by elevated temperature and low pH, conditions found within macrophage parasitophorous vacuoles (PVs). However, the signaling events controlling amastigote differentiation remain poorly understood. Recent studies revealed a novel role for iron uptake in orchestrating the differentiation of amastigotes, through a mechanism that involves production of reactive oxygen species (ROS) and is independent from pH and temperature changes. ROS are generally thought to be deleterious for pathogens, but it is becoming increasingly apparent that they can also function as signaling molecules regulating Leishmania differentiation, in a process that is tightly controlled by iron availability.
Collapse
Affiliation(s)
- Bidyottam Mittra
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | | |
Collapse
|
47
|
Goyal N. Novel approaches for the identification of inhibitors of leishmanial dipeptidylcarboxypeptidase. Expert Opin Drug Discov 2013; 8:1127-34. [PMID: 23745836 DOI: 10.1517/17460441.2013.807247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Leishmaniasis imposes a substantial burden of mortality and morbidity affecting 12 million globally and continues to be a neglected tropical disease. Control of the disease is mainly based on chemotherapy, which relies on a handful of drugs with serious limitations. Over the last decade, target-based drug discovery is also being employed in addition to the random screening of compounds. Leishmanial dipeptidylcarboxypeptidase (LDCP), an angiotensin converting enzyme (ACE) related metallopeptidase, has been recently identified as a novel drug target for antileishmanial chemotherapy. AREAS COVERED This article examines dipeptidylcarboxypeptidase (DCP) of Leishmania donovani and of other sources from the international literature regarding their biochemical and structural characterization in comparison to mammalian ACE. Furthermore, the author discusses the identification of LdDCP specific inhibitors by virtual screening and their effect on parasite multiplication. Finally, the review looks ahead at areas for further exploration of DCP inhibitors in Leishmania chemotherapy. EXPERT OPINION The first step in targeted screening is to identify a suitable drug target and its validation followed by its use in high throughput screening of compounds. Limited studies on LDCP inhibitors have established a good correlation between parasite enzyme inhibition and their biological activity. This suggests that there is a potential for LDCP inhibitors as new antileishmanial drugs.
Collapse
Affiliation(s)
- Neena Goyal
- CSIR-Central Drug Research Institute, Division of Biochemistry, Chattar Manzil Palace, PO Box 173, Lucknow-226001 (UP), India.
| |
Collapse
|
48
|
Smircich P, Forteza D, El-Sayed NM, Garat B. Genomic analysis of sequence-dependent DNA curvature in Leishmania. PLoS One 2013; 8:e63068. [PMID: 23646176 PMCID: PMC3639952 DOI: 10.1371/journal.pone.0063068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/27/2013] [Indexed: 11/26/2022] Open
Abstract
Leishmania major is a flagellated protozoan parasite of medical importance. Like other members of the Trypanosomatidae family, it possesses unique mechanisms of gene expression such as constitutive polycistronic transcription of directional gene clusters, gene amplification, mRNA trans-splicing, and extensive editing of mitochondrial transcripts. The molecular signals underlying most of these processes remain under investigation. In order to investigate the role of DNA secondary structure signals in gene expression, we carried out a genome-wide in silico analysis of the intrinsic DNA curvature. The L. major genome revealed a lower frequency of high intrinsic curvature regions as well as inter- and intra- chromosomal distribution heterogeneity, when compared to prokaryotic and eukaryotic organisms. Using a novel method aimed at detecting region-integrated intrinsic curvature (RIIC), high DNA curvature was found to be associated with regions implicated in transcription initiation. Those include divergent strand-switch regions between directional gene clusters and regions linked to markers of active transcription initiation such as acetylated H3 histone, TRF4 and SNAP50. These findings suggest a role for DNA curvature in transcription initiation in Leishmania supporting the relevance of DNA secondary structures signals.
Collapse
Affiliation(s)
- Pablo Smircich
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Montevideo, Uruguay
- Departamento de Genética, Facultad de Medicina, Montevideo, Uruguay
| | - Diego Forteza
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Montevideo, Uruguay
| | - Najib M. El-Sayed
- Department of Cell Biology and Molecular Genetics and Center for Bioinformatics and Computational Biology, University of Maryland College Park, Maryland, United States of America
| | - Beatriz Garat
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Montevideo, Uruguay
| |
Collapse
|
49
|
Mittra B, Cortez M, Haydock A, Ramasamy G, Myler PJ, Andrews NW. Iron uptake controls the generation of Leishmania infective forms through regulation of ROS levels. ACTA ACUST UNITED AC 2013; 210:401-16. [PMID: 23382545 PMCID: PMC3570109 DOI: 10.1084/jem.20121368] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
During its life cycle, Leishmania undergoes extreme environmental changes, alternating between insect vectors and vertebrate hosts. Elevated temperature and decreased pH, conditions encountered after macrophage invasion, can induce axenic differentiation of avirulent promastigotes into virulent amastigotes. Here we show that iron uptake is a major trigger for the differentiation of Leishmania amazonensis amastigotes, independently of temperature and pH changes. We found that iron depletion from the culture medium triggered expression of the ferrous iron transporter LIT1 (Leishmania iron transporter 1), an increase in iron content of the parasites, growth arrest, and differentiation of wild-type (WT) promastigotes into infective amastigotes. In contrast, LIT1-null promastigotes showed reduced intracellular iron content and sustained growth in iron-poor media, followed by cell death. LIT1 up-regulation also increased iron superoxide dismutase (FeSOD) activity in WT but not in LIT1-null parasites. Notably, the superoxide-generating drug menadione or H(2)O(2) was sufficient to trigger differentiation of WT promastigotes into fully infective amastigotes. LIT1-null promastigotes accumulated superoxide radicals and initiated amastigote differentiation after exposure to H(2)O(2) but not to menadione. Our results reveal a novel role for FeSOD activity and reactive oxygen species in orchestrating the differentiation of virulent Leishmania amastigotes in a process regulated by iron availability.
Collapse
Affiliation(s)
- Bidyottam Mittra
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Leishmaniasis is a vector-borne neglected tropical disease associated with a spectrum of clinical manifestations, ranging from self-healing cutaneous lesions to fatal visceral infections. Among the most important questions in Leishmania research is why some species like L. donovani infect visceral organs, whereas other species like L. major remain in the skin. The determinants of visceral leishmaniasis are still poorly understood, although genomic, immunologic, and animal models are beginning to provide important insight into this disease. In this review, we discuss the vector, host, and pathogen factors that mediate the development of visceral leishmaniasis. We examine the progression of the parasite from the initial site of sand fly bite to the visceral organs and its ability to survive there. The identification of visceral disease determinants is required to understand disease evolution, to understand visceral organ survival mechanisms, and potentially to develop better interventions for this largely neglected disease.
Collapse
|