1
|
Ansari I, Basak R, Mukhopadhyay A. Hemoglobin Endocytosis and Intracellular Trafficking: A Novel Way of Heme Acquisition by Leishmania. Pathogens 2022; 11:585. [PMID: 35631106 PMCID: PMC9143042 DOI: 10.3390/pathogens11050585] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Leishmania species are causative agents of human leishmaniasis, affecting 12 million people annually. Drugs available for leishmaniasis are toxic, and no vaccine is available. Thus, the major thrust is to identify new therapeutic targets. Leishmania is an auxotroph for heme and must acquire heme from the host for its survival. Thus, the major focus has been to understand the heme acquisition process by the parasites in the last few decades. It is conceivable that the parasite is possibly obtaining heme from host hemoprotein, as free heme is not available in the host. Current understanding indicates that Leishmania internalizes hemoglobin (Hb) through a specific receptor by a clathrin-mediated endocytic process and targets it to the parasite lysosomes via the Rab5 and Rab7 regulated endocytic pathway, where it is degraded to generate intracellular heme that is used by the parasite. Subsequently, intra-lysosomal heme is initially transported to the cytosol and is finally delivered to the mitochondria via different heme transporters. Studies using different null mutant parasites showed that these receptors and transporters are essential for the survival of the parasite. Thus, the heme acquisition process in Leishmania may be exploited for the development of novel therapeutics.
Collapse
Affiliation(s)
| | | | - Amitabha Mukhopadhyay
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India; (I.A.); (R.B.)
| |
Collapse
|
2
|
Carneiro MB, Peters NC. The Paradox of a Phagosomal Lifestyle: How Innate Host Cell- Leishmania amazonensis Interactions Lead to a Progressive Chronic Disease. Front Immunol 2021; 12:728848. [PMID: 34557194 PMCID: PMC8452962 DOI: 10.3389/fimmu.2021.728848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Intracellular phagosomal pathogens represent a formidable challenge for innate immune cells, as, paradoxically, these phagocytic cells can act as both host cells that support pathogen replication and, when properly activated, are the critical cells that mediate pathogen elimination. Infection by parasites of the Leishmania genus provides an excellent model organism to investigate this complex host-pathogen interaction. In this review we focus on the dynamics of Leishmania amazonensis infection and the host innate immune response, including the impact of the adaptive immune response on phagocytic host cell recruitment and activation. L. amazonensis infection represents an important public health problem in South America where, distinct from other Leishmania parasites, it has been associated with all three clinical forms of leishmaniasis in humans: cutaneous, muco-cutaneous and visceral. Experimental observations demonstrate that most experimental mouse strains are susceptible to L. amazonensis infection, including the C57BL/6 mouse, which is resistant to other species such as Leishmania major, Leishmania braziliensis and Leishmania infantum. In general, the CD4+ T helper (Th)1/Th2 paradigm does not sufficiently explain the progressive chronic disease established by L. amazonensis, as strong cell-mediated Th1 immunity, or a lack of Th2 immunity, does not provide protection as would be predicted. Recent findings in which the balance between Th1/Th2 immunity was found to influence permissive host cell availability via recruitment of inflammatory monocytes has also added to the complexity of the Th1/Th2 paradigm. In this review we discuss the roles played by innate cells starting from parasite recognition through to priming of the adaptive immune response. We highlight the relative importance of neutrophils, monocytes, dendritic cells and resident macrophages for the establishment and progressive nature of disease following L. amazonensis infection.
Collapse
Affiliation(s)
- Matheus B Carneiro
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Nathan C Peters
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
3
|
Zhang KY, Yuan WJ, Xu JD, Wang JX. Cation-dependent mannose-6-phosphate receptor functions as a pattern recognition receptor in anti-bacterial immunity of Marsupenaeus japonicus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 89:122-130. [PMID: 30118735 DOI: 10.1016/j.dci.2018.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/12/2018] [Accepted: 08/14/2018] [Indexed: 06/08/2023]
Abstract
The cation-dependent mannose-6-phosphate receptor (CD-MPR) is a member of the P-type lectin family. As a type I transmembrane glycoprotein, it functions in the delivery of newly synthesized acid hydrolases from the trans-Golgi network to endosomes for their subsequent transfer to the lysosome by binding the mannose-6-phosphate receptor-recognition moieties in the hydrolases. However, the functions of CD-MPR in immune responses are seldom reported. In the present study, we identified a CD-MPR-like molecule in Marsupenaeus japonicus and designed it as MjCD-MPR. It was significantly upregulated after challenge with Vibrio anguillarum at the mRNA and protein levels. Knockdown of MjCD-MPR resulted in a significant increase in the amount of V. anguillarum in the hemolymph of shrimp, which suggested that MjCD-MPR plays a role in shrimp antibacterial defense. The recombinant extracytoplasmic region of MjCD-MPR could bind gram-positive and gram-negative bacteria by interaction with peptidoglycan, lipopolysaccharide, and lipoteichoic acid. MjCD-MPR showed no direct bacteriostatic or bacteriocidal activity. Knockdown of MjCD-MPR decreased the expression levels of several antimicrobial peptides (Alf-C1, Alf-E1, Crustin I-2, and Crustin I-3), suggesting that MjCD-MPR promotes the expression of antimicrobial peptides in shrimp. In summary, working as a pattern recognition receptor, MjCD-MPR recognizes invading bacteria and triggers the expression of AMPs against bacterial infection in shrimp.
Collapse
Affiliation(s)
- Ke-Yi Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, 266237, Shandong, China
| | - Wen-Jie Yuan
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, 266237, Shandong, China
| | - Ji-Dong Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, 266237, Shandong, China
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, 266237, Shandong, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, China.
| |
Collapse
|
4
|
Dias BRS, de Souza CS, Almeida NDJ, Lima JGB, Fukutani KF, Dos Santos TBS, França-Cost J, Brodskyn CI, de Menezes JPB, Colombo MI, Veras PST. Autophagic Induction Greatly Enhances Leishmania major Intracellular Survival Compared to Leishmania amazonensis in CBA/j-Infected Macrophages. Front Microbiol 2018; 9:1890. [PMID: 30158914 PMCID: PMC6104192 DOI: 10.3389/fmicb.2018.01890] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 07/27/2018] [Indexed: 12/29/2022] Open
Abstract
CBA mouse macrophages control Leishmania major infection yet are permissive to Leishmania amazonensis. Few studies have been conducted to assess the role played by autophagy in Leishmania infection. Therefore, we assessed whether the autophagic response of infected macrophages may account for the differential behavior of these two parasite strains. After 24 h of infection, the LC3-II/Act ratio increased in both L. amazonensis- and L. major-infected macrophages compared to uninfected controls, but less than in chloroquine-treated cells. This suggests that L. amazonensis and L. major activate autophagy in infected macrophages, without altering the autophagic flux. Furthermore, L. major-infected cells exhibited higher percentages of DQ-BSA-labeled parasitophorous vacuoles (50%) than those infected by L. amazonensis (25%). However, L. major- and L. amazonensis-induced parasitophorous vacuoles accumulated LysoTracker similarly, indicating that the acidity in both compartment was equivalent. At as early as 30 min, endogenous LC3 was recruited to both L. amazonensis- and L. major-induced parasitophorous vacuoles, while after 24 h a greater percentage of LC3 positive vacuoles was observed in L. amazonensis-infected cells (42.36%) compared to those infected by L. major (18.10%). Noteworthy, principal component analysis (PCA) and an hierarchical cluster analysis completely discriminated L. major-infected macrophages from L. amazonensis-infected cells accordingly to infection intensity and autophagic features of parasite-induced vacuoles. Then, we evaluated whether the modulation of autophagy exerted an influence on parasite infection in macrophages. No significant changes were observed in both infection rate or parasite load in macrophages treated with the autophagic inhibitors wortmannin, chloroquine or VPS34-IN1, as well as with the autophagic inducers rapamycin or physiological starvation, in comparison to untreated control cells. Interestingly, both autophagic inducers enhanced intracellular L. amazonensis and L. major viability, while the pharmacological inhibition of autophagy exerted no effects on intracellular parasite viability. We also demonstrated that autophagy induction reduced NO production by L. amazonensis- and L. major-infected macrophages but not alters arginase activity. These findings provide evidence that although L. amazonensis-induced parasitophorous vacuoles recruit LC3 more markedly, L. amazonensis and L. major similarly activate the autophagic pathway in CBA macrophages. Interestingly, the exogenous induction of autophagy favors L. major intracellular viability to a greater extent than L. amazonensis related to a reduction in the levels of NO.
Collapse
Affiliation(s)
- Beatriz R S Dias
- Laboratory of Parasite-Host Interaction and Epidemiology, Gonçalo Moniz Institute, Salvador, Brazil
| | - Carina S de Souza
- Laboratory of Parasite-Host Interaction and Epidemiology, Gonçalo Moniz Institute, Salvador, Brazil
| | - Niara de Jesus Almeida
- Laboratory of Parasite-Host Interaction and Epidemiology, Gonçalo Moniz Institute, Salvador, Brazil
| | - José G B Lima
- Laboratory of Parasite-Host Interaction and Epidemiology, Gonçalo Moniz Institute, Salvador, Brazil
| | - Kiyoshi F Fukutani
- Laboratory of Inflammation and Biomarkers, Gonçalo Moniz Institute, Salvador, Brazil
| | - Thiale B S Dos Santos
- Laboratory of Inflammation and Biomarkers, Gonçalo Moniz Institute, Salvador, Brazil
| | - Jaqueline França-Cost
- Laboratory of Inflammation and Biomarkers, Gonçalo Moniz Institute, Salvador, Brazil.,Department of Biointeraction, Federal University of Bahia, Salvador, Brazil
| | - Claudia I Brodskyn
- Laboratory of Parasite-Host Interaction and Epidemiology, Gonçalo Moniz Institute, Salvador, Brazil
| | - Juliana P B de Menezes
- Laboratory of Parasite-Host Interaction and Epidemiology, Gonçalo Moniz Institute, Salvador, Brazil
| | - Maria I Colombo
- Laboratory of Cellular and Molecular Biology, Institute of Histology and Embryology-CONICET, National University of Cuyo, Mendoza, Argentina
| | - Patricia S T Veras
- Laboratory of Parasite-Host Interaction and Epidemiology, Gonçalo Moniz Institute, Salvador, Brazil
| |
Collapse
|
5
|
Živanović V, Semini G, Laue M, Drescher D, Aebischer T, Kneipp J. Chemical Mapping of Leishmania Infection in Live Cells by SERS Microscopy. Anal Chem 2018; 90:8154-8161. [DOI: 10.1021/acs.analchem.8b01451] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Vesna Živanović
- School of Analytical Sciences Adlershof, Humboldt-Universität zu Berlin, Albert-Einstein-Strasse 5-9, 12489 Berlin, Germany
| | | | | | | | | | - Janina Kneipp
- School of Analytical Sciences Adlershof, Humboldt-Universität zu Berlin, Albert-Einstein-Strasse 5-9, 12489 Berlin, Germany
| |
Collapse
|
6
|
Hernández-Chinea C, Maimone L, Campos Y, Mosca W, Romero PJ. Apparent isocitrate lyase activity in Leishmania amazonensis. Acta Parasitol 2017; 62:701-707. [PMID: 29035856 DOI: 10.1515/ap-2017-0084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 06/23/2017] [Indexed: 11/15/2022]
Abstract
Early reports have demonstrated the occurrence of glyoxylate cycle enzymes in several Leishmania species. However, these results have been underestimated because genes for the two key enzymes of the cycle, isocitrate lyase (ICL) and malate synthase (MS), are not annotated in Leishmania genomes. We have re-examined this issue in promastigotes of Leishmania amazonensis. Enzyme activities were assayed spectrophotometrically in cellular extracts and characterized partially. A 40 kDa band displaying ICL activity was visualized on zymograms of the extracts. By immunoblotting with mouse antibodies against ICL from Bacillus stearothermophilus, a band of approximately 40 kDa was identified, coincident with the relative molecular mass of the activity band revealed on zymograms. Indirect immunofluorescence of intact promastigotes showed that the recognized antigen is distributed as a punctuated pattern, mainly distributed beneath the subpellicular microtubules, over a diffused cytoplasmic stain. These results clearly demonstrate the existence of an apparent ICL activity in L. amazonensis promastigotes, which is associated to a 40 kDa polypeptide and distributed both diffused and as punctuate aggregates in the cytoplasm. The relevance of this activity is discussed.
Collapse
|
7
|
Semini G, Aebischer T. Phagosome proteomics to study Leishmania's intracellular niche in macrophages. Int J Med Microbiol 2017; 308:68-76. [PMID: 28927848 DOI: 10.1016/j.ijmm.2017.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/23/2017] [Accepted: 09/03/2017] [Indexed: 12/14/2022] Open
Abstract
Intracellular pathogens invade their host cells and replicate within specialized compartments. In turn, the host cell initiates a defensive response trying to kill the invasive agent. As a consequence, intracellular lifestyle implies morphological and physiological changes in both pathogen and host cell. Leishmania spp. are medically important intracellular protozoan parasites that are internalized by professional phagocytes such as macrophages, and reside within the parasitophorous vacuole inhibiting their microbicidal activity. Whereas the proteome of the extracellular promastigote form and the intracellular amastigote form have been extensively studied, the constituents of Leishmania's intracellular niche, an endolysosomal compartment, are not fully deciphered. In this review we discuss protocols to purify such compartments by means of an illustrating example to highlight generally relevant considerations and innovative aspects that allow purification of not only the intracellular parasites but also the phagosomes that harbor them and analyze the latter by gel free proteomics.
Collapse
Affiliation(s)
- Geo Semini
- Mycotic and Parasitic Agents and Mycobacteria, Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany.
| | - Toni Aebischer
- Mycotic and Parasitic Agents and Mycobacteria, Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
8
|
Semini G, Paape D, Paterou A, Schroeder J, Barrios‐Llerena M, Aebischer T. Changes to cholesterol trafficking in macrophages by Leishmania parasites infection. Microbiologyopen 2017; 6:e00469. [PMID: 28349644 PMCID: PMC5552908 DOI: 10.1002/mbo3.469] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 02/06/2017] [Accepted: 02/17/2017] [Indexed: 11/08/2022] Open
Abstract
Leishmania spp. are protozoan parasites that are transmitted by sandfly vectors during blood sucking to vertebrate hosts and cause a spectrum of diseases called leishmaniases. It has been demonstrated that host cholesterol plays an important role during Leishmania infection. Nevertheless, little is known about the intracellular distribution of this lipid early after internalization of the parasite. Here, pulse-chase experiments with radiolabeled cholesteryl esterified to fatty acids bound to low-density lipoproteins indicated that retention of this source of cholesterol is increased in parasite-containing subcellular fractions, while uptake is unaffected. This is correlated with a reduction or absence of detectable NPC1 (Niemann-Pick disease, type C1), a protein responsible for cholesterol efflux from endocytic compartments, in the Leishmania mexicana habitat and infected cells. Filipin staining revealed a halo around parasites within parasitophorous vacuoles (PV) likely representing free cholesterol accumulation. Labeling of host cell membranous cholesterol by fluorescent cholesterol species before infection revealed that this pool is also trafficked to the PV but becomes incorporated into the parasites' membranes and seems not to contribute to the halo detected by filipin. This cholesterol sequestration happened early after infection and was functionally significant as it correlated with the upregulation of mRNA-encoding proteins required for cholesterol biosynthesis. Thus, sequestration of cholesterol by Leishmania amastigotes early after infection provides a basis to understand perturbation of cholesterol-dependent processes in macrophages that were shown previously by others to be necessary for their proper function in innate and adaptive immune responses.
Collapse
Affiliation(s)
- Geo Semini
- Mycotic and Parasitic Agents and MycobacteriaDepartment of Infectious DiseasesRobert Koch‐InstituteBerlinGermany
| | - Daniel Paape
- Institute of Immunology and Infection ResearchThe University of EdinburghEdinburghUK
- Present address:
Welcome Trust Centre for Molecular Parasitology and Institute of Infection Immunity and InflammationCollege of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - Athina Paterou
- Institute of Immunology and Infection ResearchThe University of EdinburghEdinburghUK
| | - Juliane Schroeder
- Institute of Immunology and Infection ResearchThe University of EdinburghEdinburghUK
- Present address:
Welcome Trust Centre for Molecular Parasitology and Institute of Infection Immunity and InflammationCollege of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - Martin Barrios‐Llerena
- Institute of Immunology and Infection ResearchThe University of EdinburghEdinburghUK
- Present address:
Centre for Cardiovascular SciencesQueen's Medical Research Institute University of EdinburghEdinburghUK
| | - Toni Aebischer
- Mycotic and Parasitic Agents and MycobacteriaDepartment of Infectious DiseasesRobert Koch‐InstituteBerlinGermany
- Institute of Immunology and Infection ResearchThe University of EdinburghEdinburghUK
| |
Collapse
|
9
|
Zaidi A, Singh KP, Ali V. Leishmania and its quest for iron: An update and overview. Mol Biochem Parasitol 2016; 211:15-25. [PMID: 27988301 DOI: 10.1016/j.molbiopara.2016.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 11/21/2016] [Accepted: 12/11/2016] [Indexed: 12/12/2022]
Abstract
Parasites of genus Leishmania are the causative agents of complex neglected diseases called leishmaniasis and continue to be a significant health concern globally. Iron is a vital nutritional requirement for virtually all organisms, including pathogenic trypanosomatid parasites, and plays a crucial role in many facets of cellular metabolism as a cofactor of several enzymes. Iron acquisition is essential for the survival of parasites. Yet parasites are also vulnerable to the toxicity of iron and reactive oxygen species. The aim of this review is to provide an update on the current knowledge about iron acquisition and usage by Leishmania species. We have also discussed about host strategy to modulate iron availability and the strategies deployed by Leishmania parasites to overcome iron withholding defences and thus favour parasite growth within host macrophages. Since iron plays central roles in the host's response and parasite metabolism, a comprehensive understanding of the iron metabolism is beneficial to identify potential viable therapeutic opportunities against leishmaniasis.
Collapse
Affiliation(s)
- Amir Zaidi
- Laboratory of Molecular Biochemistry and Cell Biology, Dept. of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna, India
| | - Krishn Pratap Singh
- Laboratory of Molecular Biochemistry and Cell Biology, Dept. of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna, India
| | - Vahab Ali
- Laboratory of Molecular Biochemistry and Cell Biology, Dept. of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna, India.
| |
Collapse
|
10
|
Okuda K, Tong M, Dempsey B, Moore KJ, Gazzinelli RT, Silverman N. Leishmania amazonensis Engages CD36 to Drive Parasitophorous Vacuole Maturation. PLoS Pathog 2016; 12:e1005669. [PMID: 27280707 PMCID: PMC4900624 DOI: 10.1371/journal.ppat.1005669] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 05/10/2016] [Indexed: 11/19/2022] Open
Abstract
Leishmania amastigotes manipulate the activity of macrophages to favor their own success. However, very little is known about the role of innate recognition and signaling triggered by amastigotes in this host-parasite interaction. In this work we developed a new infection model in adult Drosophila to take advantage of its superior genetic resources to identify novel host factors limiting Leishmania amazonensis infection. The model is based on the capacity of macrophage-like cells, plasmatocytes, to phagocytose and control the proliferation of parasites injected into adult flies. Using this model, we screened a collection of RNAi-expressing flies for anti-Leishmania defense factors. Notably, we found three CD36-like scavenger receptors that were important for defending against Leishmania infection. Mechanistic studies in mouse macrophages showed that CD36 accumulates specifically at sites where the parasite contacts the parasitophorous vacuole membrane. Furthermore, CD36-deficient macrophages were defective in the formation of the large parasitophorous vacuole typical of L. amazonensis infection, a phenotype caused by inefficient fusion with late endosomes and/or lysosomes. These data identify an unprecedented role for CD36 in the biogenesis of the parasitophorous vacuole and further highlight the utility of Drosophila as a model system for dissecting innate immune responses to infection.
Collapse
Affiliation(s)
- Kendi Okuda
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (KO); (NS)
| | - Mei Tong
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Brian Dempsey
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Kathryn J. Moore
- Department of Medicine, New York University School of Medicine, Langone Medical Center, New York, New York, United States of America
| | - Ricardo T. Gazzinelli
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Neal Silverman
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (KO); (NS)
| |
Collapse
|
11
|
Liévin-Le Moal V, Loiseau PM. Leishmania hijacking of the macrophage intracellular compartments. FEBS J 2015; 283:598-607. [PMID: 26588037 DOI: 10.1111/febs.13601] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/08/2015] [Accepted: 11/13/2015] [Indexed: 12/15/2022]
Abstract
Leishmania spp., transmitted to humans by the bite of the sandfly vector, are responsible for the three major forms of leishmaniasis, cutaneous, diffuse mucocutaneous and visceral. Leishmania spp. interact with membrane receptors of neutrophils and macrophages. In macrophages, the parasite is internalized within a parasitophorous vacuole and engages in a particular intracellular lifestyle in which the flagellated, motile Leishmania promastigote metacyclic form differentiates into non-motile, metacyclic amastigote form. This phenomenon is induced by Leishmania-triggered events leading to the fusion of the parasitophorous vacuole with vesicular members of the host cell endocytic pathway including recycling endosomes, late endosomes and the endoplasmic reticulum. Maturation of the parasitophorous vacuole leads to the intracellular proliferation of the Leishmania amastigote forms by acquisition of host cell nutrients while escaping host defense responses.
Collapse
Affiliation(s)
- Vanessa Liévin-Le Moal
- Anti-Parasitic Chemotherapy, Faculté de Pharmacie, CNRS, UMR 8076 BioCIS, Châtenay-Malabry, France.,Université Paris-Sud, Orsay, France.,Faculté de Pharmacie, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LabEx LERMIT), Châtenay-Malabry, France
| | - Philippe M Loiseau
- Anti-Parasitic Chemotherapy, Faculté de Pharmacie, CNRS, UMR 8076 BioCIS, Châtenay-Malabry, France.,Université Paris-Sud, Orsay, France.,Faculté de Pharmacie, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LabEx LERMIT), Châtenay-Malabry, France
| |
Collapse
|
12
|
Genetically Modified Live Attenuated Leishmania donovani Parasites Induce Innate Immunity through Classical Activation of Macrophages That Direct the Th1 Response in Mice. Infect Immun 2015; 83:3800-15. [PMID: 26169275 DOI: 10.1128/iai.00184-15] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/07/2015] [Indexed: 01/05/2023] Open
Abstract
Visceral leishmaniasis (VL) causes significant mortality and there is no effective vaccine. Previously, we have shown that genetically modified Leishmania donovani parasites, here described as live attenuated parasites, induce a host protective adaptive immune response in various animal models. In this study, we demonstrate an innate immune response upon infection with live attenuated parasites in macrophages from BALB/c mice both in vitro and in vivo. In vitro infection of macrophages with live attenuated parasites (compared to that with wild-type [WT] L. donovani parasites) induced significantly higher production of proinflammatory cytokines (tumor necrosis factor alpha [TNF-α], interleukin-12 [IL-12], gamma interferon [IFN-γ], and IL-6), chemokines (monocyte chemoattractant protein 1/CCL-2, macrophage inflammatory protein 1α/CCL-3, and IP-10), reactive oxygen species (ROS), and nitric oxide, while concomitantly reducing anti-inflammatory cytokine IL-10 and arginase-1 activities, suggesting a dominant classically activated/M1 macrophage response. The classically activated response in turn helps in presenting antigen to T cells, as observed with robust CD4(+) T cell activation in vitro. Similarly, parasitized splenic macrophages from live attenuated parasite-infected mice also demonstrated induction of an M1 macrophage phenotype, indicated by upregulation of IL-1β, TNF-α, IL-12, and inducible nitric oxide synthase 2 and downregulation of genes associated with the M2 phenotype, i.e., the IL-10, YM1, Arg-1, and MRC-1 genes, compared to WT L. donovani-infected mice. Furthermore, an ex vivo antigen presentation assay showed macrophages from live attenuated parasite-infected mice induced higher IFN-γ and IL-2 but significantly less IL-10 production by ovalbumin-specific CD4(+) T cells, resulting in proliferation of Th1 cells. These data suggest that infection with live attenuated parasites promotes a state of classical activation (M1 dominant) in macrophages that leads to the generation of protective Th1 responses in BALB/c mice.
Collapse
|
13
|
Transferrin: Endocytosis and Cell Signaling in Parasitic Protozoa. BIOMED RESEARCH INTERNATIONAL 2015; 2015:641392. [PMID: 26090431 PMCID: PMC4450279 DOI: 10.1155/2015/641392] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/18/2014] [Indexed: 12/31/2022]
Abstract
Iron is the fourth most abundant element on Earth and the most abundant metal in the human body. This element is crucial for life because almost all organisms need iron for several biological activities. This is the case with pathogenic organisms, which are at the vanguard in the battle with the human host for iron. The latest regulates Fe concentration through several iron-containing proteins, such as transferrin. The transferrin receptor transports iron to each cell that needs it and maintains it away from pathogens. Parasites have developed several strategies to obtain iron as the expression of specific transferrin receptors localized on plasma membrane, internalized through endocytosis. Signal transduction pathways related to the activation of the receptor have functional importance in proliferation. The study of transferrin receptors and other proteins with action in the signaling networks is important because these proteins could be used as therapeutic targets due to their specificity or to differences with the human counterpart. In this work, we describe proteins that participate in signal transduction processes, especially those that involve transferrin endocytosis, and we compare these processes with those found in T. brucei, T. cruzi, Leishmania spp., and E. histolytica parasites.
Collapse
|
14
|
Abstract
Leishmania amazonensis is an intracellular protozoan parasite responsible for chronic cutaneous leishmaniasis (CL). CL is a neglected tropical disease responsible for infecting millions of people worldwide. L. amazonensis promotes alteration of various signaling pathways that are essential for host cell survival. Specifically, through parasite-mediated phosphorylation of extracellular signal regulated kinase (ERK), L. amazonensis inhibits cell-mediated parasite killing and promotes its own survival by co-opting multiple host cell functions. In this review, we highlight Leishmania-host cell signaling alterations focusing on those specific to (1) motor proteins, (2) prevention of NADPH subunit phosphorylation impairing reactive oxygen species production, and (3) localized endosomal signaling to up-regulate ERK phosphorylation. This review will focus upon mechanisms and possible explanations as to how Leishmania spp. evades the various layers of defense employed by the host immune response.
Collapse
|
15
|
Cecílio P, Pérez-Cabezas B, Santarém N, Maciel J, Rodrigues V, Cordeiro da Silva A. Deception and manipulation: the arms of leishmania, a successful parasite. Front Immunol 2014; 5:480. [PMID: 25368612 PMCID: PMC4202772 DOI: 10.3389/fimmu.2014.00480] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/19/2014] [Indexed: 12/12/2022] Open
Abstract
Leishmania spp. are intracellular parasitic protozoa responsible for a group of neglected tropical diseases, endemic in 98 countries around the world, called leishmaniasis. These parasites have a complex digenetic life cycle requiring a susceptible vertebrate host and a permissive insect vector, which allow their transmission. The clinical manifestations associated with leishmaniasis depend on complex interactions between the parasite and the host immune system. Consequently, leishmaniasis can be manifested as a self-healing cutaneous affliction or a visceral pathology, being the last one fatal in 85–90% of untreated cases. As a result of a long host–parasite co-evolutionary process, Leishmania spp. developed different immunomodulatory strategies that are essential for the establishment of infection. Only through deception and manipulation of the immune system, Leishmania spp. can complete its life cycle and survive. The understanding of the mechanisms associated with immune evasion and disease progression is essential for the development of novel therapies and vaccine approaches. Here, we revise how the parasite manipulates cell death and immune responses to survive and thrive in the shadow of the immune system.
Collapse
Affiliation(s)
- Pedro Cecílio
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Begoña Pérez-Cabezas
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Nuno Santarém
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Joana Maciel
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Vasco Rodrigues
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Anabela Cordeiro da Silva
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal ; Department of Biological Sciences, Faculty of Pharmacy, University of Porto , Porto , Portugal
| |
Collapse
|
16
|
Canton J, Kima PE. Targeting host syntaxin-5 preferentially blocks Leishmania parasitophorous vacuole development in infected cells and limits experimental Leishmania infections. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1348-55. [PMID: 22885104 DOI: 10.1016/j.ajpath.2012.06.041] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 06/06/2012] [Accepted: 06/20/2012] [Indexed: 02/08/2023]
Abstract
Our previous observations established a role for syntaxin-5 in the development of Leishmania parasitophorous vacuoles (LPVs). In this study, we took advantage of the recent identification of Retro-2, a small organic molecule that can cause the redistribution of syntaxin-5; we show herein that Retro-2 blocks LPV development within 2 hours of adding it to cells infected with Leishmania amazonensis. In infected cells incubated for 48 hours with Retro-2, LPV development was significantly limited; furthermore, infected cells harbored four to five times fewer parasites than infected cells incubated in vehicle alone. In vivo studies revealed that Retro-2 curbed experimental L. amazonensis infections in a dose-dependent manner. Retro-2 did not have any appreciable effect on the host cell physiological characteristics; furthermore, it had no apparent toxicity in experimental animals. An unexpected, but welcome, finding was that Retro-2 inhibited the replication of Leishmania parasites in axenic cultures. This study is significant because it identifies an endoplasmic reticulum/Golgi SNARE as a potential target for the control of Leishmania infections; moreover, it suggests that small organic molecules can be identified that can selectively disrupt the vesicle fusion machinery that promotes the development of pathogen-containing compartments without exerting toxic effects on the host.
Collapse
Affiliation(s)
- Johnathan Canton
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida 32611, USA
| | | |
Collapse
|
17
|
De Cicco NNT, Pereira MG, Corrêa JR, Andrade-Neto VV, Saraiva FB, Chagas-Lima AC, Gondim KC, Torres-Santos EC, Folly E, Saraiva EM, Cunha-E-Silva NL, Soares MJ, Atella GC. LDL uptake by Leishmania amazonensis: involvement of membrane lipid microdomains. Exp Parasitol 2012; 130:330-40. [PMID: 22381219 DOI: 10.1016/j.exppara.2012.02.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 02/10/2012] [Accepted: 02/13/2012] [Indexed: 02/02/2023]
Abstract
Leishmania amazonensis lacks a de novo mechanism for cholesterol synthesis and therefore must scavenge this lipid from the host environment. In this study we show that the L. amazonensis takes up and metabolizes human LDL(1) particles in both a time and dose-dependent manner. This mechanism implies the presence of a true LDL receptor because the uptake is blocked by both low temperature and by the excess of non-labelled LDL. This receptor is probably associated with specific microdomains in the membrane of the parasite, such as rafts, because this process is blocked by methyl-β-cyclodextrin (MCBD). Cholesteryl ester fluorescently-labeled LDL (BODIPY-cholesteryl-LDL) was used to follow the intracellular distribution of this lipid. After uptake it was localized in large compartments along the parasite body. The accumulation of LDL was analyzed by flow cytometry using FITC-labeled LDL particles. Together these data show for the first time that L. amazonensis is able to compensate for its lack of lipid synthesis through the use of a lipid importing machinery largely based on the uptake of LDL particles from the host. Understanding the details of the molecular events involved in this mechanism may lead to the identification of novel targets to block Leishmania infection in human hosts.
Collapse
Affiliation(s)
- Nuccia N T De Cicco
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21.941-902, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Real F, Mortara RA. The diverse and dynamic nature of Leishmania parasitophorous vacuoles studied by multidimensional imaging. PLoS Negl Trop Dis 2012; 6:e1518. [PMID: 22348167 PMCID: PMC3279510 DOI: 10.1371/journal.pntd.0001518] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 12/22/2011] [Indexed: 12/23/2022] Open
Abstract
An important area in the cell biology of intracellular parasitism is the customization of parasitophorous vacuoles (PVs) by prokaryotic or eukaryotic intracellular microorganisms. We were curious to compare PV biogenesis in primary mouse bone marrow-derived macrophages exposed to carefully prepared amastigotes of either Leishmania major or L. amazonensis. While tight-fitting PVs are housing one or two L. major amastigotes, giant PVs are housing many L. amazonensis amastigotes. In this study, using multidimensional imaging of live cells, we compare and characterize the PV biogenesis/remodeling of macrophages i) hosting amastigotes of either L. major or L. amazonensis and ii) loaded with Lysotracker, a lysosomotropic fluorescent probe. Three dynamic features of Leishmania amastigote-hosting PVs are documented: they range from i) entry of Lysotracker transients within tight-fitting, fission-prone L. major amastigote-housing PVs; ii) the decrease in the number of macrophage acidic vesicles during the L. major PV fission or L. amazonensis PV enlargement; to iii) the L. amazonensis PV remodeling after homotypic fusion. The high content information of multidimensional images allowed the updating of our understanding of the Leishmania species-specific differences in PV biogenesis/remodeling and could be useful for the study of other intracellular microorganisms. Leishmania parasites lodge in host cells within phagolysosome-like structures called parasitophorous vacuoles (PVs). Depending on the species, amastigote forms can be individually hosted within small, tight-fitting PVs or grouped within loose, spacious PVs. Using multidimensional live cell imaging, we examined the biogenesis of the two PV phenotypes in macrophages exposed to L. major (a representative of the tight PV phenotype) or L. amazonensis (an example of the loose PV phenotype) amastigotes. L. major PVs undergo fission as parasites divide; we demonstrate that in the course of fission there are transients of the lysosomotropic fluorescent probe Lysotracker. In contrast, during the course of amastigote population size expansion, L. amazonensis PVs do accumulate Lysotracker while increasing in diameter and volume. The large PVs fuse together, and the products of fusion undergo size and shape remodeling. The biogenesis/remodeling of the two types of Leishmania PVs is accompanied by a reduction in the number of macrophage acidic vesicles. The present imaging study adds new morphometric information to the cell biology of Leishmania amastigote intracellular parasitism.
Collapse
Affiliation(s)
- Fernando Real
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil.
| | | |
Collapse
|
19
|
McConville MJ, Naderer T. Metabolic pathways required for the intracellular survival of Leishmania. Annu Rev Microbiol 2012; 65:543-61. [PMID: 21721937 DOI: 10.1146/annurev-micro-090110-102913] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Leishmania spp. are sandfly-transmitted parasitic protozoa that cause a spectrum of important diseases and lifelong chronic infections in humans. In the mammalian host, these parasites proliferate within acidified vacuoles in several phagocytic host cells, including macrophages, dendritic cells, and neutrophils. In this review, we discuss recent progress that has been made in defining the nutrient composition of the Leishmania parasitophorous vacuole, as well as metabolic pathways required by these parasites for virulence. Analysis of the virulence phenotype of Leishmania mutants has been particularly useful in defining carbon sources and nutrient salvage pathways that are essential for parasite persistence and/or induction of pathology. We also review data suggesting that intracellular parasite stages modulate metabolic processes in their host cells in order to generate a more permissive niche.
Collapse
Affiliation(s)
- Malcolm J McConville
- Department of Biochemistry and Molecular Biology, University of Melbourne, Bio21 Institute of Molecular Science and Biotechnology, Parkville, Victoria 3010, Australia.
| | | |
Collapse
|
20
|
de Assis RR, Ibraim IC, Nogueira PM, Soares RP, Turco SJ. Glycoconjugates in New World species of Leishmania: polymorphisms in lipophosphoglycan and glycoinositolphospholipids and interaction with hosts. Biochim Biophys Acta Gen Subj 2011; 1820:1354-65. [PMID: 22093608 DOI: 10.1016/j.bbagen.2011.11.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 10/31/2011] [Accepted: 11/01/2011] [Indexed: 11/29/2022]
Abstract
BACKGROUND Protozoan parasites of the genus Leishmania cause a number of important diseases in humans and undergo a complex life cycle, alternating between a sand fly vector and vertebrate hosts. The parasites have a remarkable capacity to avoid destruction in which surface molecules are determinant for survival. Amongst the many surface molecules of Leishmania, the glycoconjugates are known to play a central role in host-parasite interactions and are the focus of this review. SCOPE OF THE REVIEW The most abundant and best studied glycoconjugates are the Lipophosphoglycans (LPGs) and glycoinositolphospholipids (GIPLs). This review summarizes the main studies on structure and biological functions of these molecules in New World Leishmania species. MAJOR CONCLUSIONS LPG and GIPLs are complex molecules that display inter- and intraspecies polymorphisms. They are key elements for survival inside the vector and to modulate the vertebrate immune response during infection. GENERAL SIGNIFICANCE Most of the studies on glycoconjugates focused on Old World Leishmania species. Here, it is reported some of the studies involving New World species and their biological significance on host-parasite interaction. This article is part of a Special Issue entitled Glycoproteomics.
Collapse
Affiliation(s)
- Rafael Ramiro de Assis
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, FIOCRUZ, Av. Augusto de Lima, 1715, Belo Horizonte, MG 30190-002, Brazil
| | | | | | | | | |
Collapse
|
21
|
Endocytosis and Sphingolipid Scavenging in Leishmania mexicana Amastigotes. Biochem Res Int 2011; 2012:691363. [PMID: 21941657 PMCID: PMC3177366 DOI: 10.1155/2012/691363] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 07/18/2011] [Accepted: 07/22/2011] [Indexed: 01/03/2023] Open
Abstract
Leishmania species are the causative agents of the leishmaniases, a spectrum of neglected tropical diseases. Amastigote stage parasites exist within macrophages and scavenge host factors for survival, for example, Leishmania species utilise host sphingolipid for synthesis of complex sphingolipid. In this study L. mexicana endocytosis was shown to be significantly upregulated in amastigotes, indicating that sphingolipid scavenging may be enhanced. However, inhibition of host sphingolipid biosynthesis had no significant effect on amastigote proliferation within a macrophage cell line. In addition, infection itself did not directly influence host biosynthesis. Notably, in contrast to L. major, L. mexicana amastigotes are indicated to possess a complete biosynthetic pathway suggesting that scavenged sphingolipids may be nonessential for proliferation. This suggested that Old and New World species differ in their interactions with the macrophage host. This will need to be considered when targeting the Leishmania sphingolipid biosynthetic pathway with novel therapeutics.
Collapse
|
22
|
|
23
|
Nutrient transport and pathogenesis in selected parasitic protozoa. EUKARYOTIC CELL 2011; 10:483-93. [PMID: 21216940 DOI: 10.1128/ec.00287-10] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Parasitic protozoa, such as malaria parasites, trypanosomes, and Leishmania, acquire a plethora of nutrients from their hosts, employing transport proteins located in the plasma membrane of the parasite. Application of molecular genetic approaches and the completion of genome projects have allowed the identification and functional characterization of a cohort of transporters and their genes in these parasites. This review focuses on a subset of these permeases that have been studied in some detail, that import critical nutrients, and that provide examples of approaches being undertaken broadly with these and other parasite transporters. Permeases reviewed include those for hexoses, purines, iron, polyamines, carboxylates, and amino acids. Topics of special emphasis include structure-function approaches, critical roles for transporters in parasite viability and physiology, regulation of transporter expression, and subcellular targeting. Investigations of parasite transporters impact a broad spectrum of basic biological problems in these protozoa.
Collapse
|
24
|
Naderer T, Heng J, McConville MJ. Evidence that intracellular stages of Leishmania major utilize amino sugars as a major carbon source. PLoS Pathog 2010; 6:e1001245. [PMID: 21203480 PMCID: PMC3009595 DOI: 10.1371/journal.ppat.1001245] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 11/29/2010] [Indexed: 11/20/2022] Open
Abstract
Intracellular parasites, such as Leishmania spp, must acquire suitable carbon sources from the host cell in order to replicate. Here we present evidence that intracellular amastigote stages of Leishmania exploit amino sugars in the phagolysosome of mammalian macrophages as a source of carbon and energy. L. major parasites are capable of using N-acetylglucosamine and glucosamine as primarily carbon sources and contain key enzymes required for conversion of these sugars to fructose-6-phosphate. The last step in this pathway is catalyzed by glucosamine-6-phosphate deaminase (GND), which was targeted to glycosomes via a canonical C-terminal targeting signal when expressed as a GFP fusion protein. Mutant parasites lacking GND were unable to grow in medium containing amino sugars as sole carbohydrate source and rapidly lost viability, concomitant with the hyper-accumulation of hexosamine-phosphates. Expression of native GND, but not a cytosolic form of GND, in Δgnd parasites restored hexosamine-dependent growth, indicating that toxicity is due to depletion of glycosomal pools of ATP. Non-lethal increases in hexosamine phosphate levels in both Δgnd and wild type parasites was associated with a defect in promastigote metacyclogenesis, suggesting that hexosamine phosphate levels may influence parasite differentiation. Promastigote and amastigote stages of the Δgnd mutant were unable to replicate within macrophages and were either completely cleared or exhibited reduced lesion development in highly susceptible Balb/c mice. Our results suggest that hexosamines are a major class of sugars in the macrophage phagolysosome and that catabolism of scavenged amino sugars is required to sustain essential metabolic pathways and prevent hexosamine toxicity. Protozoan parasites belonging to the genus Leishmania are transmitted by sandfly vectors and cause a number of important diseases in humans. These parasites proliferate within mature lysosome compartments in macrophages and other phagocytic cells in the mammalian host. How intracellular stages of Leishmania survive within this hydrolytic compartment and the extent to which they utilize different carbon sources is poorly defined. Previous studies have suggested that sugar uptake is important for growth, although the nature of these sugars is unknown. In this study we show that Leishmania express all of the enzymes needed to degrade the amino sugars, glucosamine and N-acetylglucosamine. We show that a key enzyme in this pathway is sequestered within modified peroxisomes, or glycosomes, and that this localization is essential for growth on amino sugars and avoidance of amino sugar toxicity. This pathway is also required for parasite proliferation within cultured macrophages and for normal infection of highly susceptible mice. Mutant parasites are either completely eradicated or induce small lesions in Balb/c mice after an extended lag period. These findings suggest that amino sugars generated by the lysosomal breakdown of host glycoconjugates are an important carbon source for intracellular stages of Leishmania, particularly during the early stages of infection.
Collapse
Affiliation(s)
- Thomas Naderer
- The Department of Biochemistry and Molecular Biology, University of Melbourne, Bio21 Institute of Molecular Science and Biotechnology, Parkville, Victoria, Australia
| | - Joanne Heng
- The Department of Biochemistry and Molecular Biology, University of Melbourne, Bio21 Institute of Molecular Science and Biotechnology, Parkville, Victoria, Australia
| | - Malcolm J. McConville
- The Department of Biochemistry and Molecular Biology, University of Melbourne, Bio21 Institute of Molecular Science and Biotechnology, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
25
|
Fusion between Leishmania amazonensis and Leishmania major parasitophorous vacuoles: live imaging of coinfected macrophages. PLoS Negl Trop Dis 2010; 4:e905. [PMID: 21151877 PMCID: PMC2998430 DOI: 10.1371/journal.pntd.0000905] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 11/03/2010] [Indexed: 12/14/2022] Open
Abstract
Protozoan parasites of the genus Leishmania alternate between flagellated, elongated extracellular promastigotes found in insect vectors, and round-shaped amastigotes enclosed in phagolysosome-like Parasitophorous Vacuoles (PVs) of infected mammalian host cells. Leishmania amazonensis amastigotes occupy large PVs which may contain many parasites; in contrast, single amastigotes of Leishmania major lodge in small, tight PVs, which undergo fission as parasites divide. To determine if PVs of these Leishmania species can fuse with each other, mouse macrophages in culture were infected with non-fluorescent L. amazonensis amastigotes and, 48 h later, superinfected with fluorescent L. major amastigotes or promastigotes. Fusion was investigated by time-lapse image acquisition of living cells and inferred from the colocalization of parasites of the two species in the same PVs. Survival, multiplication and differentiation of parasites that did or did not share the same vacuoles were also investigated. Fusion of PVs containing L. amazonensis and L. major amastigotes was not found. However, PVs containing L. major promastigotes did fuse with pre-established L. amazonensis PVs. In these chimeric vacuoles, L. major promastigotes remained motile and multiplied, but did not differentiate into amastigotes. In contrast, in doubly infected cells, within their own, unfused PVs metacyclic-enriched L. major promastigotes, but not log phase promastigotes - which were destroyed - differentiated into proliferating amastigotes. The results indicate that PVs, presumably customized by L. major amastigotes or promastigotes, differ in their ability to fuse with L. amazonensis PVs. Additionally, a species-specific PV was required for L. major destruction or differentiation – a requirement for which mechanisms remain unknown. The observations reported in this paper should be useful in further studies of the interactions between PVs to different species of Leishmania parasites, and of the mechanisms involved in the recognition and fusion of PVs. Many non-viral intracellular pathogens lodge within cell vesicles known as “parasitophorous vacuoles” (PVs), which exhibit a variety of pathogen-dependent functional and compositional phenotypes. PVs of the protozoan Leishmania are similar to the digestive organelles known as phagolysosomes. We asked if, in phagocytes infected with two different Leishmania species, would the two parasites be found in the same or in separate vacuoles? Of the species chosen, Leishmania amazonensis develops within large vacuoles which shelter many parasites; in contrast, Leishmania major lodges in small PVs containing one or two parasites. In the present experiments, the species and their life-cycle stages (extracellular promastigotes, and intracellular amastigotes) were distinguished by means of fluorescent markers, and the intracellular localization of the parasites was examined in living cells. We report here that, whereas L. major amastigotes remained within their individual vacuoles, L. major promastigotes were delivered to L. amazonensis vacuoles, in which they survived and multiplied but were unable to differentiate into amastigotes. A species-specific vacuole was thus required for L. major differentiation. The model should be useful in cellular and molecular studies of the biology of these parasites and of their parasitophorous vacuoles.
Collapse
|
26
|
Späth GF, Schlesinger P, Schreiber R, Beverley SM. A novel role for Stat1 in phagosome acidification and natural host resistance to intracellular infection by Leishmania major. PLoS Pathog 2009; 5:e1000381. [PMID: 19381261 PMCID: PMC2663844 DOI: 10.1371/journal.ppat.1000381] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 03/16/2009] [Indexed: 01/14/2023] Open
Abstract
Intracellular parasites of the genus Leishmania generate severe diseases in humans, which are associated with a failure of the infected host to induce a protective interferon γ (IFNγ)-mediated immune response. We tested the role of the JAK/STAT1 signaling pathway in Leishmania pathogenesis by utilizing knockout mice lacking the signal transducer and activator of transcription 1 (Stat1) and derived macrophages. Unexpectedly, infection of Stat1-deficient macrophages in vitro with promastigotes from Leishmania major and attenuated LPG1 knockout mutants (lpg−) specifically lacking lipophosphoglycan (LPG) resulted in a twofold increased intracellular growth, which was independent of IFNγ and associated with a substantial increase in phagosomal pH. Phagosomes in Stat1−/− macrophages showed normal maturation as judged by the accumulation of the lysosomal marker protein rab7, and provided normal vATPase activity, but were defective in the anion conductive pathway required for full vesicular acidification. Our results suggest a role of acidic pH in the control of intracellular Leishmania growth early during infection and identify for the first time an unexpected role of Stat1 in natural anti-microbial resistance independent from its function as IFNγ-induced signal transducer. This novel Stat1 function may have important implications to studies of other pathogens, as the acidic phagolysosomal pH plays an important role in antigen processing and the uncoating process of many viruses. Protozoan parasites of the genus Leishmania generate a variety of pathologies, collectively termed leishmaniasis, which afflict millions of people worldwide. Leishmania is transmitted during the blood meal of infested sand flies that inoculate highly infective metacyclic promastigotes into the mammalian host. Following uptake by host macrophages, metacyclics differentiate into the amastigote form that replicates inside the acidified phago-lysosome of the host cell. The cytokine interferon-γ activates infected macrophages to kill intracellular Leishmania through the production of nitric oxide. This process is mediated through Stat 1, a cytosolic transcription factor that translocates into the nucleus in response to the cytokine, where it induces a pleiotropic anti-microbial response. By utilizing Stat1-deficient macrophages we found evidence for a novel interferon-γ-independent physiological function of Stat1 in acidification of the host cell phago-lysosome. Stat1-deficient macrophages showed higher phago-lysosomal pH and increased susceptibility to Leishmania infection, which was linked to a defect in cellular chloride channel function. Vesicular pH and acidification are important factors affecting the infective cycle of bacterial and protozoan pathogens, and the uncoating process during viral entry. Thus, the role of cytokine-independent Stat1 functions in innate anti-microbial resistance may have a greater impact on host-pathogen interactions than previously appreciated.
Collapse
Affiliation(s)
- Gerald F. Späth
- Department of Molecular Microbiology, Washington University Medical School, St. Louis, Missouri, United States of America
- Department of Parasitology and Mycology, Laboratory of Parasite Virulence, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) AVENIR, Paris, France
| | - Paul Schlesinger
- Department of Cell Biology, Washington University Medical School, St. Louis, Missouri, United States of America
| | - Robert Schreiber
- Department of Pathology and Immunology, Washington University Medical School, St. Louis, Missouri, United States of America
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University Medical School, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
27
|
Nicoletti S, Seifert K, Gilbert IH. N-(2-hydroxypropyl)methacrylamide-amphotericin B (HPMA-AmB) copolymer conjugates as antileishmanial agents. Int J Antimicrob Agents 2008; 33:441-8. [PMID: 19097763 PMCID: PMC2669511 DOI: 10.1016/j.ijantimicag.2008.10.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 10/07/2008] [Accepted: 10/09/2008] [Indexed: 11/02/2022]
Abstract
Leishmaniasis is a major health problem in many parts of the world, caused by various species of Leishmania. Amastigotes are the clinically relevant form of the parasite in the human host and reside in the parasitophorous vacuole within macrophages. Polymer-drug conjugates have been used for lysosomotropic drug delivery and have already shown potential in anticancer and antileishmanial chemotherapy. We synthesised N-(2-hydroxypropyl)methacrylamide-amphotericin B (HPMA-AmB) copolymer conjugates in which the AmB was attached to the polymer through a degradable GlyPheLeuGly linker. Antileishmanial activity was assessed in vitro against intracellular amastigotes in host macrophages [murine peritoneal exudate macrophages (PEMs), murine bone marrow-derived macrophages (BMMs) and differentiated THP-1 cells]. The most potent copolymers had 50% effective concentration (EC(50)) values of 0.03 microg/mL AmB equivalent against Leishmania donovani amastigotes in PEMs and BMMs and an EC(50) of 0.57 microg/mL AmB equivalent against L. donovani in THP-1 cells. This activity was comparable with free AmB (EC(50)=0.03-0.07 microg/mL against L. donovani in PEMs and BMMs and 0.24-0.42 microg/mL against amastigotes in THP-1 cells) and Fungizone (EC(50)=0.04-0.07 microg/mL against amastigotes in PEMs). Conjugates also showed potent in vivo activity with ca. 50% inhibition of parasite burden at 1mg/kg body weight.
Collapse
Affiliation(s)
- Salvatore Nicoletti
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Sir James Black Centre, Dundee DD1 5EH, UK
| | | | | |
Collapse
|
28
|
Pinheiro RO, Nunes MP, Pinheiro CS, D'Avila H, Bozza PT, Takiya CM, Côrte-Real S, Freire-de-Lima CG, DosReis GA. Induction of autophagy correlates with increased parasite load of Leishmania amazonensis in BALB/c but not C57BL/6 macrophages. Microbes Infect 2008; 11:181-90. [PMID: 19070676 DOI: 10.1016/j.micinf.2008.11.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 11/03/2008] [Accepted: 11/11/2008] [Indexed: 12/12/2022]
Abstract
We investigated the role of autophagy in infection of macrophages by Leishmania amazonensis. Induction of autophagy by IFN-gamma or starvation increased intracellular parasite load and the percentages of infected macrophages from BALB/c but not from C57BL/6 mice. In contrast, starvation did not affect the replication of either Leishmania major or Trypanosoma cruzi in BALB/c macrophages. In BALB/c macrophages, starvation resulted in increased monodansylcadaverine staining and in the appearance of double-membrane and myelin-like vesicles characteristic of autophagosomes. Increased parasite load was associated with a reduction in NO levels and was attenuated by wortmannin, an inhibitor of autophagy. In infected macrophages from BALB/c, but not from C57BL/6 mice, starvation increased the number of lipid bodies and the amounts of PGE(2) produced. Exogenous PGE(2) increased parasite load in macrophages from BALB/c, but not C57BL/6 mice. The cyclooxygenase inhibitor indomethacin prevented the increase of parasite load in starved BALB/c macrophages, and actually induced parasite killing. These results suggest that autophagy regulates the outcome of L. amazonensis infection in macrophages in a host strain specific manner.
Collapse
|
29
|
SopE-mediated recruitment of host Rab5 on phagosomes inhibits Salmonella transport to lysosomes. Methods Mol Biol 2008; 445:417-37. [PMID: 18425466 DOI: 10.1007/978-1-59745-157-4_27] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Phagocytosis is a process by which invading organisms are taken up by macrophages and targeted to the lysosomes, where they are degraded. However, many pathogens modulate this central process of macrophage-mediated killing by inhibiting their transport to the lysosomes through a variety of pathogen-derived mechanisms. Given the importance of Rab proteins in the regulation of intracellular transport pathways, we investigated the role of different host endocytic Rabs on the maturation of Salmonella-containing phagosomes in macrophages. Initially, we have developed a ligand mixing assay to measure the transport of the Salmonella-containing phagosomes to lysosomes. Using this assay we have shown that Salmonella decline their transport to the lysosomes. In order to determine whether inhibition of Salmonella transport to lysosomes is due to their sustained fusion with early endosomes, we have developed an in vitro fusion assay between Salmonella-containing phagosomes and early endosomes. Here, we have discussed how these methodologies are helpful to determine the mechanism of evasion of Salmonella transport to the lysosomes.
Collapse
|
30
|
Leishmania (L.) amazonensis: fusion between parasitophorous vacuoles in infected bone-marrow derived mouse macrophages. Exp Parasitol 2008; 119:15-23. [PMID: 18346736 DOI: 10.1016/j.exppara.2007.12.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 10/02/2007] [Accepted: 12/11/2007] [Indexed: 11/20/2022]
Abstract
[Leishmania(L.)] amazonensis amastigotes reside in macrophages within spacious parasitophorous vacuoles (PVs) which may contain numerous parasites. After sporadic fusion events were detected by time-lapse cinemicrography, PV fusion was examined in two different models. In single infections, it was inferred from the reduction in PV numbers per cell. In a reinfection model, macrophages infected with unlabeled amastigotes were reinfected with GFP-transfected- or carboxyfluorescein diacetate succinimidyl ester-labeled parasites, and fusion was detected by the colocalization of labeled and unlabeled amastigotes in the same PVs. The main findings were: (1) as expected, fusion frequency increased with the multiplicity of infection; (2) most fusion events took place in the first 24h of infection or reinfection, prior to the multiplication of incoming parasites; (3) resident and incoming parasites multiplied at similar rates in fused PVs. The model should be useful in studies of parasite and host cell factors and mechanisms involved in PV fusogenicity.
Collapse
|
31
|
Locksley RM, Wakil AE, Corry DB, Pingel S, Bix M, Fowell DJ. The development of effector T cell subsets in murine Leishmania major infection. CIBA FOUNDATION SYMPOSIUM 2007; 195:110-7; discussion 117-22. [PMID: 8724833 DOI: 10.1002/9780470514849.ch8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Leishmania major infection has proven an exceptional model for CD4+ subset development in inbred mice. Most strains contain infection coincident with the appearance of T helper 1 (Th1) cells that produce gamma-interferon (IFN-gamma) required for macrophage activation. In contrast, mice on the BALB background are unable to control infection due to the development of Th2 cells that produce counter-regulatory cytokines, particularly interleukin 4 (IL-4), capable of abrogating the effects of IFN-gamma. Selective gene disruption studies in mice have illustrated critical components of the host response to L. major. Mice deficient in beta 2 microglobulin, which have no major histocompatibility complex (MHC) class I or CD8+ T cells, control infection as well as wild-type mice, whereas mice deficient in MHC class II (and CD4+ T cells) suffer fatal infection. Mice with disruption of the gene coding IFN-gamma are also incapable of containing infection, reflecting absolute requirements for this cytokine. A number of interventions have been demonstrated to abrogate Th2 cell development in BALB mice, enabling these mice to control infection. Each of these--IL-12, anti-IL-4, anti-IL-2, anti-CD4 and CTLA4-Ig--has in common the capacity to make IL-4 rate limiting at the time of CD4+ cell priming.
Collapse
Affiliation(s)
- R M Locksley
- Department of Medicine, University of California, San Francisco 94143, USA
| | | | | | | | | | | |
Collapse
|
32
|
Figarella K, Uzcategui NL, Zhou Y, LeFurgey A, Ouellette M, Bhattacharjee H, Mukhopadhyay R. Biochemical characterization of Leishmania major aquaglyceroporin LmAQP1: possible role in volume regulation and osmotaxis. Mol Microbiol 2007; 65:1006-17. [PMID: 17640270 DOI: 10.1111/j.1365-2958.2007.05845.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The Leishmania major aquaglyceroporin, LmAQP1, is responsible for the transport of trivalent metalloids, arsenite and antimonite. We have earlier shown that downregulation of LmAQP1 provides resistance to trivalent antimony compounds whereas increased expression of LmAQP1 in drug-resistant parasites can reverse the resistance. In this paper we describe the biochemical characterization of LmAQP1. Expression of LmAQP1 in Xenopus oocytes rendered them permeable to water, glycerol, methylglyoxal, dihydroxyacetone and sugar alcohols. The transport property of LmAQP1 was severely affected when a critical Arg230, located inside the pore of the channel, was altered to either alanine or lysine. Immunofluorescence and immuno-electron microscopy revealed LmAQP1 to be localized to the flagellum of Leishmania promastigotes and in the flagellar pocket membrane and contractile vacuole/spongiome complex of amastigotes. This is the first report of an aquaglyceroporin being localized to the flagellum of any microbe. Leishmania promastigotes and amastigotes expressing LmAQP1 could regulate their volume in response to hypoosmotic stress. Additionally, Leishmania promastigotes overexpressing LmAQP1 were found to migrate faster towards an osmotic gradient. These results taken together suggest that Leishmania LmAQP1 has multiple physiological roles, being involved in solute transport, volume regulation and osmotaxis.
Collapse
Affiliation(s)
- Katherine Figarella
- Department of Biochemistry and Molecular Biology, Wayne State University, School of Medicine, Detroit, MI, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Dey R, Khan S, Pahari S, Srivastava N, Jadhav M, Saha B. Functional paradox in host–pathogen interaction dictates the fate of parasites. Future Microbiol 2007; 2:425-37. [PMID: 17683278 DOI: 10.2217/17460913.2.4.425] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The interactions between the protozoan parasite Leishmania and host macrophages are complex and involve several paradoxical functions that are meant for protection of the host but exploited by the parasite for its survival. The initial interaction of the parasite surface molecules with the host-cell receptors plays a major role in the final outcome of the disease state. While the interactions between macrophages and a virulent strain of Leishmania trigger a cascade of cell-signaling events leading to immunosuppression, the interaction with an avirulent strain triggers host-protective immune effector functions. Thus, an incisive study on Leishmania–macrophage interactions reveals functional paradoxes that highlight the concept of ‘relativity in parasite virulence’. Using Leishmania infection as a model, we propose that virulence of a pathogen and the resistance (or susceptibility) of a host to the pathogen are relative properties that equate to combinatorial functions of several sets of molecular processes.
Collapse
Affiliation(s)
- Ranadhir Dey
- National Centre for Cell Science, Ganeshkhind, Pune, India.
| | | | | | | | | | | |
Collapse
|
34
|
McConville MJ, de Souza D, Saunders E, Likic VA, Naderer T. Living in a phagolysosome; metabolism of Leishmania amastigotes. Trends Parasitol 2007; 23:368-75. [PMID: 17606406 DOI: 10.1016/j.pt.2007.06.009] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 04/26/2007] [Accepted: 06/13/2007] [Indexed: 12/11/2022]
Abstract
Leishmania amastigotes primarily proliferate within macrophages in the mammalian host. Genome-based metabolic reconstructions, combined with biochemical, reverse genetic and mRNA or protein profiling studies are providing new insights into the metabolism of this intracellular stage. We propose that the complex nutritional requirements of amastigotes have contributed to the tropism of these parasites for the amino acid-rich phagolysosome of macrophages. Amastigote metabolism in this compartment is robust because many metabolic mutants are capable of either growing normally or persisting long term in susceptible animals. New approaches for measuring amastigote metabolism in vivo are discussed.
Collapse
Affiliation(s)
- Malcolm J McConville
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia.
| | | | | | | | | |
Collapse
|
35
|
Castro R, Scott K, Jordan T, Evans B, Craig J, Peters EL, Swier K. The ultrastructure of the parasitophorous vacuole formed by Leishmania major. J Parasitol 2007; 92:1162-70. [PMID: 17304790 DOI: 10.1645/ge-841r.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Protozoan parasites of Leishmania spp. invade macrophages as promastigotes and differentiate into replicative amastigotes within parasitophorous vacuoles. Infection of inbred strains of mice with Leishmania major is a well-studied model of the mammalian immune response to Leishmania species, but the ultrastructure and biochemical properties of the parasitophorous vacuole occupied by this parasite have been best characterized for other species of Leishmania. We examined the parasitophorous vacuole occupied by L. major in lymph nodes of infected mice and in bone marrow-derived macrophages infected in vitro. At all time points after infection, single L. major amastigotes were wrapped tightly by host membrane, suggesting that amastigotes segregate into separate vacuoles during replication. This small, individual vacuole contrasts sharply with the large, communal vacuoles occupied by Leishmania amazonensis. An extensive survey of the literature revealed that the single vacuoles occupied by L. major are characteristic of those formed by Old World species of Leishmania, while New World species of Leishmania form large vacuoles occupied by many amastigotes.
Collapse
Affiliation(s)
- Ramon Castro
- Department of Biological Sciences, Chicago State University, 9501 South King Drive, Chicago, Illinois 60628, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Dey R, Majumder N, Bhattacharjee S, Majumdar SB, Banerjee R, Ganguly S, Das P, Majumdar S. Leishmania donovani-induced ceramide as the key mediator of Akt dephosphorylation in murine macrophages: role of protein kinase Czeta and phosphatase. Infect Immun 2007; 75:2136-42. [PMID: 17220321 PMCID: PMC1865788 DOI: 10.1128/iai.01589-06] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Leishmania donovani is an intracellular protozoan parasite that impairs the host macrophage immune response to render it suitable for its survival and establishment. L. donovani-induced immunosuppression and alteration of host cell signaling is mediated by ceramide, a pleiotropic second messenger playing an important role in regulation of several kinases, including mitogen-activated protein kinase and phosphatases. We observed that the endogenous ceramide generated during leishmanial infection led to the dephosphorylation of protein kinase B (PKB) (Akt) in infected cells. The study of ceramide-mediated Akt phosphorylation revealed that Akt was dephosphorylated at both Thr308 and Ser473 sites in infected cells. Further investigation demonstrated that ceramide was also responsible for the induction of PKCzeta, an atypical Ca-independent stress kinase, as well as the ceramide-activated protein phosphatases (e.g., protein phosphatase 2A [PP2A]). We found that Akt dephosphorylation was mediated by ceramide-induced PKCzeta-Akt association and PP2A activation. In addition, treatment of L. donovani-infected macrophages with PKCzeta-specific inhibitor peptide could restore the translocation of phosphorylated Akt to the cell membrane. This study also revealed that ceramide is involved in the inhibition of proinflammatory cytokine tumor necrosis factor alpha release by infected macrophages. These observations strongly suggest the importance of ceramide in the alteration of normal cellular functions, impairment of the kinase/phosphatase balance, and thereby establishment of leishmaniasis in the hostile macrophage environment.
Collapse
Affiliation(s)
- Ranadhir Dey
- Dept. of Microbiology, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, India
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Lucía Bonilla-Escobar D. Respuesta immune a la leishmaniasis: algo más que linfocitos T. ACTA ACUST UNITED AC 2005. [DOI: 10.1016/s0213-9251(05)72311-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
38
|
Yao C, Luo J, Hsiao C, Donelson JE, Wilson ME. Internal and surface subpopulations of the major surface protease (MSP) of Leishmania chagasi. Mol Biochem Parasitol 2005; 139:173-83. [PMID: 15664652 DOI: 10.1016/j.molbiopara.2004.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Revised: 10/19/2004] [Accepted: 11/03/2004] [Indexed: 11/21/2022]
Abstract
Major surface protease (MSP) facilitates Leishmania promastigote evasion of complement-mediated lysis in the mammalian host and enhances host macrophage phagocytosis of the promastigotes. We previously showed that the steady-state abundance of MSP protein increases 14-fold during in vitro cultivation of L. chagasi promastigotes from logarithmic to stationary phase, despite the fact that the total amount of MSP mRNA does not increase. Furthermore, 10 major MSP isoforms are differentially expressed in different promastigote growth phases, and attenuation of parasites by long-term in vitro cultivation influences MSP isoform expression. Herein, we report that although about two-thirds of newly synthesized MSP becomes surface localized, the rest of the MSP does not reach the promastigote surface. This internal MSP is stable without detectable decrease in abundance up to 6 days after biosynthesis. Furthermore, surface-localized MSP is released at different rates from logarithmic and stationary phase virulent Leishmania promastigotes. These data are consistent with the hypothesis that the major mechanism regulating MSP abundance is the rate of loss of surface-localized MSP from the promastigote surface, and that internally localized MSP is very stable.
Collapse
Affiliation(s)
- Chaoqun Yao
- VA Medical Center, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | | | |
Collapse
|
39
|
Kima PE, Dunn W. Exploiting calnexin expression on phagosomes to isolate Leishmania parasitophorous vacuoles. Microb Pathog 2005; 38:139-45. [PMID: 15797809 DOI: 10.1016/j.micpath.2004.11.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2004] [Revised: 11/17/2004] [Accepted: 11/28/2004] [Indexed: 11/23/2022]
Abstract
We have developed a simple scheme for the isolation of parasitophorous vacuoles (PVs) that harbor Leishmania parasites. This scheme exploits the observation that PVs display endoplasmic reticulum molecules, including the transmembrane protein calnexin. The presence of calnexin at the surface of the PVs distinguishes them from late endosomal vesicles of comparable density. As a result, PVs can be isolated by calnexin affinity selection from an enriched PV fraction obtained by sucrose density fractionation.
Collapse
Affiliation(s)
- Peter E Kima
- Department of Microbiology and Cell Science, University of Florida, Building 981, Box 110700, Gainesville, FL 32611, USA.
| | | |
Collapse
|
40
|
Singh VK, Balaraman S, Tewary P, Madhubala R. Leishmania donovani activates nuclear transcription factor-kappaB in macrophages through reactive oxygen intermediates. Biochem Biophys Res Commun 2004; 322:1086-95. [PMID: 15336576 DOI: 10.1016/j.bbrc.2004.08.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Indexed: 11/28/2022]
Abstract
Interaction of Leishmania donovani with macrophages antagonizes host defense mechanisms by interfering with a cascade of cell signaling processes in the macrophages. An early intracellular signaling event that follows receptor engagement is the activation of transcription factor NF-kappaB. It has been reported earlier that NF-kappaB-dependent signaling pathway regulates proinflammatory cytokine release. We therefore investigated the effect of L. donovani infectivity on this nuclear transcription factor in macrophage cell line J774A.1. Both L. donovani and its surface molecule lipophosphoglycan (LPG) resulted in a dose- and time-dependent activation of NF-kappaB-DNA binding activity in an electrophoretic mobility shift assay. We also report the involvement of IkappaB-alpha and IkappaB-beta in the persistent activation of NF-kappaB by L. donovani. We demonstrate that the NF-kappaB activation was independent of viability of the parasite. Electrophoretic mobility supershift assay indicated that the NF-kappaB complex consists of p65 and c-rel subunits. The interaction of parasite with the macrophages and not the cellular uptake was important for NF-kappaB activation. Both p38 and ERK mitogen activated protein kinase (MAP) activation appears to be necessary for NF-kappaB activation by LPG. Preincubation of cells with antioxidants resulted in inhibition of L. donovani induced NF-kappaB activation, thereby suggesting a potential role of reactive oxygen species in L. donovani induced intracellular signaling. The present data indicate that antioxidants could play an important role in working out various therapeutic modalities to control leishmaniasis.
Collapse
Affiliation(s)
- Vandana Km Singh
- School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| | | | | | | |
Collapse
|
41
|
Russo R, Nigro L, Panarello G, Montineri A. Clinical survey of Leishmania/HIV co-infection in Catania, Italy: the impact of highly active antiretroviral therapy (HAART). ANNALS OF TROPICAL MEDICINE AND PARASITOLOGY 2004; 97 Suppl 1:149-55. [PMID: 14678642 DOI: 10.1179/000349803225002624] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The clinical and parasitological features of visceral leishmaniasis (VL) were investigated, retrospectively, in 27 HIV-infected patients who attended the out-patient clinic of Catania University's Department of Infectious Diseases between 1990 and 1998. The aim was to evaluate the epidemiological, clinical, therapeutic and prognostic characteristics of the co-infection, to determine if there were any interactions between the two infections, and to see if the use of highly active antiretroviral therapy (HAART) had any impact on the leishmaniasis. The most dramatic observation was a marked, HAART-attributable reduction in the annual incidence of VL relapses among the patients.
Collapse
Affiliation(s)
- R Russo
- Department of Infectious Diseases, Catania University, Via Passo Gravina 187, 95125 Catania, Italy.
| | | | | | | |
Collapse
|
42
|
Späth GF, Garraway LA, Turco SJ, Beverley SM. The role(s) of lipophosphoglycan (LPG) in the establishment of Leishmania major infections in mammalian hosts. Proc Natl Acad Sci U S A 2003; 100:9536-41. [PMID: 12869694 PMCID: PMC170953 DOI: 10.1073/pnas.1530604100] [Citation(s) in RCA: 212] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The abundant cell surface glycolipid lipophosphoglycan (LPG) was implicated in many steps of the Leishmania infectious cycle by biochemical tests. The presence of other abundant surface or secreted glycoconjugates sharing LPG domains, however, has led to uncertainty about the relative contribution of LPG in vivo. Here we used an Leishmania major lpg1- mutant, which lacks LPG alone and shows attenuated virulence, to dissect the role of LPG in the establishment of macrophage infections in vivo. lpg1- was highly susceptible to human complement, had lost the ability to inhibit phagolysosomal fusion transiently, and was oxidant sensitive. Studies of mouse mutants defective in relevant defense mechanisms confirmed the role of LPG in oxidant resistance but called into question the importance of transient inhibition of phagolysosomal fusion for Leishmania macrophage survival. Moreover, the limited lytic activity of mouse complement appears to be an ineffective pathogen defense mechanism in vitro and in vivo, unlike human hosts. In contrast, lpg1- parasites bound C3b and resisted low pH and proteases normally, entered macrophages efficiently and silently, and continued to inhibit host-signaling pathways. These studies illustrate the value of mechanistic approaches focusing on both parasite and host defense pathways in dissecting the specific biological roles of complex virulence factors such as LPG.
Collapse
Affiliation(s)
- Gerald F Späth
- Department of Molecular Microbiology, Washington University Medical School, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Chlamydiae, bacterial obligate intracellular pathogens, are the etiologic agents of several human diseases. A large part of the chlamydial intracellular survival strategy involves the formation of a unique organelle called the inclusion that provides a protected site within which they replicate. The chlamydial inclusion is effectively isolated from endocytic pathways but is fusogenic with a subset of exocytic vesicles that deliver sphingomyelin from the Golgi apparatus to the plasma membrane. A combination of host and parasite functions contribute to the biogenesis of this compartment. Establishment of the mature inclusion is accompanied by the insertion of multiple chlamydial proteins, suggesting that chlamydiae actively modify the inclusion to define its interactions with the eukaryotic host cell. Despite being sequestered within a membrane-bound vacuole, chlamydiae clearly communicate with and manipulate the host cell from within this privileged intracellular niche.
Collapse
Affiliation(s)
- Kenneth A Fields
- Host-Parasite Interactions Section, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana 59840, USA
| | | |
Collapse
|
44
|
Gatfield J, Pieters J. Molecular Mechanisms of Host–Pathogen Interaction: Entry and Survival of Mycobacteria in Macrophages. Adv Immunol 2003; 81:45-96. [PMID: 14711053 DOI: 10.1016/s0065-2776(03)81002-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- John Gatfield
- Biozentrum der Universitaet Basel, Department of Biochemistry, Klingelbergstrasse 50-70, 4056 Basel, Switzerland
| | | |
Collapse
|
45
|
Morgan GW, Hall BS, Denny PW, Carrington M, Field MC. The kinetoplastida endocytic apparatus. Part I: a dynamic system for nutrition and evasion of host defences. Trends Parasitol 2002; 18:491-6. [PMID: 12473365 DOI: 10.1016/s1471-4922(02)02391-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The endocytic system of kinetoplastid parasites is a highly polarized membrane network focused on the flagellar pocket localized at one end of the cell. When first characterized, the endosomal network was envisioned as a simple system for uptake of extracellular material by fluid-phase or receptor-mediated mechanisms. Subsequently, it has become clear that the kinetoplastid endosomal system has an active and vital role in avoiding the host immune system and virulence, as well as providing the basic functions to fulfil cellular nutritional requirements. In two reviews, recent advances in the definition and comprehension of kinetoplastida endocytosis are discussed and, in Trypanosoma brucei in particular as the more developed experimental system. In Part 1, the endocytic system is considered in context of the surface molecules and their potential roles in virulence.
Collapse
Affiliation(s)
- Gareth W Morgan
- Wellcome Trust Laboratories for Molecular Parasitology, Dept of Biological Sciences, Imperial College of Science, Technology and Medicine, Exhibition Road, London, UK
| | | | | | | | | |
Collapse
|
46
|
Morehead J, Coppens I, Andrews NW. Opsonization modulates Rac-1 activation during cell entry by Leishmania amazonensis. Infect Immun 2002; 70:4571-80. [PMID: 12117970 PMCID: PMC128177 DOI: 10.1128/iai.70.8.4571-4580.2002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Lesions caused by Leishmania amazonensis normally heal, but relapses occur due to parasite persistence in host tissues. It has been proposed that infection of fibroblasts plays an important role in this process by providing the parasites with a safe haven in which to replicate. However, most previous studies have focused on the entry of Leishmania into macrophages, a process mediated by serum opsonins. To gain insight into a possible role of nonopsonic entry in the intracellular persistence of amastigotes, we examined the invasion of Chinese hamster ovary (CHO) cells. Amastigotes entered CHO cells by a cytochalasin D, genistein, wortmannin, and 2,3-butanedione monoxime-sensitive pathway and replicated within phagolysosomes. However, unlike most phagocytic processes described to date, amastigote internalization in CHO cells involved activation of the GTPases Rho and Cdc42 but not Rac-1. When uptake was mediated by fibronectin or when amastigotes were opsonized with immunoglobulin G and internalized by Fc receptor-expressing CHO cells, Rac-1 activation was restored and found to be required for parasite internalization. Given the essential role of Rac in assembly of the respiratory burst oxidase, invasion through this nonopsonic, Rac-1-independent pathway may play a central role in the intracellular survival of Leishmania in immune hosts.
Collapse
Affiliation(s)
- J Morehead
- Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06536, USA
| | | | | |
Collapse
|
47
|
Fratti RA, Chua J, Deretic V. Cellubrevin alterations and Mycobacterium tuberculosis phagosome maturation arrest. J Biol Chem 2002; 277:17320-6. [PMID: 11825909 DOI: 10.1074/jbc.m200335200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The intracellular trafficking processes controlling phagosomal maturation remain to be fully delineated. Mycobacterium tuberculosis var. bovis BCG, an organism that causes phagosomal maturation arrest, has emerged as a tool for dissection of critical phagosome biogenesis events. In this work, we report that cellubrevin, a v-SNARE functioning in endosomal recycling and implicated in endosomal interactions with post-Golgi compartments, plays a role in phagosomal maturation and that it is altered on mycobacterial phagosomes. Both mycobacterial phagosomes, which undergo maturation arrest, and model phagosomes containing latex beads, which follow the normal pathway of maturation into phagolysosomes, acquired cellubrevin. However, the mycobacterial and model phagosomes differed, as a discrete proteolytic degradation of this SNARE was detected on mycobacterial phagosomes. The observed cellubrevin alteration on mycobacterial phagosomes was not a passive event secondary to a maturation arrest at another checkpoint of the phagosome maturation pathway, since pharmacological inhibitors of phagosomal/endosomal pathways blocking phagosomal maturation did not cause cellubrevin degradation on model phagosomes. Cellubrevin status on phagosomes had consequences on phagosomal membrane and lumenal content trafficking, involving plasma membrane marker recycling and delivery of lysosomal enzymes. These results suggest that cellubrevin plays a role in phagosomal maturation and that it is a target for modification by mycobacteria or by infection-induced processes in the host cell.
Collapse
Affiliation(s)
- Rutilio A Fratti
- Department of Microbiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
48
|
Webster P. Early intracellular events during internalization of Listeria monocytogenes by J774 cells. J Histochem Cytochem 2002; 50:503-18. [PMID: 11897803 DOI: 10.1177/002215540205000407] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The gram-positive bacillus Listeria monocytogenes gains entry into host cells through a phagosome membrane that forms around entering bacteria. During the early stages of internalization the invading bacteria appear to modify the protein composition of the forming phagosome membrane in J774 cells. MHC class II molecules on the cell surface and exposed surface molecules available for biotinylation are excluded from the bacteria-host cell membrane interface and from the forming phagosome. This exclusion of MHC class II molecules from the early phagosome may partially help to explain previous reports suggesting that L. monocytogenes is able to interfere with antigen presentation. Inside the host cell, MHC class II molecules are delivered to the phagosome membrane. This is followed by delivery of LAMP 1, a marker of late endocytic compartments, and fusion with low-pH compartments. The bacteria then escape into the cell cytoplasm, possibly assisted by rapid delivery of this low-pH environment.
Collapse
Affiliation(s)
- Paul Webster
- House Ear Institute, 2100 West Third Street, Los Angeles, CA 90057, USA.
| |
Collapse
|
49
|
Swanson MS, Fernandez-Moreira E, Fernandez-Moreia E. A microbial strategy to multiply in macrophages: the pregnant pause. Traffic 2002; 3:170-7. [PMID: 11886587 DOI: 10.1034/j.1600-0854.2002.030302.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Humans live in harmony with much of the microbial world, thanks to a sophisticated immune system. As the first line of defense, macrophages engulf, digest, and display foreign material, then recruit specialists to eliminate potential threats. Yet infiltrators exist: certain fungi, viruses, parasites, and bacteria thrive within sentinel macrophages. By scrutinizing the life styles of these shrewd microbes, we can deduce how macrophages routinely mount an effective immune response. The bimorphic life cycles of three pathogens have dramatic consequences for phagosome traffic. In the transmissible state, Leishmania spp., Coxiella burnetii, and Legionella pneumophila block phagosome maturation; after a pregnant pause, replicative forms emerge and thrive in lysosomes.
Collapse
Affiliation(s)
- Michele S Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109-0620, USA.
| | | | | |
Collapse
|
50
|
Abstract
Leishmania have a digenetic life cycle, involving a motile, extracellular stage (promastigote) which parasitises the alimentary tract of a sandfly vector. Bloodfeeding activity by an infected sandfly can result in transmission of infective (metacyclic) promastigotes to mammalian hosts, including humans. Leishmania promastigotes are rapidly phagocytosed but may survive and transform into non-motile amastigote forms which can persist as intracellular parasites. Leishmania amastigotes multiply in an acidic intracellular compartment, the parasitophorous vacuole. pH plays a central role in the developmental switch between promastigote and amastigote stages, and amastigotes are metabolically most active when their environment is acidic, although the cytoplasm of the amastigote is regulated at near-neutral pH by an active process of proton extrusion. A steep proton gradient is thus maintained across the amastigote surface and all membrane processes must be adapted to function under these conditions. Amastigote uptake systems for glucose, amino acids, nucleosides and polyamines are optimally active at acidic pH. Promastigote uptake systems are kinetically distinct and function optimally at more neutral environmental pH, indicating that membrane transport activity is developmentally regulated. The nutrient environment encountered by amastigotes is not well understood but the parasitophorous vacuole can fuse with endosomes, phagosomes and autophagosomes, suggesting that a diverse range of macromolecules will be present. The parasitophorous vacuole is a hydrolytic compartment in which such material will be rapidly degraded to low molecular weight components which are typical substrates for membrane transporters. Amastigote surface transporters must compete for these substrates with equivalent host transporters in the membrane of the parasitophorous vacuole. The elaboration of accumulative transporters with high affinity will be beneficial to amastigotes in this environment. The influence of environmental pH on membrane transporter function is discussed, with emphasis on the potential role of a transmembrane proton gradient in active, high affinity transport.
Collapse
Affiliation(s)
- R J Burchmore
- Institute of Biomedical and Life Sciences, Division of Infection and Immunity, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, UK.
| | | |
Collapse
|