301
|
Matheoud D, Moradin N, Bellemare-Pelletier A, Shio MT, Hong WJ, Olivier M, Gagnon E, Desjardins M, Descoteaux A. Leishmania evades host immunity by inhibiting antigen cross-presentation through direct cleavage of the SNARE VAMP8. Cell Host Microbe 2014; 14:15-25. [PMID: 23870310 DOI: 10.1016/j.chom.2013.06.003] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 04/08/2013] [Accepted: 05/17/2013] [Indexed: 11/18/2022]
Abstract
During phagocytosis, microorganisms are taken up by immune cells into phagosomes. Through membrane-trafficking events mediated by SNARE proteins, phagosomes fuse with lysosomes, generating degradative phagolysosomes. Phagolysosomes contribute to host immunity by linking microbial killing within these organelles with antigen processing for presentation on MHC class I or II molecules to T cells. We show that the intracellular parasite Leishmania evades immune recognition by inhibiting phagolysosome biogenesis. The Leishmania cell surface metalloprotease GP63 cleaves a subset of SNAREs, including VAMP8. GP63-mediated VAMP8 inactivation or Vamp8 disruption prevents the NADPH oxidase complex from assembling on phagosomes, thus altering their pH and degradative properties. Consequently, the presentation of exogenous Leishmania antigens on MHC class I molecules, also known as cross-presentation, is inhibited, resulting in reduced T cell activation. These findings indicate that Leishmania subverts immune recognition by altering phagosome function and highlight the importance of VAMP8 in phagosome biogenesis and antigen cross-presentation.
Collapse
Affiliation(s)
- Diana Matheoud
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
302
|
Frézard F, Monte-Neto R, Reis PG. Antimony transport mechanisms in resistant leishmania parasites. Biophys Rev 2014; 6:119-132. [PMID: 28509965 DOI: 10.1007/s12551-013-0134-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 12/05/2013] [Indexed: 11/26/2022] Open
Abstract
Antimonial compounds have been used for more than a century in the treatment of the parasitic disease leishmaniasis. Although pentavalent antimonials are still first-line drugs in several developing countries, this class of drugs is no longer recommended in the Indian sub-continent because of the emergence of drug resistance. The precise mechanisms involved in the resistance of leishmania parasites to antimony are still subject to debate. It is now well documented that drug resistance in leishmania parasites is a multifactorial phenomenon involving multiple genes whose expression pattern synergistically leads to the resistance phenotype. The reduction of intracellular antimony accumulation is a frequent change observed in resistant leishmania cells; however, no comprehensive transport model has been presented so far to explain this change and its contribution to Leishmania resistance. The present review firstly covers the actual knowledge on the metabolism of antimonial drugs, the mechanisms of their transmembrane transport and intracellular processing in Leishmania. It further describes both the functional and molecular changes associated with Sb resistance in this organism. Possible transport models based on the actual knowledge are then presented, as well as their functional implications. Biophysical and pharmacological strategies are finally proposed for the precise identification of the transport pathways.
Collapse
Affiliation(s)
- Frédéric Frézard
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Av. Antônio Carlos 6627, Pampulha, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| | - Rubens Monte-Neto
- Centre de Recherche en Infectiologie du Centre Hospitalier de l'Université Laval, 2705, Boulevard Laurier, RC-709, G1V 4G2, Québec, QC, Canada
| | - Priscila G Reis
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Av. Antônio Carlos 6627, Pampulha, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
303
|
de Jesus JB, Mesquita-Rodrigues C, Cuervo P. Proteomics advances in the study of Leishmania parasites and leishmaniasis. Subcell Biochem 2014; 74:323-349. [PMID: 24264252 DOI: 10.1007/978-94-007-7305-9_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Leishmania spp. are digenetic parasites which cause a broad spectrum of fatal diseases in humans. These parasites, as well as the other trypanosomatid, regulate gene expression at the post-transcriptional and post-translational levels, so that a poor correlation is observed between mRNA content and translated proteins. The completion of the genomic sequencing of several Leishmania species has enormous relevance to the study of the leishmaniasis pathogenesis. The combination of the available genomic resources of these parasites with powerful high-throughput proteomic analysis has shed light on various aspects of Leishmania biology as well as on the mechanisms underlying the disease. Diverse proteomic approaches have been used to describe and catalogue global protein profiles of Leishmania spp., reveal changes in protein expression during development, determine the subcellular localization of gene products, evaluate host-parasite interactions and elucidate drug resistance mechanisms. The characterization of these proteins has advanced, although many fundamental questions remain unanswered. Here, we present a historic review summarizing the different proteomic technologies applied to the study of Leishmania parasites during the last decades and we discuss the proteomic discoveries that have contributed to the understanding of Leishmania parasites biology and leishmaniasis.
Collapse
Affiliation(s)
- Jose Batista de Jesus
- Departamento de Engenharia de Biossistemas, Universidade Federal de São João Del Rei, São João Del Rei, MG, Brazil,
| | | | | |
Collapse
|
304
|
Pharmacological assessment defines Leishmania donovani casein kinase 1 as a drug target and reveals important functions in parasite viability and intracellular infection. Antimicrob Agents Chemother 2013; 58:1501-15. [PMID: 24366737 DOI: 10.1128/aac.02022-13] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Protein kinase inhibitors have emerged as new drugs in various therapeutic areas, including leishmaniasis, an important parasitic disease. Members of the Leishmania casein kinase 1 (CK1) family represent promising therapeutic targets. Leishmania casein kinase 1 isoform 2 (CK1.2) has been identified as an exokinase capable of phosphorylating host proteins, thus exerting a potential immune-suppressive action on infected host cells. Moreover, its inhibition reduces promastigote growth. Despite these important properties, its requirement for intracellular infection and its chemical validation as a therapeutic target in the disease-relevant amastigote stage remain to be established. In this study, we used a multidisciplinary approach combining bioinformatics, biochemical, and pharmacological analyses with a macrophage infection assay to characterize and define Leishmania CK1.2 as a valid drug target. We show that recombinant and transgenic Leishmania CK1.2 (i) can phosphorylate CK1-specific substrates, (ii) is sensitive to temperature, and (iii) is susceptible to CK1-specific inhibitors. CK1.2 is constitutively expressed at both the promastigote insect stage and the vertebrate amastigote stage. We further demonstrated that reduction of CK1 activity by specific inhibitors, such as D4476, blocks promastigote growth, strongly compromises axenic amastigote viability, and decreases the number of intracellular Leishmania donovani and L. amazonensis amastigotes in infected macrophages. These results underline the potential role of CK1 kinases in intracellular survival. The identification of differences in structure and inhibition profiles compared to those of mammalian CK1 kinases opens new opportunities for Leishmania CK1.2 antileishmanial drug development. Our report provides the first chemical validation of Leishmania CK1 protein kinases, required for amastigote intracellular survival, as therapeutic targets.
Collapse
|
305
|
Pope SM, Lässer C. Toxoplasma gondii infection of fibroblasts causes the production of exosome-like vesicles containing a unique array of mRNA and miRNA transcripts compared to serum starvation. J Extracell Vesicles 2013; 2:22484. [PMID: 24363837 PMCID: PMC3862870 DOI: 10.3402/jev.v2i0.22484] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/30/2013] [Accepted: 10/11/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Until recently thought to be of little significance unless occurring during pregnancy, Toxoplasma gondii infection of human hosts is now known to play a larger role in mental health and is a growing concern in the health care community. We sought to elucidate a possible mechanism by which Toxoplasma infection may cause some of the behavioural pathology now associated with infection. We hypothesized that exosomes may be playing a role. METHODS We utilized electron microscopy to detect the presence and size of extracellular vesicles in the supernatants of Toxoplasma-infected human foreskin fibroblasts (HFF). We then utilized microarray analysis to discern mRNA and miRNA content of the vesicles isolated from supernatants of Toxoplasma-infected (Toxo) and serum-starved (SS) HFF. RESULTS We recovered extracellular vesicles with a size consistent with exosomes that we called exosome-like vesicles (ELVs) from the supernatants of SS and Toxo cultures. The mRNA and miRNA content of these ELVs was highly regulated creating specific and unique expression profiles comparing Toxo ELVs, SS ELVs and RNA isolated from whole cell homogenates. Interestingly, among the most enriched mRNA isolated from ELVs of Toxo cells are 4 specific mRNA species that have been described in the literature as having neurologic activity: Rab-13, eukaryotic translation elongation factor 1 alpha 1, thymosin beta 4 and LLP homolog. In addition, miRNA species uniquely expressed in Toxo ELVs include miR-23b, a well-known regulator of IL-17. CONCLUSION While the production of ELVs containing mRNAs that modify behaviour are consistent with reported Toxoplasma pathology, the mechanism of enrichment and ultimate in vivo effect of these mRNA and miRNA containing ELVs remains to be investigated.
Collapse
Affiliation(s)
- Samuel M Pope
- Department of Biomedical Sciences, Marian University College of Osteopathic Medicine, Indianapolis IN, USA
| | - Cecilia Lässer
- Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
306
|
Dan-Goor M, Nasereddin A, Jaber H, Jaffe CL. Identification of a secreted casein kinase 1 in Leishmania donovani: effect of protein over expression on parasite growth and virulence. PLoS One 2013; 8:e79287. [PMID: 24260187 PMCID: PMC3829951 DOI: 10.1371/journal.pone.0079287] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/25/2013] [Indexed: 12/14/2022] Open
Abstract
Casein kinase 1 (CK1) plays an important role in eukaryotic signaling pathways, and their substrates include key regulatory proteins involved in cell differentiation, proliferation and chromosome segregation. The Leishmania genome encodes six potential CK1 isoforms, of which five have orthologs in other trypanosomatidae. Leishmania donovani CK1 isoform 4 (Ldck1.4, orthologous to LmjF27.1780) is unique to Leishmania and contains a putative secretion signal peptide. The full-length gene and three shorter constructs were cloned and expressed in E. coli as His-tag proteins. Only the full-length 62.3 kDa protein showed protein kinase activity indicating that the N-terminal and C-terminal domains are essential for protein activity. LdCK1.4-FLAG was stably over expressed in L. donovani, and shown by immunofluorescence to be localized primarily in the cytosol. Western blotting using anti-FLAG and anti-CK1.4 antibodies showed that this CK1 isoform is expressed and secreted by promastigotes. Over expression of LdCK1.4 had a significant effect on promastigote growth in culture with these parasites growing to higher cell densities than the control parasites (wild-type or Ld:luciferase, P<0.001). Analysis by flow cytometry showed a higher percentage, ∼4-5-fold, of virulent metacyclic promastigotes on day 3 among the LdCK1.4 parasites. Finally, parasites over expressing LdCK1.4 gave significantly higher infections of mouse peritoneal macrophages compared to wild-type parasites, 28.6% versus 6.3%, respectively (p = 0.0005). These results suggest that LdCK1.4 plays an important role in parasite survival and virulence. Further studies are needed to validate CK1.4 as a therapeutic target in Leishmania.
Collapse
Affiliation(s)
- Mary Dan-Goor
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, National Center for Leishmaniasis, IMRIC, Hebrew University–Hadassah Medical School, Jerusalem, Israel
| | - Abedelmajeed Nasereddin
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, National Center for Leishmaniasis, IMRIC, Hebrew University–Hadassah Medical School, Jerusalem, Israel
| | - Hanan Jaber
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, National Center for Leishmaniasis, IMRIC, Hebrew University–Hadassah Medical School, Jerusalem, Israel
| | - Charles L. Jaffe
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, National Center for Leishmaniasis, IMRIC, Hebrew University–Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
307
|
Gerbaba TK, Gedamu L. Cathepsin B gene disruption induced Leishmania donovani proteome remodeling implies cathepsin B role in secretome regulation. PLoS One 2013; 8:e79951. [PMID: 24244582 PMCID: PMC3828211 DOI: 10.1371/journal.pone.0079951] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 09/29/2013] [Indexed: 11/29/2022] Open
Abstract
Leishmania cysteine proteases are potential vaccine candidates and drug targets. To study the role of cathepsin B cysteine protease, we have generated and characterized cathepsin B null mutant L. donovani parasites. L. donovani cathepsin B null mutants grow normally in culture, but they show significantly attenuated virulence inside macrophages. Quantitative proteome profiling of wild type and null mutant parasites indicates cathepsin B disruption induced remodeling of L. donovani proteome. We identified 83 modulated proteins, of which 65 are decreased and 18 are increased in the null mutant parasites, and 66% (55/83) of the modulated proteins are L. donovani secreted proteins. Proteins involved in oxidation-reduction (trypanothione reductase, peroxidoxins, tryparedoxin, cytochromes) and translation (ribosomal proteins) are among those decreased in the null mutant parasites, and most of these proteins belong to the same complex network of proteins. Our results imply virulence role of cathepsin B via regulation of Leishmania secreted proteins.
Collapse
Affiliation(s)
- Teklu Kuru Gerbaba
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Lashitew Gedamu
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
308
|
Figuera L, Gómez-Arreaza A, Avilán L. Parasitism in optima forma: exploiting the host fibrinolytic system for invasion. Acta Trop 2013; 128:116-23. [PMID: 23850506 DOI: 10.1016/j.actatropica.2013.06.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 06/20/2013] [Accepted: 06/30/2013] [Indexed: 02/08/2023]
Abstract
The interaction of pathogenic bacteria with the host fibrinolytic system through the plasminogen molecule has been well documented. It has been shown, using animal models, to be important in invasion into the host and establishment of the infection. From a number of recent observations with parasitic protists and helminths, emerges evidence that also in these organisms the interaction with plasminogen may be important for infection and virulence. A group of molecules that act as plasminogen receptors have been identified in parasites. This group comprises the glycolytic enzymes enolase, glyceraldehyde-3-phosphate dehydrogenase and fructose-1,6-biphosphate aldolase, in common with the plasminogen receptors known in prokaryotic pathogens. The interaction with the fibrinolytic system may arm the parasites with the host protease plasmin, thus helping them to migrate and cross barriers, infect cells and avoid clot formation. In this context, plasminogen receptors on the parasite surface or as secreted molecules, may be considered virulence factors. A possible evolutionary scenario for the recruitment of glycolytic enzymes as plasminogen receptors by widely different pathogens is discussed.
Collapse
|
309
|
Ghosh J, Bose M, Roy S, Bhattacharyya SN. Leishmania donovani targets Dicer1 to downregulate miR-122, lower serum cholesterol, and facilitate murine liver infection. Cell Host Microbe 2013; 13:277-88. [PMID: 23498953 PMCID: PMC3605572 DOI: 10.1016/j.chom.2013.02.005] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 11/20/2012] [Accepted: 02/08/2013] [Indexed: 12/21/2022]
Abstract
Leishmania donovani causes visceral leishmaniasis (VL) where the parasite infects and resides inside liver and spleen tissue macrophages. Given the abnormal lipid profile observed in VL patients, we examined the status of serum lipids in an experimental murine model of VL. The murine VL liver displayed altered expression of lipid metabolic genes, many of which are direct or indirect targets of the liver-specific microRNA-122. Concomitant reduction of miR-122 expression was observed in VL liver. High serum cholesterol caused resistance to L. donovani infection, while downregulation of miR-122 is coupled with low serum cholesterol in VL mice. Exosomes secreted by the infective parasites caused reduction in miR-122 activity in hepatic cells. Leishmania surface glycoprotein gp63, a Zn-metalloprotease, targets pre-miRNA processor Dicer1 to prevent miRNP formation in L. donovani-interacting hepatic cells. Conversely, restoration of miR-122 or Dicer1 levels in VL mouse liver increased serum cholesterol and reduced liver parasite burden.
Collapse
Affiliation(s)
- June Ghosh
- RNA Biology Research Laboratory, Molecular and Human Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | | | | | | |
Collapse
|
310
|
Barteneva NS, Maltsev N, Vorobjev IA. Microvesicles and intercellular communication in the context of parasitism. Front Cell Infect Microbiol 2013; 3:49. [PMID: 24032108 PMCID: PMC3764926 DOI: 10.3389/fcimb.2013.00049] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 08/20/2013] [Indexed: 01/18/2023] Open
Abstract
There is a rapidly growing body of evidence that production of microvesicles (MVs) is a universal feature of cellular life. MVs can incorporate microRNA (miRNA), mRNA, mtDNA, DNA and retrotransposons, camouflage viruses/viral components from immune surveillance, and transfer cargo between cells. These properties make MVs an essential player in intercellular communication. Increasing evidence supports the notion that MVs can also act as long-distance vehicles for RNA molecules and participate in metabolic synchronization and reprogramming eukaryotic cells including stem and germinal cells. MV ability to carry on DNA and their general distribution makes them attractive candidates for horizontal gene transfer, particularly between multi-cellular organisms and their parasites; this suggests important implications for the co-evolution of parasites and their hosts. In this review, we provide current understanding of the roles played by MVs in intracellular pathogens and parasitic infections. We also discuss the possible role of MVs in co-infection and host shifting.
Collapse
Affiliation(s)
- Natasha S Barteneva
- Program in Cellular and Molecular Medicine, Children's Hospital Boston and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA. Natasha.Barteneva@ childrens.harvard.edu
| | | | | |
Collapse
|
311
|
Microvesicles and exosomes as vehicles between protozoan and host cell communication. Biochem Soc Trans 2013; 41:252-7. [PMID: 23356292 DOI: 10.1042/bst20120217] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cells release extracellular vesicles in response to external factors or in a physiological way. Microvesicles and exosomes originate in cells in different ways and, depending on their contents, may have multiple biological effects on other cells and the environment. The host cell-parasite relationship could be changed dramatically by the plasticity of a new type of communication through extracellular vesicles. In the present paper, we discuss how protozoans use this new resource to evade the immune system and establish infection.
Collapse
|
312
|
Twu O, de Miguel N, Lustig G, Stevens GC, Vashisht AA, Wohlschlegel JA, Johnson PJ. Trichomonas vaginalis exosomes deliver cargo to host cells and mediate host∶parasite interactions. PLoS Pathog 2013; 9:e1003482. [PMID: 23853596 PMCID: PMC3708881 DOI: 10.1371/journal.ppat.1003482] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 05/23/2013] [Indexed: 12/21/2022] Open
Abstract
Trichomonas vaginalis is a common sexually transmitted parasite that colonizes the human urogential tract where it remains extracellular and adheres to epithelial cells. Infections range from asymptomatic to highly inflammatory, depending on the host and the parasite strain. Here, we use a combination of methodologies including cell fractionation, immunofluorescence and electron microscopy, RNA, proteomic and cytokine analyses and cell adherence assays to examine pathogenic properties of T. vaginalis. We have found that T.vaginalis produces and secretes microvesicles with physical and biochemical properties similar to mammalian exosomes. The parasite-derived exosomes are characterized by the presence of RNA and core, conserved exosomal proteins as well as parasite-specific proteins. We demonstrate that T. vaginalis exosomes fuse with and deliver their contents to host cells and modulate host cell immune responses. Moreover, exosomes from highly adherent parasite strains increase the adherence of poorly adherent parasites to vaginal and prostate epithelial cells. In contrast, exosomes from poorly adherent strains had no measurable effect on parasite adherence. Exosomes from parasite strains that preferentially bind prostate cells increased binding of parasites to these cells relative to vaginal cells. In addition to establishing that parasite exosomes act to modulate host∶parasite interactions, these studies are the first to reveal a potential role for exosomes in promoting parasite∶parasite communication and host cell colonization. Trichomoniasis, the most common non-viral sexually transmitted disease worldwide, infects over 275 million people annually. Infection results from the colonization of the human urogenital tract by the parasite Trichomonas vaginalis. To establish and maintain infection the parasite adheres to host cells, a process that is poorly understood. Here, we show that T. vaginalis secretes small vesicles called exosomes that are capable of fusing with and delivering their contents to host cells. Parasite exosomes were found to induce changes in the host cell and to mediate the interaction of T. vaginalis with host by increasing the adherence of the parasite to host cells. Exosomes have been primarily studied in mammalian cells where they have been shown to mediate intercellular communication and have been implicated in processes including development, antigen presentation and cancer metastasis. Our data extend the function of exosomes to mediating host∶parasite interactions, cellular communication between two species and promoting colonization of an extracellular parasite. Research on T. vaginalis exosomes holds the potential for developing applications that would allow exosomes to be used in detecting and diagnosing trichomoniasis and for targeting drugs to the site of infection.
Collapse
Affiliation(s)
- Olivia Twu
- Molecular Biology Institute, University of California, Los Angeles, California, United States of America
| | - Natalia de Miguel
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States of America
- IIB-INTECH, CONICET-UNSAM, Camino de Circunvalación Laguna Km. 6, Buenos Aires, Argentina
| | - Gila Lustig
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States of America
| | - Grant C. Stevens
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States of America
| | - Ajay A. Vashisht
- Department of Biological Chemistry, University of California, Los Angeles, California, United States of America
| | - James A. Wohlschlegel
- Molecular Biology Institute, University of California, Los Angeles, California, United States of America
- Department of Biological Chemistry, University of California, Los Angeles, California, United States of America
| | - Patricia J. Johnson
- Molecular Biology Institute, University of California, Los Angeles, California, United States of America
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
313
|
Queiroz RML, Charneau S, Motta FN, Santana JM, Roepstorff P, Ricart CAO. Comprehensive proteomic analysis of Trypanosoma cruzi epimastigote cell surface proteins by two complementary methods. J Proteome Res 2013; 12:3255-63. [PMID: 23682730 DOI: 10.1021/pr400110h] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Trypanosoma cruzi is a protozoan that causes Chagas' disease, a neglected infectious illness that affects millions of people, mostly in Latin America. Here, the cell surface subproteome of the T. cruzi epimastigote life form was characterized. In order to prepare samples enriched in epimastigote plasma membrane protein, two distinct methodologies were optimized and evaluated. The first methodology was based on cell surface trypsinization (Shave) of intact living cells while the second approach used biotinylation of cell surface proteins followed by streptavidin affinity chromatography isolation of the labeled proteins. Both T. cruzi subproteomes were analyzed by LC-MS/MS. The results showed that the methodologies offered comprehensive and complementary information about the parasite's plasma membrane subproteome.
Collapse
Affiliation(s)
- Rayner M L Queiroz
- Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, CEP 70910-900, Brazil
| | | | | | | | | | | |
Collapse
|
314
|
Santarém N, Racine G, Silvestre R, Cordeiro-da-Silva A, Ouellette M. Exoproteome dynamics in Leishmania infantum. J Proteomics 2013; 84:106-18. [DOI: 10.1016/j.jprot.2013.03.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 03/14/2013] [Accepted: 03/20/2013] [Indexed: 12/14/2022]
|
315
|
Where do they come from and where do they go: candidates for regulating extracellular vesicle formation in fungi. Int J Mol Sci 2013; 14:9581-603. [PMID: 23644887 PMCID: PMC3676800 DOI: 10.3390/ijms14059581] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 04/11/2013] [Accepted: 04/17/2013] [Indexed: 01/23/2023] Open
Abstract
In the past few years, extracellular vesicles (EVs) from at least eight fungal species were characterized. EV proteome in four fungal species indicated putative biogenesis pathways and suggested interesting similarities with mammalian exosomes. Moreover, as observed for mammalian exosomes, fungal EVs were demonstrated to be immunologically active. Here we review the seminal and most recent findings related to the production of EVs by fungi. Based on the current literature about secretion of fungal molecules and biogenesis of EVs in eukaryotes, we focus our discussion on a list of cellular proteins with the potential to regulate vesicle biogenesis in the fungi.
Collapse
|
316
|
Attenuation of Leishmania infantum chagasi metacyclic promastigotes by sterol depletion. Infect Immun 2013; 81:2507-17. [PMID: 23630964 DOI: 10.1128/iai.00214-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The infectious metacyclic promastigotes of Leishmania protozoa establish infection in a mammalian host after they are deposited into the dermis by a sand fly vector. Several Leishmania virulence factors promote infection, including the glycosylphosphatidylinositol membrane-anchored major surface protease (MSP). Metacyclic Leishmania infantum chagasi promastigotes were treated with methyl-beta-cyclodextrin (MβCD), a sterol-chelating reagent, causing a 3-fold reduction in total cellular sterols as well as enhancing MSP release without affecting parasite viability in vitro. MβCD-treated promastigotes were more susceptible to complement-mediated lysis than untreated controls and reduced the parasite load 3-fold when inoculated into BALB/c mice. Paradoxically, MβCD-treated promastigotes caused a higher initial in vitro infection rate in human or murine macrophages than untreated controls, although their intracellular multiplication was hindered upon infection establishment. There was a corresponding larger amount of covalently bound C3b than iC3b on the parasite surfaces of MβCD-treated promastigotes exposed to healthy human serum in vitro, as well as loss of MSP, a protease that enhances C3b cleavage to iC3b. Mass spectrometry showed that MβCD promotes the release of proteins into the extracellular medium, including both MSP and MSP-like protein (MLP), from virulent metacyclic promastigotes. These data support the hypothesis that plasma membrane sterols are important for the virulence of Leishmania protozoa at least in part through retention of membrane virulence proteins.
Collapse
|
317
|
Barteneva NS, Fasler-Kan E, Bernimoulin M, Stern JNH, Ponomarev ED, Duckett L, Vorobjev IA. Circulating microparticles: square the circle. BMC Cell Biol 2013; 14:23. [PMID: 23607880 PMCID: PMC3651414 DOI: 10.1186/1471-2121-14-23] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 03/20/2013] [Indexed: 01/05/2023] Open
Abstract
Background The present review summarizes current knowledge about microparticles (MPs) and provides a systematic overview of last 20 years of research on circulating MPs, with particular focus on their clinical relevance. Results MPs are a heterogeneous population of cell-derived vesicles, with sizes ranging between 50 and 1000 nm. MPs are capable of transferring peptides, proteins, lipid components, microRNA, mRNA, and DNA from one cell to another without direct cell-to-cell contact. Growing evidence suggests that MPs present in peripheral blood and body fluids contribute to the development and progression of cancer, and are of pathophysiological relevance for autoimmune, inflammatory, infectious, cardiovascular, hematological, and other diseases. MPs have large diagnostic potential as biomarkers; however, due to current technological limitations in purification of MPs and an absence of standardized methods of MP detection, challenges remain in validating the potential of MPs as a non-invasive and early diagnostic platform. Conclusions Improvements in the effective deciphering of MP molecular signatures will be critical not only for diagnostics, but also for the evaluation of treatment regimens and predicting disease outcomes.
Collapse
Affiliation(s)
- Natasha S Barteneva
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, D-249, 200 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
318
|
Lynn MA, Marr AK, McMaster WR. Differential quantitative proteomic profiling of Leishmania infantum and Leishmania mexicana density gradient separated membranous fractions. J Proteomics 2013; 82:179-92. [PMID: 23466312 DOI: 10.1016/j.jprot.2013.02.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 02/08/2013] [Accepted: 02/09/2013] [Indexed: 12/22/2022]
Abstract
UNLABELLED Leishmaniasis, caused by infection with Leishmania, is a major public health concern affecting more than 20million people globally. Leishmania has a digenetic lifecycle consisting of an extracellular flagellated promastigote, adapted to live in the mid-gut of the sand fly host and an aflagellated intracellular amastigote that resides within the macrophage of the mammalian host. Leishmania mexicana and Leishmania infantum are causative agents of cutaneous and visceral leishmaniasis, respectively. Membrane proteins play a pivotal role in host-pathogen interactions and in regulatory pathways. As the genome of Leishmania is essentially constitutively expressed, regulation of protein expression during differentiation occurs post-transcriptionally and/or post-translationally. Quantitative mass spectrometry using iTRAQ labeling identified differences in the proteomes of density gradient separated membranous fractions of promastigote and amastigote life-stages. We identified 189 L. infantum and 107 L. mexicana non-redundant proteins of which 20-40% showed differential expression levels between promastigote and amastigote lifecycle stages. Differentially expressed proteins mapped to several pathways including cell motility, metabolism, and infectivity as well as virulence factors such as eEF-1α, amastin and leishmanolysin (GP63). Western blot analysis validated iTRAQ quantitation for leishmanolysin. Focusing on differentially expressed proteins essential for pathogenesis, may ultimately lead to the identification of novel potential therapeutic targets. BIOLOGICAL SIGNIFICANCE Leishmania, protozoan parasites of the Trypanosomatidae family, are the causative agents of leishmaniasis that represents a major public health concern affecting more than 20million people globally Membrane associated proteins play a pivotal role in host-pathogen interactions and in regulatory pathways. Quantitative proteomic analysis of the membranous fractions from L. mexicana and L. infantum (causative agents of cutaneous and visceral leishmaniasis, respectively) identified a number of proteins that may have important stage-specific functions in either the sand fly or mammalian host. The function of these proteins includes roles in virulence, as well as differences in metabolic process between life stages. Many of the proteins identified may act as virulence factors playing significant roles in parasite invasion, host-parasite interaction or parasite survival and thus may have therapeutic potential as drug target candidates.
Collapse
Affiliation(s)
- Miriam A Lynn
- Infection and Immunity Research Centre, Vancouver Coastal Health Research Institute, 2660 Oak Street, Vancouver, B.C., V6H 3Z6, Canada
| | | | | |
Collapse
|
319
|
Fernandes AC, Soares DC, Saraiva EM, Meyer-Fernandes JR, Souto-Padrón T. Different secreted phosphatase activities inLeishmania amazonensis. FEMS Microbiol Lett 2013; 340:117-28. [DOI: 10.1111/1574-6968.12080] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/03/2013] [Accepted: 01/07/2013] [Indexed: 12/21/2022] Open
Affiliation(s)
- Anne C.S. Fernandes
- Instituto de Microbiologia Paulo de Góes; Centro de Ciências da Saúde; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brasil
| | - Deivid C. Soares
- Instituto de Microbiologia Paulo de Góes; Centro de Ciências da Saúde; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brasil
| | - Elvira M. Saraiva
- Instituto de Microbiologia Paulo de Góes; Centro de Ciências da Saúde; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brasil
| | - José R. Meyer-Fernandes
- Instituto de Bioquímica Médica; Centro de Ciências da Saúde; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brasil
| | - Thaïs Souto-Padrón
- Instituto de Microbiologia Paulo de Góes; Centro de Ciências da Saúde; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brasil
| |
Collapse
|
320
|
Chiu KH, Chang YH, Liao PC. Secretome analysis using a hollow fiber culture system for cancer biomarker discovery. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2285-92. [PMID: 23376430 DOI: 10.1016/j.bbapap.2013.01.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 12/30/2012] [Accepted: 01/24/2013] [Indexed: 12/22/2022]
Abstract
Secreted proteins, collectively referred to as the secretome, were suggested as valuable biomarkers in disease diagnosis and prognosis. However, some secreted proteins from cell cultures are difficult to detect because of their intrinsically low abundance; they are frequently masked by the released proteins from lysed cells and the substantial amounts of serum proteins used in culture medium. The hollow fiber culture (HFC) system is a commercially available system composed of small fibers sealed in a cartridge shell; cells grow on the outside of the fiber. Recently, because this system can help cells grow at a high density, it has been developed and applied in a novel analytical platform for cell secretome collection in cancer biomarker discovery. This article focuses on the advantages of the HFC system, including the effectiveness of the system for collection of secretomes, and reviews the process of cell secretome collection by the HFC system and proteomic approaches to discover cancer biomarkers. The HFC system not only provides a high-density three-dimensional (3D) cell culture system to mimic tumor growth conditions in vivo but can also accommodate numerous cells in a small volume, allowing secreted proteins to be accumulated and concentrated. In addition, cell lysis rates can be greatly reduced, decreasing the amount of contamination by abundant cytosolic proteins from lysed cells. Therefore, the HFC system is useful for preparing a wide range of proteins from cell secretomes and provides an effective method for collecting higher amounts of secreted proteins from cancer cells. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
Affiliation(s)
- Kuo-Hsun Chiu
- Department and Graduate Institute of Aquaculture, National Kaohsiung Marine University, Kaohsiung, Taiwan
| | | | | |
Collapse
|
321
|
Bayer-Santos E, Aguilar-Bonavides C, Rodrigues SP, Cordero EM, Marques AF, Varela-Ramirez A, Choi H, Yoshida N, da Silveira JF, Almeida IC. Proteomic Analysis of Trypanosoma cruzi Secretome: Characterization of Two Populations of Extracellular Vesicles and Soluble Proteins. J Proteome Res 2013; 12:883-97. [DOI: 10.1021/pr300947g] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Ethel Bayer-Santos
- Departamento de Microbiologia,
Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, SP 04023-062, Brazil
| | - Clemente Aguilar-Bonavides
- The Border Biomedical Research
Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas 79968, United States
- Computational Science Program,
The Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Silas Pessini Rodrigues
- The Border Biomedical Research
Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Esteban Maurício Cordero
- The Border Biomedical Research
Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Alexandre Ferreira Marques
- The Border Biomedical Research
Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Armando Varela-Ramirez
- The Border Biomedical Research
Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Hyungwon Choi
- Saw Swee Hock School of Public
Health, National University of Singapore, Singapore
| | - Nobuko Yoshida
- Departamento de Microbiologia,
Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, SP 04023-062, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia,
Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, SP 04023-062, Brazil
| | - Igor C. Almeida
- The Border Biomedical Research
Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas 79968, United States
| |
Collapse
|
322
|
Weber SS, Parente AFA, Borges CL, Parente JA, Bailão AM, de Almeida Soares CM. Analysis of the secretomes of Paracoccidioides mycelia and yeast cells. PLoS One 2012; 7:e52470. [PMID: 23272246 PMCID: PMC3525554 DOI: 10.1371/journal.pone.0052470] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 11/13/2012] [Indexed: 11/19/2022] Open
Abstract
Paracoccidioides, a complex of several phylogenetic species, is the causative agent of paracoccidioidomycosis. The ability of pathogenic fungi to develop a multifaceted response to the wide variety of stressors found in the host environment is important for virulence and pathogenesis. Extracellular proteins represent key mediators of the host-parasite interaction. To analyze the expression profile of the proteins secreted by Paracoccidioides, Pb01 mycelia and yeast cells, we used a proteomics approach combining two-dimensional electrophoresis with matrix-assisted laser desorption ionization quadrupole time-of-flight mass spectrometry (MALDI-Q-TOF MS/MS). From three biological replicates, 356 and 388 spots were detected, in mycelium and yeast cell secretomes, respectively. In this study, 160 non-redundant proteins/isoforms were indentified, including 30 and 24 proteins preferentially secreted in mycelia and yeast cells, respectively. In silico analyses revealed that 65% of the identified proteins/isoforms were secreted primarily via non-conventional pathways. We also investigated the influence of protein export inhibition in the phagocytosis of Paracoccidioides by macrophages. The addition of Brefeldin A to the culture medium significantly decreased the production of secreted proteins by both Paracoccidioides and internalized yeast cells by macrophages. In contrast, the addition of concentrated culture supernatant to the co-cultivation significantly increased the number of internalized yeast cells by macrophages. Importantly, the proteins detected in the fungal secretome were also identified within macrophages. These results indicate that Paracoccidioides extracellular proteins are important for the fungal interaction with the host.
Collapse
Affiliation(s)
- Simone Schneider Weber
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Ana Flávia Alves Parente
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Clayton Luiz Borges
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Juliana Alves Parente
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Alexandre Melo Bailão
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Célia Maria de Almeida Soares
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
- * E-mail:
| |
Collapse
|
323
|
Olivier M, Atayde VD, Isnard A, Hassani K, Shio MT. Leishmania virulence factors: focus on the metalloprotease GP63. Microbes Infect 2012; 14:1377-89. [DOI: 10.1016/j.micinf.2012.05.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 05/16/2012] [Accepted: 05/28/2012] [Indexed: 12/20/2022]
|
324
|
Abstract
Membrane vesicles secreted by Leishmania mexicana were collected and analyzed. These vesicles can bind plasminogen and were shown to contain enolase, previously identified as a plasminogen-binding protein. In addition, another plasminogen-binding protein was identified, the small myristoylated protein, SMP-1. Recombinant SMP-1 was able to bind plasminogen in a lysine-dependent manner with a K(d) value of 0.24 μM. The C-terminal lysine seems to be responsible for this binding, since this recognition decreases upon carboxypeptidase B treatment. This protein was present within the secreted membrane vesicles as demonstrated by its protection from trypsin digestion in the absence of Triton X-100. Plasminogen-binding proteins in the secreted vesicles may be involved in parasite invasion in the mammalian host.
Collapse
|
325
|
Cryptococcus neoformans-derived microvesicles enhance the pathogenesis of fungal brain infection. PLoS One 2012; 7:e48570. [PMID: 23144903 PMCID: PMC3492498 DOI: 10.1371/journal.pone.0048570] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 09/27/2012] [Indexed: 12/22/2022] Open
Abstract
Cryptococcal meningoencephalitis is the most common fungal disease in the central nervous system. The mechanisms by which Cryptococcus neoformans invades the brain are largely unknown. In this study, we found that C. neoformans-derived microvesicles (CnMVs) can enhance the traversal of the blood-brain barrier (BBB) by C. neoformans invitro. The immunofluorescence imaging demonstrates that CnMVs can fuse with human brain microvascular endothelial cells (HBMECs), the constituents of the BBB. This activity is presumably due to the ability of the CnMVs to activate HBMEC membrane rafts and induce cell fusogenic activity. CnMVs also enhanced C. neoformans infection of the brain, found in both infected brains and cerebrospinal fluid. In infected mouse brains, CnMVs are distributed inside and around C. neoformans-induced cystic lesions. GFAP (glial fibrillary acidic protein)-positive astrocytes were found surrounding the cystic lesions, overlapping with the 14-3-3-GFP (14-3-3-green fluorescence protein fusion) signals. Substantial changes could be observed in areas that have a high density of CnMV staining. This is the first demonstration that C. neoformans-derived microvesicles can facilitate cryptococcal traversal across the BBB and accumulate at lesion sites of C. neoformans-infected brains. Results of this study suggested that CnMVs play an important role in the pathogenesis of cryptococcal meningoencephalitis.
Collapse
|
326
|
Extracellular vesicles from parasitic helminths contain specific excretory/secretory proteins and are internalized in intestinal host cells. PLoS One 2012; 7:e45974. [PMID: 23029346 PMCID: PMC3454434 DOI: 10.1371/journal.pone.0045974] [Citation(s) in RCA: 264] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 08/23/2012] [Indexed: 11/30/2022] Open
Abstract
The study of host-parasite interactions has increased considerably in the last decades, with many studies focusing on the identification of parasite molecules (i.e. surface or excretory/secretory proteins (ESP)) as potential targets for new specific treatments and/or diagnostic tools. In parallel, in the last few years there have been significant advances in the field of extracellular vesicles research. Among these vesicles, exosomes of endocytic origin, with a characteristic size ranging from 30–100 nm, carry several atypical secreted proteins in different organisms, including parasitic protozoa. Here, we present experimental evidence for the existence of exosome-like vesicles in parasitic helminths, specifically the trematodes Echinostoma caproni and Fasciola hepatica. These microvesicles are actively released by the parasites and are taken up by host cells. Trematode extracellular vesicles contain most of the proteins previously identified as components of ESP, as confirmed by proteomic, immunogold labeling and electron microscopy studies. In addition to parasitic proteins, we also identify host proteins in these structures. The existence of extracellular vesicles explains the secretion of atypical proteins in trematodes, and the demonstration of their uptake by host cells suggests an important role for these structures in host-parasite communication, as described for other infectious agents.
Collapse
|
327
|
Moradin N, Descoteaux A. Leishmania promastigotes: building a safe niche within macrophages. Front Cell Infect Microbiol 2012; 2:121. [PMID: 23050244 PMCID: PMC3445913 DOI: 10.3389/fcimb.2012.00121] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 09/04/2012] [Indexed: 12/16/2022] Open
Abstract
Upon their internalization by macrophages, Leishmania promastigotes inhibit phagolysosome biogenesis. The main factor responsible for this inhibition is the promastigote surface glycolipid lipophosphoglycan (LPG). This glycolipid has a profound impact on the phagosome, causing periphagosomal accumulation of F-actin and disruption of phagosomal lipid microdomains. Functionally, this LPG-mediated inhibition of phagosome maturation is characterized by an impaired assembly of the NADPH oxidase and the exclusion of the vesicular proton-ATPase from phagosomes. In this chapter, we review the current knowledge concerning the nature of the intra-macrophage compartment in which Leishmania donovani promastigotes establish infection. We also describe how LPG enables this parasite to remodel the parasitophorous vacuole.
Collapse
Affiliation(s)
- Neda Moradin
- INRS - Institut Armand-Frappier and Center for Host-Parasite Interactions Laval, QC, Canada
| | | |
Collapse
|
328
|
Torrecilhas AC, Schumacher RI, Alves MJM, Colli W. Vesicles as carriers of virulence factors in parasitic protozoan diseases. Microbes Infect 2012; 14:1465-74. [PMID: 22892602 DOI: 10.1016/j.micinf.2012.07.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 07/06/2012] [Accepted: 07/09/2012] [Indexed: 01/08/2023]
Abstract
Different types of shed vesicles as, for example, exosomes, plasma-membrane-derived vesicles or microparticles, are the focus of intense research in view of their potential role in cell-cell communication and under the perspective that they might be good tools for immunotherapy, vaccination or diagnostic purposes. This review discusses ways employed by pathogenic trypanosomatids to interact with the host by shedding vesicles that contain molecules important for the establishment of infection, as opposed to previous beliefs considering them as a waste of cellular metabolism. Trypanosomatids are compared with Apicomplexa, which circulate parasite antigens bound to vesicles shed by host cells. The knowledge of the origin and chemical composition of these different vesicles might lead to the understanding of the mechanisms that determine their biological function.
Collapse
Affiliation(s)
- Ana Claudia Torrecilhas
- Departamento de Ciências Biológicas, Campus Diadema, Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brazil
| | | | | | | |
Collapse
|
329
|
Yamada T, Inoshima Y, Matsuda T, Ishiguro N. Comparison of methods for isolating exosomes from bovine milk. J Vet Med Sci 2012; 74:1523-5. [PMID: 22785357 DOI: 10.1292/jvms.12-0032] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Four methods were evaluated for isolating exosomes from bovine milk: (1) ExoQuick precipitation, (2) ultracentrifugation with ExoQuick precipitation, (3) ultracentrifugation with density gradient centrifugation, and (4) human milk exosome isolation. Methods 1 and 4 failed due to differences between bovine and human milk. Exosomes were efficiently isolated by ultracentrifugation with either ExoQuick precipitation (method 2) or density gradient centrifugation (method 3). The highest yield of exosomes was achieved using ultracentrifugation with ExoQuick precipitation, whereas higher quality exosome isolation with intact morphological structures was achieved by ultracentrifugation with density gradient centrifugation.
Collapse
Affiliation(s)
- Tetsuya Yamada
- Laboratory of Food and Environmental Hygiene, Department of Veterinary Medicine, Gifu University, Gifu, Japan
| | | | | | | |
Collapse
|
330
|
van der Pol E, Böing AN, Harrison P, Sturk A, Nieuwland R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev 2012; 64:676-705. [PMID: 22722893 DOI: 10.1124/pr.112.005983] [Citation(s) in RCA: 1336] [Impact Index Per Article: 102.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Both eukaryotic and prokaryotic cells release small, phospholipid-enclosed vesicles into their environment. Why do cells release vesicles? Initial studies showed that eukaryotic vesicles are used to remove obsolete cellular molecules. Although this release of vesicles is beneficial to the cell, the vesicles can also be a danger to their environment, for instance in blood, where vesicles can provide a surface supporting coagulation. Evidence is accumulating that vesicles are cargo containers used by eukaryotic cells to exchange biomolecules as transmembrane receptors and genetic information. Because also bacteria communicate to each other via extracellular vesicles, the intercellular communication via extracellular cargo carriers seems to be conserved throughout evolution, and therefore vesicles are likely to be a highly efficient, robust, and economic manner of exchanging information between cells. Furthermore, vesicles protect cells from accumulation of waste or drugs, they contribute to physiology and pathology, and they have a myriad of potential clinical applications, ranging from biomarkers to anticancer therapy. Because vesicles may pass the blood-brain barrier, they can perhaps even be considered naturally occurring liposomes. Unfortunately, pathways of vesicle release and vesicles themselves are also being used by tumors and infectious diseases to facilitate spreading, and to escape from immune surveillance. In this review, the different types, nomenclature, functions, and clinical relevance of vesicles will be discussed.
Collapse
Affiliation(s)
- Edwin van der Pol
- Department of Clinical Chemistry, Academic Medical Centre of the University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
331
|
Deatherage BL, Cookson BT. Membrane vesicle release in bacteria, eukaryotes, and archaea: a conserved yet underappreciated aspect of microbial life. Infect Immun 2012; 80:1948-57. [PMID: 22409932 PMCID: PMC3370574 DOI: 10.1128/iai.06014-11] [Citation(s) in RCA: 529] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Interaction of microbes with their environment depends on features of the dynamic microbial surface throughout cell growth and division. Surface modifications, whether used to acquire nutrients, defend against other microbes, or resist the pressures of a host immune system, facilitate adaptation to unique surroundings. The release of bioactive membrane vesicles (MVs) from the cell surface is conserved across microbial life, in bacteria, archaea, fungi, and parasites. MV production occurs not only in vitro but also in vivo during infection, underscoring the influence of these surface organelles in microbial physiology and pathogenesis through delivery of enzymes, toxins, communication signals, and antigens recognized by the innate and adaptive immune systems. Derived from a variety of organisms that span kingdoms of life and called by several names (membrane vesicles, outer membrane vesicles [OMVs], exosomes, shedding microvesicles, etc.), the conserved functions and mechanistic strategies of MV release are similar, including the use of ESCRT proteins and ESCRT protein homologues to facilitate these processes in archaea and eukaryotic microbes. Although forms of MV release by different organisms share similar visual, mechanistic, and functional features, there has been little comparison across microbial life. This underappreciated conservation of vesicle release, and the resulting functional impact throughout the tree of life, explored in this review, stresses the importance of vesicle-mediated processes throughout biology.
Collapse
Affiliation(s)
| | - Brad T. Cookson
- Department of Microbiology
- Laboratory Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
332
|
Isnard A, Shio MT, Olivier M. Impact of Leishmania metalloprotease GP63 on macrophage signaling. Front Cell Infect Microbiol 2012; 2:72. [PMID: 22919663 PMCID: PMC3417651 DOI: 10.3389/fcimb.2012.00072] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Accepted: 05/03/2012] [Indexed: 11/18/2022] Open
Abstract
The intramacrophage protozoan parasites of Leishmania genus have developed sophisticated ways to subvert the innate immune response permitting their infection and propagation within the macrophages of the mammalian host. Several Leishmania virulence factors have been identified and found to be of importance for the development of leishmaniasis. However, recent findings are now further reinforcing the critical role played by the zinc-metalloprotease GP63 as a virulence factor that greatly influence host cell signaling mechanisms and related functions. GP63 has been found to be involved not only in the cleavage and degradation of various kinases and transcription factors, but also to be the major molecule modulating host negative regulatory mechanisms involving for instance protein tyrosine phosphatases (PTPs). Those latter being well recognized for their pivotal role in the regulation of a great number of signaling pathways. In this review article, we are providing a complete overview about the role of Leishmania GP63 in the mechanisms underlying the subversion of macrophage signaling and functions.
Collapse
Affiliation(s)
- Amandine Isnard
- Faculty of Medicine, Department of Medicine, Microbiology, and Immunology, The Research Institute of the McGill University Health Centre, McGill University Montréal, QC, Canada
| | | | | |
Collapse
|
333
|
SILVA AM, TAVARES J, SILVESTRE R, OUAISSI A, COOMBS GH, CORDEIRO-da-SILVA A. Characterization of Leishmania infantum thiol-dependent reductase 1 and evaluation of its potential to induce immune protection. Parasite Immunol 2012; 34:345-50. [DOI: 10.1111/j.1365-3024.2012.01361.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
334
|
Morrison LS, Goundry A, Faria MS, Tetley L, Eschenlauer SC, Westrop GD, Dostalova A, Volf P, Coombs GH, Lima APCA, Mottram JC. Ecotin-like serine peptidase inhibitor ISP1 of Leishmania major plays a role in flagellar pocket dynamics and promastigote differentiation. Cell Microbiol 2012; 14:1271-86. [PMID: 22486816 PMCID: PMC3440592 DOI: 10.1111/j.1462-5822.2012.01798.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Leishmania ISPs are ecotin-like natural peptide inhibitors of trypsin-family serine peptidases, enzymes that are absent from the Leishmania genome. This led to the proposal that ISPs inhibit host serine peptidases and we have recently shown that ISP2 inhibits neutrophil elastase, thereby enhancing parasite survival in murine macrophages. In this study we show that ISP1 has less serine peptidase inhibitory activity than ISP2, and in promastigotes both are generally located in the cytosol and along the flagellum. However, in haptomonad promastigotes there is a prominent accumulation of ISP1 and ISP2 in the hemidesmosome and for ISP2 on the cell surface. An L. major mutant deficient in all three ISP genes (Δisp1/2/3) was generated and compared with Δisp2/3 mutants to elucidate the physiological role of ISP1. In in vitro cultures, the Δisp1/2/3 mutant contained more haptomonad, nectomonad and leptomonad promastigotes with elongated flagella and reduced motility compared with Δisp2/3 populations, moreover it was characterized by very high levels of release of exosome-like vesicles from the flagellar pocket. These data suggest that ISP1 has a primary role in flagellar homeostasis, disruption of which affects differentiation and flagellar pocket dynamics.
Collapse
Affiliation(s)
- Lesley S Morrison
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
335
|
Hu G, Drescher KM, Chen XM. Exosomal miRNAs: Biological Properties and Therapeutic Potential. Front Genet 2012; 3:56. [PMID: 22529849 PMCID: PMC3330238 DOI: 10.3389/fgene.2012.00056] [Citation(s) in RCA: 273] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 03/27/2012] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs), small non-coding regulatory RNAs that regulate gene expression at the post-transcriptional level, are master regulators of a wide array of cellular processes. Altered miRNA expression could be a determinant of disease development and/or progression and manipulation of miRNA expression represents a potential avenue of therapy. Exosomes are cell-derived extracellular vesicles that promote cell–cell communication and immunoregulatory functions. These “bioactive vesicles” shuttle various molecules, including miRNAs, to recipient cells. Inappropriate release of miRNAs from exosomes may cause significant alterations in biological pathways that affect disease development, supporting the concept that miRNA-containing exosomes could serve as targeted therapies for particular diseases. This review briefly summarizes recent advances in the biology, function, and therapeutic potential of exosomal miRNAs.
Collapse
Affiliation(s)
- Guoku Hu
- Department of Medical Microbiology and Immunology, Creighton University Medical Center Omaha, NE, USA
| | | | | |
Collapse
|
336
|
Bogdan C. Leishmaniasis in rheumatology, haematology and oncology: epidemiological, immunological and clinical aspects and caveats. Ann Rheum Dis 2012; 71 Suppl 2:i60-6. [PMID: 22460140 DOI: 10.1136/annrheumdis-2011-200596] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Leishmaniasis is an intracellular protozoan infection that can lead to cutaneous, mucocutaneous, visceral or systemic manifestations depending on the parasite species and virulence and on the host immune response. It is endemic in countries of Europe (Mediterranean basin), Asia, Africa, Central and South America, but autochthonous cases begin to emerge outside classical disease areas. CD4+ T helper cells, interferon γ, dendritic cells and macrophages are the key components of antileishmanial defence. Leishmaniasis is an important differential diagnosis in patients with chronic lesions of the skin or mucous membranes or with fever, hepatosplenomegaly, lymphadenopathy, pancytopenia, histocytosis, haemophagocytic syndrome or glomerulonephritis. Organ transplant recipients and patients with autoimmune syndromes are at particular risk of developing visceral leishmaniasis following immunosuppressive therapy (eg, with steroids, methotrexate, ciclosporin or tumour necrosis factor-neutralising biological agents). Diagnosis and adequate treatment of leishmaniasis requires the combined use of culture, microscopic and nucleic acid amplication methods and species identification by sequencing and other molecular techniques. Standard regimens for the treatment of visceral leishmaniasis are intravenous liposomal amphotericin B (3 mg/kg body weight for 10 days) or oral miltefosine (150 mg/day for 28 days).
Collapse
Affiliation(s)
- Christian Bogdan
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Wasserturmstrasse 3/5, D-91054 Erlangen, Germany.
| |
Collapse
|
337
|
Lambertz U, Silverman JM, Nandan D, McMaster WR, Clos J, Foster LJ, Reiner NE. Secreted virulence factors and immune evasion in visceral leishmaniasis. J Leukoc Biol 2012; 91:887-99. [PMID: 22442494 DOI: 10.1189/jlb.0611326] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Evasion or subversion of host immune responses is a well-established paradigm in infection with visceralizing leishmania. In this review, we summarize current findings supporting a model in which leishmania target host regulatory molecules and pathways, such as the PTP SHP-1 and the PI3K/Akt signaling cascade, to prevent effective macrophage activation. Furthermore, we describe how virulence factors, secreted by leishmania, interfere with macrophage intracellular signaling. Finally, we discuss mechanisms of secretion and provide evidence that leishmania use a remarkably adept, exosome-based secretion mechanism to export and deliver effector molecules to host cells. In addition to representing a novel mechanism for trafficking of virulence factors across membranes, recent findings indicate that leishmania exosomes may have potential as vaccine candidates.
Collapse
Affiliation(s)
- Ulrike Lambertz
- Department of Medicine Division of Infectious Diseases and the Experimental Medicine Program, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | | | |
Collapse
|
338
|
Rodrigues ML, Nosanchuk JD, Schrank A, Vainstein MH, Casadevall A, Nimrichter L. Vesicular transport systems in fungi. Future Microbiol 2012; 6:1371-81. [PMID: 22082294 DOI: 10.2217/fmb.11.112] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Canonical and unconventional mechanisms of secretion in many eukaryotic cells are relatively well known. In contrast to the situation in animal cells, mechanisms of secretion in fungi must include the capacity for trans-cell wall passage of macromolecules to the extracellular space. Although these mechanisms remain somewhat elusive, several studies in recent years have suggested that vesicular transport is required for trans-cell wall secretion of large molecules. Several fungal molecules, including proteins, lipids, polysaccharides and pigments, are released to the extracellular space in vesicles. In pathogenic fungi, a number of these vesicular components are associated with fungal virulence. Indeed, extracellular vesicles produced by fungi can interfere with the immunomodulatory activity of host cells. Fungal vesicles share many functional aspects with mammalian exosomes and extracellular vesicles produced by bacteria, plants and protozoa, but their cellular origin remains unknown. Here, we discuss the involvement of vesicular transport systems in fungal physiology and pathogenesis, making parallels with the mammalian, bacterial, protozoan and plant cell literature.
Collapse
Affiliation(s)
- Marcio L Rodrigues
- Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Brazil.
| | | | | | | | | | | |
Collapse
|
339
|
Maclean LM, O'Toole PJ, Stark M, Marrison J, Seelenmeyer C, Nickel W, Smith DF. Trafficking and release of Leishmania metacyclic HASPB on macrophage invasion. Cell Microbiol 2012; 14:740-61. [PMID: 22256896 PMCID: PMC3491706 DOI: 10.1111/j.1462-5822.2012.01756.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Proteins of the Leishmania hydrophilic acylated surface protein B (HASPB) family are only expressed in infective parasites (both extra- and intracellular stages) and, together with the peripheral membrane protein SHERP (small hydrophilic endoplasmic reticulum-associated protein), are essential for parasite differentiation (metacyclogenesis) in the sand fly vector. HASPB is a ‘non-classically’ secreted protein, requiring N-terminal acylation for trafficking to and exposure on the plasma membrane. Here, we use live cell imaging methods to further explore this pathway to the membrane and flagellum. Unlike HASPB trafficking in transfected mammalian cells, we find no evidence for a phosphorylation-regulated recycling pathway in metacyclic parasites. Once at the plasma membrane, HASPB18–GFP (green fluorescent protein) can undergo bidirectional movement within the inner leaflet of the membrane and on the flagellum. Transfer of fluorescent protein between the flagellum and the plasma membrane is compromised, however, suggesting the presence of a diffusion barrier at the base of the Leishmania flagellum. Full-length HASPB is released from the metacyclic parasite surface on to macrophages during phagocytosis but while expression is maintained in intracellular amastigotes, HASPB cannot be detected on the external surface in these cells. Thus HASPB may be a dual function protein that is shed by the infective metacyclic but retained internally once Leishmania are taken up by macrophages.
Collapse
Affiliation(s)
- Lorna M Maclean
- Centre for Immunology and Infection, Department of Biology/Hull York Medical School, University of York, York YO10 5DD, UK
| | | | | | | | | | | | | |
Collapse
|
340
|
Innate immune activation and subversion of Mammalian functions by leishmania lipophosphoglycan. J Parasitol Res 2012; 2012:165126. [PMID: 22523640 PMCID: PMC3317186 DOI: 10.1155/2012/165126] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 11/10/2011] [Indexed: 12/19/2022] Open
Abstract
Leishmania promastigotes express several prominent glycoconjugates, either secreted or anchored to the parasite surface. Of these lipophosphoglycan (LPG) is the most abundant, and along with other phosphoglycan-bearing molecules, plays important roles in parasite infectivity and pathogenesis in both the sand fly and the mammalian host. Besides its contribution for parasite survival in the sand fly vector, LPG is important for modulation the host immune responses to favor the establishment of mammalian infection. This review will summarize the current knowledge regarding the role of LPG in Leishmania infectivity, focusing on the interaction of LPG and innate immune cells and in the subversion of mammalian functions by this molecule.
Collapse
|
341
|
Leishmania donovani HslV does not interact stably with HslU proteins. Int J Parasitol 2012; 42:329-39. [PMID: 22370310 DOI: 10.1016/j.ijpara.2012.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 01/10/2012] [Accepted: 01/16/2012] [Indexed: 11/21/2022]
Abstract
Genes for HslVU-type peptidases are found in bacteria and in a few select Eukaryota, among those such important pathogens as Plasmodium spp. and Leishmania spp. In this study, we performed replacements of all three HslV/HslU gene homologues and found one of those, HslV, to be essential for Leishmania donovani viability. The Leishmania HslV gene can also partially relieve the thermosensitive phenotype of a combined HslVU/Lon/ClpXP knockout mutant of Escherichia coli, indicating a conserved function. However, we found that the role and function of the two Leishmania HslU genes has diverged since neither of those interacts stably with HslV. The latter forms a dodecameric complex by itself and shows a punctate distribution. We conclude that whilst the basic function of HslV may be conserved in Leishmania, its organisation and interaction with its canonical complex partner HslU is not. Nevertheless, given the absence of HslV from the proteome of mammals and its essential role in Leishmania viability, HslV is a promising target for intervention.
Collapse
|
342
|
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: 4.8] [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
|
343
|
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: 104] [Impact Index Per Article: 8.0] [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
|
344
|
Silverman JM, Reiner NE. Leishmania exosomes deliver preemptive strikes to create an environment permissive for early infection. Front Cell Infect Microbiol 2012; 1:26. [PMID: 22919591 PMCID: PMC3417360 DOI: 10.3389/fcimb.2011.00026] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 12/23/2011] [Indexed: 01/20/2023] Open
Abstract
Herein, we review evidence supporting a role for Leishmania exosomes during early infection. We suggest a model in which Leishmania secreted microvesicles released into the extracellular milieu deliver effector cargo to host target cells. This cargo mediates immunosuppression and functionally primes host cells for Leishmania invasion. Leishmania ssp. release microvesicles and the amount of vesicle release and the specific protein cargo of the vesicles is sensitive to changes in environmental conditions that mimic infection. Leishmania exosomes influence the phenotype of treated immune cells. For example, wild-type (WT) exosomes attenuate interferon-γ-induced pro-inflammatory cytokine production (TNF-α) by Leishmania-infected monocytes while conversely enhancing production of the anti-inflammatory cytokine IL-10. The Leishmania proteins GP63 and elongation factor-1α (EF-1α) are found in secreted vesicles and are likely important effectors responsible for these changes in phenotype. GP63 and EF-1α access host cell cytosol and activate multiple host protein-tyrosine phosphatases (PTPs). Activation of these PTPs negatively regulates interferon-γ signaling and this prevents effective expression of the macrophage microbicidal arsenal, including TNF-α and nitric oxide. In addition to changing macrophage phenotype, WT vesicles dampen the immune response of monocyte-derived dendritic cells and CD4+ T lymphocytes. This capacity is lost when the protein cargo of the vesicles is modified, specifically when the amount of GP63 and EF-1α in the vesicles is reduced. It appears that exosome delivery of effector proteins results in activation of host PTPs and the negative regulatory effects of the latter creates a pro-parasitic environment. The data suggest that Leishmania exosomes secreted upon initial infection are capable of delivering effector cargo to naïve target cells wherein the cargo primes host cells for infection by interfering with host cell signaling pathways.
Collapse
Affiliation(s)
- Judith Maxwell Silverman
- Brain Research Center, Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
345
|
Puri V, Goyal A, Sankaranarayanan R, Enright AJ, Vaidya T. Evolutionary and functional insights into Leishmania META1: evidence for lateral gene transfer and a role for META1 in secretion. BMC Evol Biol 2011; 11:334. [PMID: 22093578 PMCID: PMC3270026 DOI: 10.1186/1471-2148-11-334] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 11/17/2011] [Indexed: 12/30/2022] Open
Abstract
Background Leishmania META1 has for long been a candidate molecule for involvement in virulence: META1 transcript and protein are up-regulated in metacyclic Leishmania. Yet, how META1 contributes to virulence remains unclear. We sought insights into the possible functions of META1 by studying its evolutionary origins. Results Using multiple criteria including sequence similarity, nucleotide composition, phylogenetic analysis and selection pressure on gene sequence, we present evidence that META1 originated in trypanosomatids as a result of a lateral gene transfer of a bacterial heat-inducible protein, HslJ. Furthermore, within the Leishmania genome, META1 sequence is under negative selection pressure against change/substitution. Using homology modeling of Leishmania META1 based on solved NMR structure of HslJ, we show that META1 and HslJ share a similar structural fold. The best hit for other proteins with similar fold is MxiM, a protein involved in the type III secretion system in Shigella. The striking structural similarity shared by META1, HslJ and MxiM suggests a possibility of shared functions. Upon structural superposition with MxiM, we have observed a putative hydrophobic cavity in META1. Mutagenesis of select hydrophobic residues in this cavity affects the secretion of the secreted acid phosphatase (SAP), indicating META1's involvement in secretory processes in Leishmania. Conclusions Overall, this work uses an evolutionary biology approach, 3D-modeling and site-directed mutagenesis to arrive at new insights into functions of Leishmania META1.
Collapse
Affiliation(s)
- Vidhi Puri
- Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad - 500 007, Andhra Pradesh, India
| | | | | | | | | |
Collapse
|
346
|
Koppen T, Weckmann A, Müller S, Staubach S, Bloch W, Dohmen RJ, Schwientek T. Proteomics analyses of microvesicles released by Drosophila Kc167 and S2 cells. Proteomics 2011; 11:4397-410. [PMID: 21901833 DOI: 10.1002/pmic.201000774] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 08/19/2011] [Accepted: 08/24/2011] [Indexed: 01/30/2023]
Abstract
Distinct types of vesicles are formed in eukaryotic cells that conduct a variable set of functions depending on their origin. One subtype designated circulating microvesicles (MVs) provides a novel form of intercellular communication and recent work suggested the release and uptake of morphogens in vesicles by Drosophila cells. In this study, we have examined cells of the hemocyte-like cell lines Kc167 and S2 and identified secreted vesicles in the culture supernatant. The vesicles were isolated and found to have characteristics comparable to exosomes and plasma membrane MVs released by mammalian cells. In wingless-transfected cells, the full-length protein was detected in the vesicle isolates. Proteomics analyses of the vesicles identified 269 proteins that include various orthologs of marker proteins and proteins with putative functions in vesicle formation and release. Analogous to their mammalian counterparts, the subcellular origin of the vesicular constituents of both cell lines is dominated by membrane-associated and cytosolic proteins with functions that are consistent with their localization in MVs. The analyses revealed a significant overlap of the Kc167 and S2 vesicle proteomes and confirmed a close correlation with non-mammalian and mammalian exosomes.
Collapse
Affiliation(s)
- Tim Koppen
- Center for Biochemistry, Medical Faculty, University of Cologne, Köln, Germany
| | | | | | | | | | | | | |
Collapse
|
347
|
Maia ACRG, Detoni ML, Porcino GN, Soares TV, do Nascimento Gusmão MA, Fessel MR, Marques MJ, Souza MA, Coelho PMZ, Estanislau JASG, da Costa Rocha MO, de Oliveira Santos M, Faria-Pinto P, Vasconcelos EG. Occurrence of a conserved domain in ATP diphosphohydrolases from pathogenic organisms associated to antigenicity in human parasitic diseases. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:1059-1067. [PMID: 21527274 DOI: 10.1016/j.dci.2011.03.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 02/15/2011] [Accepted: 03/27/2011] [Indexed: 05/30/2023]
Abstract
A polypeptide (r78-117) belonging to the potato apyrase was identified as a conserved domain shared with apyrase-like proteins from distinct pathogenic organisms, and was obtained as a 6xHis tag polypeptide (r-Domain B). By ELISA, high IgG, and IgG1 and IgG2a subtypes levels were detected in BALB/c mice pre-inoculated with r-Domain B. In Schistosoma mansoni adult worm or Leishmania (V.) braziliensis promastigote preparation, anti-r-Domain B antibodies inhibit 22-72% of the phosphohydrolytic activities and when immobilized on Protein A-Sepharose immunoprecipitate 42-91% of them. Western blots of the immunoprecipitated resin-antibody-antigen complexes identified bands of mw similar to those predicted for parasite proteins. Total IgG and subclasses of patients with leishmaniasis or schistosomiasis exhibited cross-immunoreactivity with r-Domain B. Therefore, the domain B within both S. mansoni SmATPDase 2 (r156-195) and L. (V.) braziliensis NDPase (r83-122) are potentially involved in the host immune response, and also seem to be conserved during host and parasites co-evolution.
Collapse
Affiliation(s)
- Ana Carolina Ribeiro Gomes Maia
- Departamento de Bioquímica, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, MG, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
348
|
Lemgruber L, Lupetti P, Martins-Duarte ES, De Souza W, Vommaro RC. The organization of the wall filaments and characterization of the matrix structures of Toxoplasma gondii cyst form. Cell Microbiol 2011; 13:1920-32. [PMID: 21899696 DOI: 10.1111/j.1462-5822.2011.01681.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The encystation process is a key step in Toxoplasma gondii life cycle, allowing the parasite to escape from the host immune system and the transmission among the hosts. A detailed characterization of the formation and structure of the cyst stage is essential for a better knowledge of toxoplasmosis. Here we isolated cysts from mice brains and analysed the cyst wall structure and cyst matrix organization using different electron microscopy techniques. Images obtained showed that the cyst wall presented a filamentous aspect, with circular openings on its surface. The filaments were organized in two layers: a compact one, facing the exterior of the whole cyst and a more loosen one, facing the matrix. Within the cyst wall, we observed tubules and a large number of vesicles. The cyst matrix presented vesicles of different sizes and tubules, which were organized in a network connecting the bradyzoites to each other and to the cyst wall. Large vesicles, with a granular material in their lumen of glycidic nature were observed. Similar vesicles were also found associated with the posterior pole of the bradyzoites and in proximity to the cyst wall.
Collapse
Affiliation(s)
- Leandro Lemgruber
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
349
|
Sen S, Roy K, Mukherjee S, Mukhopadhyay R, Roy S. Restoration of IFNγR subunit assembly, IFNγ signaling and parasite clearance in Leishmania donovani infected macrophages: role of membrane cholesterol. PLoS Pathog 2011; 7:e1002229. [PMID: 21931549 PMCID: PMC3169561 DOI: 10.1371/journal.ppat.1002229] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 07/14/2011] [Indexed: 01/10/2023] Open
Abstract
Despite the presence of significant levels of systemic Interferon gamma (IFNγ), the host protective cytokine, Kala-azar patients display high parasite load with downregulated IFNγ signaling in Leishmania donovani (LD) infected macrophages (LD-MØs); the cause of such aberrant phenomenon is unknown. Here we reveal for the first time the mechanistic basis of impaired IFNγ signaling in parasitized murine macrophages. Our study clearly shows that in LD-MØs IFNγ receptor (IFNγR) expression and their ligand-affinity remained unaltered. The intracellular parasites did not pose any generalized defect in LD-MØs as IL-10 mediated signal transducer and activator of transcription 3 (STAT3) phosphorylation remained unaltered with respect to normal. Previously, we showed that LD-MØs are more fluid than normal MØs due to quenching of membrane cholesterol. The decreased rigidity in LD-MØs was not due to parasite derived lipophosphoglycan (LPG) because purified LPG failed to alter fluidity in normal MØs. IFNγR subunit 1 (IFNγR1) and subunit 2 (IFNγR2) colocalize in raft upon IFNγ stimulation of normal MØs, but this was absent in LD-MØs. Oddly enough, such association of IFNγR1 and IFNγR2 could be restored upon liposomal delivery of cholesterol as evident from the fluorescence resonance energy transfer (FRET) experiment and co-immunoprecipitation studies. Furthermore, liposomal cholesterol treatment together with IFNγ allowed reassociation of signaling assembly (phospho-JAK1, JAK2 and STAT1) in LD-MØs, appropriate signaling, and subsequent parasite killing. This effect was cholesterol specific because cholesterol analogue 4-cholestene-3-one failed to restore the response. The presence of cholesterol binding motifs [(L/V)-X(1-5)-Y-X(1-5)-(R/K)] in the transmembrane domain of IFNγR1 was also noted. The interaction of peptides representing this motif of IFNγR1 was studied with cholesterol-liposome and analogue-liposome with difference of two orders of magnitude in respective affinity (K(D): 4.27×10(-9) M versus 2.69×10(-7) M). These observations reinforce the importance of cholesterol in the regulation of function of IFNγR1 proteins. This study clearly demonstrates that during its intracellular life-cycle LD perturbs IFNγR1 and IFNγR2 assembly and subsequent ligand driven signaling by quenching MØ membrane cholesterol.
Collapse
Affiliation(s)
- Subha Sen
- Division of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| | - Koushik Roy
- Division of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| | - Sandip Mukherjee
- Division of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| | - Rupkatha Mukhopadhyay
- Division of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| | - Syamal Roy
- Division of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
| |
Collapse
|
350
|
Sir2-Related Protein 1 from Leishmania amazonensis is a glycosylated NAD+-dependent deacetylase. Parasitology 2011; 138:1245-58. [PMID: 21819639 DOI: 10.1017/s0031182011001077] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Sirtuin proteins form a family of NAD+-dependent protein deacetylases that are considered potential drug targets against parasites. Here, we present the first characterization of a sirtuin orthologue from Leishmania amazonensis, an aetiological agent of American tegumentary leishmaniasis that has been the subject of many studies focused in the development of therapeutic approaches. The protein has high sequence identity with other Kinetoplastid Silent information regulator 2 Related Protein 1 (Sir2RP1) and was named LaSir2RP1. The gene exists as a single copy, encoding a monomeric protein (LaSir2RP1) of approximately 41 kDa that has NAD+-dependent deacetylase activity. LaSir2RP1 was immunodetected in total protein extracts, in cytoplasmic granules, and in the secreted material of both promastigotes and lesion-derived amastigotes. Analysis of both lectin‑affinity purified promastigote and amastigote extracts revealed the presence of a major enriched protein of approximately 66 kDa that was recognized by an anti-LaSir2RP1 serum, suggesting that a parasite sirtuin could be glycosylated in vivo.
Collapse
|