1
|
Bhattacharya S, Chakraborty S, Manna D, Thakur P, Chakravorty N, Mukherjee B. Deciphering the intricate dynamics of inflammasome regulation in visceral and post-kala-azar dermal leishmaniasis: A meta-analysis of consistencies. Acta Trop 2024; 257:107313. [PMID: 38964632 DOI: 10.1016/j.actatropica.2024.107313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Post Kala-azar dermal leishmaniasis (PKDL) arises as a significant dermal sequel following Visceral leishmaniasis (VL) caused by protozoan parasite Leishmania donovani (LD). PKDL acts as a significant constrain for VL elimination serving as a crucial reservoir for LD. PKDL patients exhibit depigmented macular and papular lesions on their skin, which results in social discrimination due to loss of natural skin color. Inflammatory reactions, prevalent in both VL and PKDL, potentially lead to tissue damage in areas harboring the parasite. Disruption of the immune-inflammasomal network not only facilitates LD persistence but also leads to the skin hypopigmentation seen in PKDL, impacting social well-being. Activation of inflammasomal markers like STAT1, NLRP1, NLRP3, AIM2, CASP11, and NLRP12 have been identified as a common host-defense mechanism across various Leishmania infections. Conversely, Leishmania modulates inflammasome activation to sustain its presence within the host. Nevertheless, in specific instances of Leishmania infection, inflammasome activation can worsen disease pathology by promoting parasite proliferation and persistence. This study encompasses recent transcriptomic analyses conducted between 2016 and 2023 on human and murine subjects afflicted with VL/PKDL, elucidating significant alterations in inflammasomal markers in both conditions. It offers a comprehensive understanding how these markers contribute in disease progression, drawing upon available literature for logical analysis. Furthermore, our analysis identifies validated miRNA network that could potentially disrupt this crucial immune-inflammasomal network, thereby offering a plausible explanation on how secreted LD-factors could enable membrane-bound LD, isolated from the host cytoplasm, to modulate cytoplasmic inflammasomal markers. Insights from this study could guide the development of host-directed therapeutics to impede transmission and address hypopigmentation, thereby mitigating the social stigma associated with PKDL.
Collapse
Affiliation(s)
| | | | - Debolina Manna
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India
| | - Pradipti Thakur
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India
| | - Nishant Chakravorty
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India
| | - Budhaditya Mukherjee
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India.
| |
Collapse
|
2
|
Kuhlmann FM, Key PN, Hickerson SM, Turk J, Hsu FF, Beverley SM. Inositol phosphorylceramide synthase null Leishmania are viable and virulent in animal infections where salvage of host sphingomyelin predominates. J Biol Chem 2022; 298:102522. [PMID: 36162499 PMCID: PMC9637897 DOI: 10.1016/j.jbc.2022.102522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 12/03/2022] Open
Abstract
Many pathogens synthesize inositol phosphorylceramide (IPC) as the major sphingolipid (SL), differing from the mammalian host where sphingomyelin (SM) or more complex SLs predominate. The divergence between IPC synthase and mammalian SL synthases has prompted interest as a potential drug target. However, in the trypanosomatid protozoan Leishmania, cultured insect stage promastigotes lack de novo SL synthesis (Δspt2-) and SLs survive and remain virulent, as infective amastigotes salvage host SLs and continue to produce IPC. To further understand the role of IPC, we generated null IPCS mutants in Leishmania major (Δipcs-). Unexpectedly and unlike fungi where IPCS is essential, Δipcs- was remarkably normal in culture and highly virulent in mouse infections. Both IPCS activity and IPC were absent in Δipcs- promastigotes and amastigotes, arguing against an alternative route of IPC synthesis. Notably, salvaged mammalian SM was highly abundant in purified amastigotes from both WT and Δipcs-, and salvaged SLs could be further metabolized into IPC. SM was about 7-fold more abundant than IPC in WT amastigotes, establishing that SM is the dominant amastigote SL, thereby rendering IPC partially redundant. These data suggest that SM salvage likely plays key roles in the survival and virulence of both WT and Δipcs- parasites in the infected host, confirmation of which will require the development of methods or mutants deficient in host SL/SM uptake in the future. Our findings call into question the suitability of IPCS as a target for chemotherapy, instead suggesting that approaches targeting SM/SL uptake or catabolism may warrant further emphasis.
Collapse
Affiliation(s)
- F. Matthew Kuhlmann
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA,Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Phillip N. Key
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Suzanne M. Hickerson
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - John Turk
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Fong-Fu Hsu
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA,For correspondence: Stephen M. Beverley
| |
Collapse
|
3
|
Gurjar D, Kumar Patra S, Bodhale N, Lenka N, Saha B. Leishmania intercepts IFN-γR signaling at multiple levels in macrophages. Cytokine 2022; 157:155956. [PMID: 35785668 DOI: 10.1016/j.cyto.2022.155956] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022]
Abstract
IFN-γ, a type 2 interferon and a cytokine, is critical for both innate and adaptive immunity. IFN-γ binds to the IFN-γRs on the cell membrane of macrophages, signals through JAK1-STAT-1 pathway and induces IFN-γ-stimulated genes (ISGs). As Leishmania amastigotes reside and replicate within macrophages, IFN-γ mediated macrophage activation eventuate in Leishmania elimination. As befits the principle of parasitism, the impaired IFN-γ responsiveness in macrophages ensures Leishmania survival. IFN-γ responsiveness is a function of integrated molecular events at multiple levels in the cells that express IFN-γ receptors. In Leishmania-infected macrophages, reduced IFN-γRα expression, impaired IFN-γRα and IFN-γRβ hetero-dimerization due to altered membrane lipid composition, reduced JAK-1 and STAT-1 phosphorylation but increased STAT-1 degradation and impaired ISGs induction collectively determine the IFN-γ responsiveness and the efficacy of IFN-γ induced antileishmanial function of macrophages. Therefore, parasite load is not only decided by the levels of IFN-γ produced but also by the IFN-γ responsiveness. Indeed, in Leishmania-infected patients, IFN-γ is produced but IFN-γ signalling is downregulated. However, the molecular mechanisms of IFN-γ responsiveness remain unclear. Therefore, we review the current understanding of IFN-γ responsiveness of Leishmania-infected macrophages.
Collapse
Affiliation(s)
- Dhiraj Gurjar
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | | | - Neelam Bodhale
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Nibedita Lenka
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| |
Collapse
|
4
|
Rostami MN, Khamesipour A. Potential biomarkers of immune protection in human leishmaniasis. Med Microbiol Immunol 2021; 210:81-100. [PMID: 33934238 PMCID: PMC8088758 DOI: 10.1007/s00430-021-00703-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/22/2021] [Indexed: 12/30/2022]
Abstract
Leishmaniasis is a vector-borne neglected tropical disease endemic in over 100 countries around the world. Available control measures are not always successful, therapeutic options are limited, and there is no vaccine available against human leishmaniasis, although several candidate antigens have been evaluated over the last decades. Plenty of studies have aimed to evaluate the immune response development and a diverse range of host immune factors have been described to be associated with protection or disease progression in leishmaniasis; however, to date, no comprehensive biomarker(s) have been identified as surrogate marker of protection or exacerbation, and lack of enough information remains a barrier for vaccine development. Most of the current understanding of the role of different markers of immune response in leishmaniasis has been collected from experimental animal models. Although the data generated from the animal models are crucial, it might not always be extrapolated to humans. Here, we briefly review the events during Leishmania invasion of host cells and the immune responses induced against Leishmania in animal models and humans and their potential role as a biomarker of protection against human leishmaniasis.
Collapse
Affiliation(s)
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, 14155-6383, Tehran, Iran.
| |
Collapse
|
5
|
Krayem I, Lipoldová M. Role of host genetics and cytokines in Leishmania infection. Cytokine 2020; 147:155244. [PMID: 33059974 DOI: 10.1016/j.cyto.2020.155244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/20/2020] [Accepted: 08/08/2020] [Indexed: 12/29/2022]
Abstract
Cytokines and chemokines are important regulators of innate and specific responses in leishmaniasis, a disease that currently affects 12 million people. We overviewed the current information about influences of genetically engineered mouse models of cytokine and chemokine on leishmaniasis. We found that genetic background of the host, parasite species and sub-strain, as well as experimental design often modify effects of genetically engineered cytokine genes. Next we analyzed genes and QTLs (quantitative trait loci) that control response to Leishmania species in mouse in order to establish relationship between genetic control of cytokine expression and organ pathology. These studies revealed a network-like complexity of the combined effects of the multiple functionally diverse QTLs and their individual specificity. Genetic control of organ pathology and systemic immune response overlap only partially. Some QTLs control both organ pathology and systemic immune response, but the effects of genes and loci with the strongest impact on disease are cytokine-independent, whereas several loci modify cytokines levels in serum without influencing organ pathology. Understanding this genetic control might be important in development of vaccines designed to stimulate certain cytokine spectrum.
Collapse
Affiliation(s)
- Imtissal Krayem
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic; Department of Natural Sciences, Faculty of Biomedical Engineering, Czech Technical University in Prague, Sítná 3105, 272 01 Kladno, Czech Republic.
| |
Collapse
|
6
|
The macrophage microtubule network acts as a key cellular controller of the intracellular fate of Leishmania infantum. PLoS Negl Trop Dis 2020; 14:e0008396. [PMID: 32722702 PMCID: PMC7386624 DOI: 10.1371/journal.pntd.0008396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/16/2020] [Indexed: 11/19/2022] Open
Abstract
The parasitophorous vacuoles (PVs) that insulate Leishmania spp. in host macrophages are vacuolar compartments wherein promastigote forms differentiate into amastigote that are the replicative form of the parasite and are also more resistant to host responses. We revisited the biogenesis of tight-fitting PVs that insulate L. infantum in promastigote-infected macrophage-like RAW 264.7 cells by time-dependent confocal laser multidimensional imaging analysis. Pharmacological disassembly of the cellular microtubule network and silencing of the dynein gene led to an impaired interaction of L. infantum-containing phagosomes with late endosomes and lysosomes, resulting in the tight-fitting parasite-containing phagosomes never transforming into mature PVs. Analysis of the shape of the L. infantum parasite within PVs, showed that factors that impair promastigote-amastigote differentiation can also result in PVs whose maturation is arrested. These findings highlight the importance of the MT-dependent interaction of L. infantum-containing phagosomes with the host macrophage endolysosomal pathway to secure the intracellular fate of the parasite. Kinetoplastid parasites of the genus Leishmania are responsible for a diverse spectrum of mammalian infectious diseases, the leishmaniases, including cutaneous, mucocutaneous, and mucosal pathologies. Infectious metacyclic promastigotes of infected female Phlebotomus sandflies are injected into the host at the site of the bite during the sandfly blood meal, after which they are internalized by host professional phagocytic neutrophils and macrophages. Leishmania infantum is an etiological agent of potentially fatal visceral pathology. This study molecularly dissects the maturation of L. infantum-containing phagosomes/parasitophorous vacuoles (PVs) in host macrophages. We reveal the requirement of vacuolar movement along macrophage microtubule tracks for the phagosome trafficking toward the endolysosomal pathway necessary for the development of the mature tight-fitting PV crucial for L. infantum survival and proliferation.
Collapse
|
7
|
Zutshi S, Kumar S, Chauhan P, Bansode Y, Nair A, Roy S, Sarkar A, Saha B. Anti-Leishmanial Vaccines: Assumptions, Approaches, and Annulments. Vaccines (Basel) 2019; 7:vaccines7040156. [PMID: 31635276 PMCID: PMC6963565 DOI: 10.3390/vaccines7040156] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/24/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022] Open
Abstract
Leishmaniasis is a neglected protozoan parasitic disease that occurs in 88 countries but a vaccine is unavailable. Vaccination with live, killed, attenuated (physically or genetically) Leishmania have met with limited success, while peptide-, protein-, or DNA-based vaccines showed promise only in animal models. Here, we critically assess several technical issues in vaccination and expectation of a host-protective immune response. Several studies showed that antigen presentation during priming and triggering of the same cells in infected condition are not comparable. Altered proteolytic processing, antigen presentation, protease-susceptible sites, and intracellular expression of pathogenic proteins during Leishmania infection may vary dominant epitope selection, MHC-II/peptide affinity, and may deter the reactivation of desired antigen-specific T cells generated during priming. The robustness of the memory T cells and their functions remains a concern. Presentation of the antigens by Leishmania-infected macrophages to antigen-specific memory T cells may lead to change in the T cells' functional phenotype or anergy or apoptosis. Although cells may be activated, the peptides generated during infection may be different and cross-reactive to the priming peptides. Such altered peptide ligands may lead to suppression of otherwise active antigen-specific T cells. We critically assess these different immunological issues that led to the non-availability of a vaccine for human use.
Collapse
Affiliation(s)
| | - Sunil Kumar
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Prashant Chauhan
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Yashwant Bansode
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Arathi Nair
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Somenath Roy
- Department of Human Physiology with Community Health, Vidyasagar University, Midnapore 721102, India.
| | - Arup Sarkar
- Department of Biotechnology, Trident Academy of Creative Technology, Bhubaneswar 751024, India.
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
- Department of Biotechnology, Trident Academy of Creative Technology, Bhubaneswar 751024, India.
| |
Collapse
|
8
|
Dos-Santos JS, Firmino-Cruz L, Ramos TD, da Fonseca-Martins AM, Oliveira-Maciel D, De-Medeiros JVR, Chaves SP, Gomes DCO, de Matos Guedes HL. Characterization of Sv129 Mice as a Susceptible Model to Leishmania amazonensis. Front Med (Lausanne) 2019; 6:100. [PMID: 31192210 PMCID: PMC6548835 DOI: 10.3389/fmed.2019.00100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/24/2019] [Indexed: 11/30/2022] Open
Abstract
Leishmaniasis is a complex of neglected diseases caused by parasites of the genus Leishmania, such as Leishmania (Leishmania) amazonensis, the ethiologic agent of diffuse cutaneous leishmaniasis in Brazil. In this work, we investigated a new experimental model of infection for L. amazonensis: the Sv129 mouse. First, we subcutaneously infected Sv129 mice with 2 × 105 or 2 × 106L. amazonensis parasites of the Josefa strain. A progressive lesion developed for both inoculation doses, showing that Sv129 mice are susceptible, independent of parasite dose. We next investigated the mechanisms associated with the pathogenesis of infection. We did not observe an increase of frequency of interferon-gamma (IFN- γ)-producing CD4+ and CD8+ T cells, a phenotype similar to that seen in BALB/c mice. There was an increased of frequency and number of IL-17-producing γδ (gamma-delta) T cells in infected Sv129 mice compared to naïve SV129 and an increased frequency of this population compared to infected BALB/c mice. In addition, Sv129 mice presented high levels of both IgG1 and IgG2a, suggesting a mixed Th1 and Th2 response with a skew toward IgG1 production based on IgG1/IgG2a ratio. Susceptibility of the Sv129 mice was further confirmed with the use of another strain of L. amazonensis, LTB0016. In this work, we characterized the Sv129 mice as a new model of susceptibility to Leishmania amazonensis infection, during infection there was controlled IFN-γ production by CD4+ or CD8+ T cells and induced IL-17 production by γδ T cells.
Collapse
Affiliation(s)
- Júlio Souza Dos-Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luan Firmino-Cruz
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tadeu Diniz Ramos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Diogo Oliveira-Maciel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | - Herbert Leonel de Matos Guedes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Núcleo Multidisciplinar de Pesquisa UFRJ - Xerém em Biologia, UFRJ Campus Duque de Caxias Professor Geraldo Cidade - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Kowalski E, Geng S, Rathes A, Lu R, Li L. Toll-interacting protein differentially modulates HIF1α and STAT5-mediated genes in fibroblasts. J Biol Chem 2018; 293:12239-12247. [PMID: 29921584 DOI: 10.1074/jbc.ra118.003382] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/11/2018] [Indexed: 11/06/2022] Open
Abstract
Toll-interacting protein (Tollip) deficiency has been implicated in complex inflammatory and infectious diseases whose mechanisms are poorly understood. Comparing the gene expression profiles of WT and Tollip-deficient murine embryonic fibroblasts, we observed here that Tollip deficiency selectively reduces the expression of the inflammatory cytokines interleukin 6 (IL-6), IL-12, and tumor necrosis factor α (TNFα) but potentiates the expression of fatty acid-binding protein 4 (FABP4) in these cells. We also observed that expression of hypoxia-inducible factor 1-α (HIF1α) is reduced, whereas that of signal transducer and activator of transcription 5 (STAT5) is elevated, in Tollip-deficient cells, correlating with the decreased expression of inflammatory cytokines and increased expression of FABP4 in these cells. We further found that the coupling of ubiquitin to ER degradation (CUE) domain of Tollip is required for stimulating HIF1α activity, because Tollip CUE-domain mutant cells exhibited reduced levels of HIF1α and selected cytokines. Tollip is known to mediate autophagy and lysosome fusion, and herein we observed that Tollip's autophagy function is required for modulating STAT5 and FABP4 expression. Bafilomycin A, an inhibitor of lysosome fusion, enhanced STAT5 and FABP4 expression in WT fibroblasts, whereas torin 2, an activator of autophagy, reduced STAT5 and FABP4 expression in Tollip-deficient fibroblasts. Taken together, our study reveals that Tollip differentially modulates HIF1α and STAT5 expression in fibroblasts, potentially explaining the complex and context-dependent contribution of Tollip to disease development.
Collapse
Affiliation(s)
- Elizabeth Kowalski
- Department of Biological Sciences and Biochemistry, Virginia Tech, Blacksburg, Virginia 24061
| | - Shuo Geng
- Department of Biological Sciences and Biochemistry, Virginia Tech, Blacksburg, Virginia 24061
| | - Allison Rathes
- Department of Biological Sciences and Biochemistry, Virginia Tech, Blacksburg, Virginia 24061
| | - Ran Lu
- Department of Biological Sciences and Biochemistry, Virginia Tech, Blacksburg, Virginia 24061
| | - Liwu Li
- Department of Biological Sciences and Biochemistry, Virginia Tech, Blacksburg, Virginia 24061.
| |
Collapse
|
10
|
Azami M, Ranjkesh Adermanabadi V, Khanahmad H, Mohaghegh MA, Zaherinejad E, Aghaei M, Jalali A, Hejazi SH. Immunology and Genetic of Leishmania infantum: The Role of Endonuclease G in the Apoptosis. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2018; 23:36. [PMID: 29887904 PMCID: PMC5961285 DOI: 10.4103/jrms.jrms_705_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 01/13/2018] [Accepted: 01/31/2018] [Indexed: 12/13/2022]
Abstract
Leishmania infantum is the causative agent of infantile visceral leishmaniasis (VL) in the Mediterranean region. Despite developing protective responses, the disease progresses due to many of factors. These include the action of suppressive cytokines, exhaustion of specific T cells, loss of lymphoid tissue, and defective humoral response. Genetic changes that occur inside the genome of alienated or parasite cells, along with immune responses, play an important role in controlling or progressing the disease. Proapoptotic proteins such as Smac/DIABLO, EndoG, AIF (apoptosis-inducing factor), and cytochrome C are effective in apoptosis. EndoG is a mitochondrion-specific nuclease that translocates to the nucleus during apoptosis. Once released from mitochondria, endoG cleaves chromatin DNA into nucleosomal fragments independently of caspases. Therefore, endoG represents a caspase-independent apoptotic pathway initiated from the mitochondria. A comprehensive understanding of the immune and genetic events that occur during VL is very important for designing immunotherapy strategies and developing effective vaccines for disease prevention. In this review which explained the immunological responses and also the important factors that can contribute to parasite apoptosis and are used in subsequent studies as a target for the preparation of drugs or recombinant vaccines against parasites are briefly reviewed.
Collapse
Affiliation(s)
- Mehdi Azami
- Skin Diseases and Leishmaniasis Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Hossein Khanahmad
- Department of Molecular Biology and Genetics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Ali Mohaghegh
- Department of Laboratory Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Health Sciences Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ebtesam Zaherinejad
- School of Pharmacy, Department of Health Sciences, Curtin University of Technology, Bentley Campus, Australia
| | - Maryam Aghaei
- Skin Diseases and Leishmaniasis Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Akram Jalali
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Hossein Hejazi
- Skin Diseases and Leishmaniasis Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
11
|
Mori H, Chen JQ, Cardiff RD, Pénzváltó Z, Hubbard NE, Schuetter L, Hovey RC, Trott JF, Borowsky AD. Pathobiology of the 129:Stat1 -/- mouse model of human age-related ER-positive breast cancer with an immune infiltrate-excluded phenotype. Breast Cancer Res 2017; 19:102. [PMID: 28865492 PMCID: PMC5581425 DOI: 10.1186/s13058-017-0892-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 08/07/2017] [Indexed: 01/05/2023] Open
Abstract
Background Stat1 gene-targeted knockout mice (129S6/SvEvTac-Stat1tm1Rds) develop estrogen receptor-positive (ER+), luminal-type mammary carcinomas at an advanced age. There is evidence for both host environment as well as tumor cell-intrinsic mechanisms to initiate tumorigenesis in this model. In this report, we summarize details of the systemic and mammary pathology at preneoplastic and tumor-bearing time points. In addition, we investigate tumor progression in the 129:Stat1−/− host compared with wild-type 129/SvEv, and we describe the immune cell reaction to the tumors. Methods Mice housed and treated according to National Institutes of Health guidelines and Institutional Animal Care and Use Committee-approved methods were evaluated by histopathology, and their tissues were subjected to immunohistochemistry with computer-assisted quantitative image analysis. Tumor cell culture and conditioned media from cell culture were used to perform macrophage (RAW264.7) cell migration assays, including the 129:Stat1−/−-derived SSM2 cells as well as control Met1 and NDL tumor cells and EpH4 normal cells. Results Tumorigenesis in 129:Stat1−/− originates from a population of FoxA1+ large oval pale cells that initially appear and accumulate along the mammary ducts in segments or regions of the gland prior to giving rise to mammary intraepithelial neoplasias. Progression to invasive carcinoma is accompanied by a marked local stromal and immune cell response composed predominantly of T cells and macrophages. In conditioned media experiments, cells derived from 129:Stat1−/− tumors secrete both chemoattractant and chemoinhibitory factors, with greater attraction in the extracellular vesicular fraction and inhibition in the soluble fraction. The result appears to be recruitment of the immune reaction to the periphery of the tumor, with exclusion of immune cell infiltration into the tumor. Conclusions 129:Stat1−/− is a unique model for studying the critical origins and risk reduction strategies in age-related ER+ breast cancer. In addition, it can be used in preclinical trials of hormonal and targeted therapies as well as immunotherapies. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0892-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hidetoshi Mori
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA
| | - Jane Q Chen
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA
| | - Robert D Cardiff
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA.,Department of Pathology and Laboratory Medicine, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Zsófia Pénzváltó
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA
| | - Neil E Hubbard
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA
| | - Louis Schuetter
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA
| | - Russell C Hovey
- Department of Animal Science, University of California at Davis, Davis, CA, USA
| | - Josephine F Trott
- Department of Animal Science, University of California at Davis, Davis, CA, USA
| | - Alexander D Borowsky
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA. .,Department of Pathology and Laboratory Medicine, School of Medicine, University of California at Davis, Sacramento, CA, USA.
| |
Collapse
|
12
|
De Muylder G, Vanhollebeke B, Caljon G, Wolfe AR, McKerrow J, Dujardin JC. Naloxonazine, an Amastigote-Specific Compound, Affects Leishmania Parasites through Modulation of Host-Encoded Functions. PLoS Negl Trop Dis 2016; 10:e0005234. [PMID: 28036391 PMCID: PMC5201425 DOI: 10.1371/journal.pntd.0005234] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 12/06/2016] [Indexed: 11/24/2022] Open
Abstract
Host-directed therapies (HDTs) constitute promising alternatives to traditional therapy that directly targets the pathogen but is often hampered by pathogen resistance. HDT could represent a new treatment strategy for leishmaniasis, a neglected tropical disease caused by the obligate intracellular parasite Leishmania. This protozoan develops exclusively within phagocytic cells, where infection relies on a complex molecular interplay potentially exploitable for drug targets. We previously identified naloxonazine, a compound specifically active against intracellular but not axenic Leishmania donovani. We evaluated here whether this compound could present a host cell-dependent mechanism of action. Microarray profiling of THP-1 macrophages treated with naloxonazine showed upregulation of vATPases, which was further linked to an increased volume of intracellular acidic vacuoles. Treatment of Leishmania-infected macrophages with the vATPase inhibitor concanamycin A abolished naloxonazine effects, functionally demonstrating that naloxonazine affects Leishmania amastigotes indirectly, through host cell vacuolar remodeling. These results validate amastigote-specific screening approaches as a powerful way to identify alternative host-encoded targets. Although the therapeutic value of naloxonazine itself is unproven, our results further demonstrate the importance of intracellular acidic compartments for host defense against Leishmania, highlighting the possibility of targeting this host cell compartment for anti-leishmanial therapy.
Collapse
Affiliation(s)
- Géraldine De Muylder
- Institute of Tropical Medicine, Department of Biomedical Sciences, Antwerp, Belgium
- University of California, San Francisco, San Francisco, CA, United States of America
| | - Benoit Vanhollebeke
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, Gosselies, Belgium
| | - Guy Caljon
- Institute of Tropical Medicine, Department of Biomedical Sciences, Antwerp, Belgium
| | - Alan R. Wolfe
- University of California, San Francisco, San Francisco, CA, United States of America
| | - James McKerrow
- University of California, San Francisco, San Francisco, CA, United States of America
| | - Jean-Claude Dujardin
- Institute of Tropical Medicine, Department of Biomedical Sciences, Antwerp, Belgium
- Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
13
|
Attenuated Leishmania induce pro-inflammatory mediators and influence leishmanicidal activity by p38 MAPK dependent phagosome maturation in Leishmania donovani co-infected macrophages. Sci Rep 2016; 6:22335. [PMID: 26928472 PMCID: PMC4772118 DOI: 10.1038/srep22335] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/08/2016] [Indexed: 12/21/2022] Open
Abstract
Promastigote form of Leishmania, an intracellular pathogen, delays phagosome maturation and resides inside macrophages. But till date limited study has been done to manipulate the phagosomal machinery of macrophages to restrict Leishmania growth. Attenuated Leishmania strain exposed RAW 264.7 cells showed a respiratory burst and enhanced production of pro-inflammatory mediators. The augmentation of pro-inflammatory activity is mostly attributed to p38 MAPK and p44/42 MAPK. In our study, these activated macrophages are found to induce phagosome maturation when infected with pathogenic Leishmania donovani. Increased co-localization of carboxyfluorescein succinimidyl ester labeled pathogenic L. donovani with Lysosome was found. Moreover, increased co-localization was observed between pathogenic L. donovani and late phagosomal markers viz. Rab7, Lysosomal Associated Membrane Protein 1, Cathepsin D, Rab9, and V-ATPase which indicate phagosome maturation. It was also observed that inhibition of V-type ATPase caused significant hindrance in attenuated Leishmania induced phagosome maturation. Finally, it was confirmed that p38 MAPK is the key player in acidification and maturation of phagosome in attenuated Leishmania strain pre-exposed macrophages. To our knowledge, this study for the first time reported an approach to induce phagosome maturation in L. donovani infected macrophages which could potentiate short-term prophylactic response in future.
Collapse
|
14
|
Chen JQ, Mori H, Cardiff RD, Trott JF, Hovey RC, Hubbard NE, Engelberg JA, Tepper CG, Willis BJ, Khan IH, Ravindran RK, Chan SR, Schreiber RD, Borowsky AD. Abnormal Mammary Development in 129:STAT1-Null Mice is Stroma-Dependent. PLoS One 2015; 10:e0129895. [PMID: 26075897 PMCID: PMC4468083 DOI: 10.1371/journal.pone.0129895] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 05/14/2015] [Indexed: 11/18/2022] Open
Abstract
Female 129:Stat1-null mice (129S6/SvEvTac-Stat1tm1Rds homozygous) uniquely develop estrogen-receptor (ER)-positive mammary tumors. Herein we report that the mammary glands (MG) of these mice have altered growth and development with abnormal terminal end buds alongside defective branching morphogenesis and ductal elongation. We also find that the 129:Stat1-null mammary fat pad (MFP) fails to sustain the growth of 129S6/SvEv wild-type and Stat1-null epithelium. These abnormalities are partially reversed by elevated serum progesterone and prolactin whereas transplantation of wild-type bone marrow into 129:Stat1-null mice does not reverse the MG developmental defects. Medium conditioned by 129:Stat1-null epithelium-cleared MFP does not stimulate epithelial proliferation, whereas it is stimulated by medium conditioned by epithelium-cleared MFP from either wild-type or 129:Stat1-null females having elevated progesterone and prolactin. Microarrays and multiplexed cytokine assays reveal that the MG of 129:Stat1-null mice has lower levels of growth factors that have been implicated in normal MG growth and development. Transplanted 129:Stat1-null tumors and their isolated cells also grow slower in 129:Stat1-null MG compared to wild-type recipient MG. These studies demonstrate that growth of normal and neoplastic 129:Stat1-null epithelium is dependent on the hormonal milieu and on factors from the mammary stroma such as cytokines. While the individual or combined effects of these factors remains to be resolved, our data supports the role of STAT1 in maintaining a tumor-suppressive MG microenvironment.
Collapse
Affiliation(s)
- Jane Q. Chen
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Hidetoshi Mori
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Robert D. Cardiff
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Josephine F. Trott
- Department of Animal Science, University of California, Davis, California, United States of America
| | - Russell C. Hovey
- Department of Animal Science, University of California, Davis, California, United States of America
| | - Neil E. Hubbard
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Jesse A. Engelberg
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Clifford G. Tepper
- Division of Basic Sciences, Cancer Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, United States of America
| | - Brandon J. Willis
- Mouse Biology Program, University of California, Davis, California, United States of America
| | - Imran H. Khan
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Resmi K. Ravindran
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Szeman R. Chan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Robert D. Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Alexander D. Borowsky
- Center for Comparative Medicine, University of California, Davis, California, United States of America
- Department of Pathology and Laboratory Medicine, University of California, Davis, School of Medicine, Sacramento, California, United States of America
- * E-mail:
| |
Collapse
|
15
|
Stahl P, Schwarz RT, Debierre-Grockiego F, Meyer T. Trypanosoma cruzi parasites fight for control of the JAK-STAT pathway by disarming their host. JAKSTAT 2015; 3:e1012964. [PMID: 26413423 DOI: 10.1080/21623996.2015.1012964] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 12/13/2022] Open
Abstract
The zoonotic Chagas' disease is caused by infections with the hemoflagellate Trypanosoma cruzi (T. cruzi) which is endemic in Latin America. Despite recent advances in our understanding of the pathogenesis of the disease, the underlying molecular processes involved in host-parasite interactions are only poorly understood. In particular, the mechanisms for parasite persistence in host cells remain largely unknown. Cytokine-driven transcription factors from the family of STAT (signal transducer and activator of transcription) proteins appear to play a central role in the fight against T. cruzi infection. However, amastigotes proliferating in the cytoplasm of infected host cells develop effective strategies to circumvent the attack executed by STAT proteins. This review highlights the interactions between T. cruzi parasites and human host cells in terms of cytokine signaling and, in particular, discusses the impact of STATs on the balance between parasite invasion and clearance.
Collapse
Affiliation(s)
- Philipp Stahl
- Institute of Virology; Parasitology Unit; University of Marburg ; Marburg, Germany
| | - Ralph T Schwarz
- Institute of Virology; Parasitology Unit; University of Marburg ; Marburg, Germany ; Laboratory for Structural and Functional Glycobiology; University of Lille 1 for Sciences and Technologies ; Lille, France
| | - Françoise Debierre-Grockiego
- Mixed Research Unit 1282; François Rabelais University of Tours-INRA; Infectious Diseases and Public Health ; Tours, France
| | - Thomas Meyer
- Department of Psychosomatic Medicine and Psychotherapy; University of Göttingen ; Göttingen, Germany ; German Center for Cardiovascular Research ; Göttingen, Germany
| |
Collapse
|
16
|
Forestier CL. Imaging host-Leishmania interactions: significance in visceral leishmaniasis. Parasite Immunol 2014; 35:256-66. [PMID: 23772814 DOI: 10.1111/pim.12044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 06/04/2013] [Indexed: 01/12/2023]
Abstract
Leishmaniasis is a neglected disease that is associated with a spectrum of clinical manifestations ranging from self-healing cutaneous lesions to fatal visceral infections, which primarily depends on the parasite species. In visceral leishmaniasis (VL), as opposed to cutaneous leishmaniasis (CL), parasites that infect host cells at the sand fly bite site have the striking ability to disseminate to visceral organs where they proliferate and persist for long periods of time. Imaging the dynamics of the host-Leishmania interaction in VL provides a powerful approach to understanding the mechanisms underlying host cell invasion, Leishmania dissemination and persistence within visceral organs and, to dissecting the immune responses to infection. Therefore, by allowing the visualization of the critical steps involved in the pathogenesis of VL, state-of-the-art microscopy technologies have the great potential to aid in the identification of better intervention strategies for this devastating disease. In this review, we emphasize the current knowledge and the potential significance of imaging technologies in understanding the infection process of visceralizing Leishmania species. Then, we discuss how application of innovative microscopy technologies to the study of VL will provide rich opportunities for investigating host-parasite interactions at a previously unexplored level and elucidating visceral disease-promoting mechanisms.
Collapse
Affiliation(s)
- C-L Forestier
- INSERM U1095, URMITE-UMR CNRS 7278, University of Aix-Marseille, Marseille, France.
| |
Collapse
|
17
|
Oghumu S, Gupta G, Snider HM, Varikuti S, Terrazas CA, Papenfuss TL, Kaplan MH, Satoskar AR. STAT4 is critical for immunity but not for antileishmanial activity of antimonials in experimental visceral leishmaniasis. Eur J Immunol 2013; 44:450-9. [PMID: 24242758 DOI: 10.1002/eji.201343477] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Revised: 09/07/2013] [Accepted: 10/07/2013] [Indexed: 11/11/2022]
Abstract
We and others have previously shown that IL-12 is indispensable for immunity and is required for the optimal antiparasitic activity of antimonials in experimental visceral leishmaniasis caused by Leishmania donovani. Here we investigated the role of STAT4 in immunity against L. donovani using STAT4 knockout mice and also determined the effect of STAT4 deficiency in response to antimonial therapy. Upon infection with L. donovani, stat4⁻/⁻ BALB/c and C57BL/6 mice showed enhanced susceptibility to Leishmania during late time points of infection which was associated with a marked reduction in Th1 responses and hepatic immunopathology. Interestingly, these defects in Th1 responses in stat4⁻/⁻ did not impair the antimonial chemotherapy as both stat4⁻/⁻ and WT mice showed comparable levels of parasite clearance from the liver and spleen. These findings highlight the role of STAT4 in immunity to L. donovani infection and also provide evidence that STAT4 is dispensable for antimonial-based chemotherapy.
Collapse
Affiliation(s)
- Steve Oghumu
- Department of Pathology, Ohio State University Medical Center, Columbus, OH, USA; Department of Oral Biology, Ohio State University College of Dentistry, Columbus, OH, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Polando R, Dixit UG, Carter CR, Jones B, Whitcomb JP, Ballhorn W, Harintho M, Jerde CL, Wilson ME, McDowell MA. The roles of complement receptor 3 and Fcγ receptors during Leishmania phagosome maturation. J Leukoc Biol 2013; 93:921-32. [PMID: 23543768 DOI: 10.1189/jlb.0212086] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Leishmania are intracellular parasites adapted to surviving in macrophages, whose primary function is elimination of invading pathogens. Leishmania entry into host cells is receptor-mediated. These parasites are able to engage multiple host cell-surface receptors, including MR, TLRs, CR3, and FcγRs. Here, we investigated the role of CR3 and FcγR engagement on the maturation of Leishmania-containing phagosomes using CD11b-/- and FcγR-/- macrophages, and assessing EEA1 and lysosome-associated proteins is necessary for the phagosome maturation delay, characteristic of Leishmania infection. Leishmania-containing phagosomes do not fuse with lyosomes until 5 h postinfection in WT mice. Phagolysosome fusion occurs by 1 h in CD11b and FcγR common chain KO macrophages, although receptor deficiency does not influence Leishmania entry or viability. We also investigated the influence of serum components and their effects on phagosome maturation progression. Opsonization with normal mouse serum, complement-deficient serum, or serum from Leishmania-infected mice all influenced phagosome maturation progression. Our results indicate that opsonophagocytosis influences phagosomal trafficking of Leishmania without altering the intracellular fate.
Collapse
Affiliation(s)
- Rachel Polando
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Bowick GC, Airo AM, Bente DA. Expression of interferon-induced antiviral genes is delayed in a STAT1 knockout mouse model of Crimean-Congo hemorrhagic fever. Virol J 2012; 9:122. [PMID: 22713837 PMCID: PMC3489622 DOI: 10.1186/1743-422x-9-122] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 06/19/2012] [Indexed: 01/14/2023] Open
Abstract
Background Crimean Congo hemorrhagic fever (CCHF) is a tick-borne hemorrhagic zoonosis associated with high mortality. Pathogenesis studies and the development of vaccines and antivirals against CCHF have been severely hampered by the lack of suitable animal model. We recently developed and characterized a mature mouse model for CCHF using mice carrying STAT1 knockout (KO). Findings Given the importance of interferons in controlling viral infections, we investigated the expression of interferon pathway-associated genes in KO and wild-type (WT) mice challenged with CCHF virus. We expected that the absence of the STAT1 protein would result in minimal expression of IFN-related genes. Surprisingly, the KO mice showed high levels of IFN-stimulated gene expression, beginning on day 2 post-infection, while in WT mice challenged with virus the same genes were expressed at similar levels on day 1. Conclusions We conclude that CCHF virus induces similar type I IFN responses in STAT1 KO and WT mice, but the delayed response in the KO mice permits rapid viral dissemination and fatal illness.
Collapse
Affiliation(s)
- Gavin C Bowick
- Department of Microbiology & Immunology, Center for Biodefense and Emerging Infectious Diseases, Sealy Center for Vaccine Development, Institute for Human Infections & Immunity, Galveston, TX, USA
| | | | | |
Collapse
|
20
|
Liu D, Uzonna JE. The early interaction of Leishmania with macrophages and dendritic cells and its influence on the host immune response. Front Cell Infect Microbiol 2012; 2:83. [PMID: 22919674 PMCID: PMC3417671 DOI: 10.3389/fcimb.2012.00083] [Citation(s) in RCA: 233] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 05/27/2012] [Indexed: 01/26/2023] Open
Abstract
The complicated interactions between Leishmania and the host antigen-presenting cells (APCs) have fundamental effects on the final outcome of the disease. Two major APCs, macrophages and dendritic cells (DCs), play critical roles in mediating resistance and susceptibility during Leishmania infection. Macrophages are the primary resident cell for Leishmania: they phagocytose and permit parasite proliferation. However, these cells are also the major effector cells to eliminate infection. The effective clearance of parasites by macrophages depends on activation of appropriate immune response, which is usually initiated by DCs. Here, we review the early interaction of APCs with Leishmania parasites and how these interactions profoundly impact on the ensuing adaptive immune response. We also discuss how the current knowledge will allow further refinement of our understanding of the interplay between Leishmania and its hosts that leads to resistance or susceptibility.
Collapse
Affiliation(s)
- Dong Liu
- Department of Immunology, University of Manitoba, Winnipeg MB, Canada
| | | |
Collapse
|
21
|
Vijayan A, Gómez CE, Espinosa DA, Goodman AG, Sanchez-Sampedro L, Sorzano COS, Zavala F, Esteban M. Adjuvant-like effect of vaccinia virus 14K protein: a case study with malaria vaccine based on the circumsporozoite protein. THE JOURNAL OF IMMUNOLOGY 2012; 188:6407-17. [PMID: 22615208 DOI: 10.4049/jimmunol.1102492] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Development of subunit vaccines for malaria that elicit a strong, long-term memory response is an intensive area of research, with the focus on improving the immunogenicity of a circumsporozoite (CS) protein-based vaccine. In this study, we found that a chimeric protein, formed by fusing vaccinia virus protein 14K (A27) to the CS of Plasmodium yoelii, induces strong effector memory CD8(+) T cell responses in addition to high-affinity Abs when used as a priming agent in the absence of any adjuvant, followed by an attenuated vaccinia virus boost expressing CS in murine models. Moreover, priming with the chimeric protein improved the magnitude and polyfunctionality of cytokine-secreting CD8(+) T cells. This fusion protein formed oligomers/aggregates that led to activation of STAT-1 and IFN regulatory factor-3 in human macrophages, indicating a type I IFN response, resulting in NO, IL-12, and IL-6 induction. Furthermore, this vaccination regimen inhibited the liver stage development of the parasite, resulting in sterile protection. In summary, we propose a novel approach in designing CS based pre-erythrocytic vaccines against Plasmodium using the adjuvant-like effect of the immunogenic vaccinia virus protein 14K.
Collapse
Affiliation(s)
- Aneesh Vijayan
- Departamento de Biología Celular y Molecular, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Cientificas, 28049 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
22
|
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
|
23
|
Wenzel UA, Bank E, Florian C, Förster S, Zimara N, Steinacker J, Klinger M, Reiling N, Ritter U, van Zandbergen G. Leishmania major parasite stage-dependent host cell invasion and immune evasion. FASEB J 2011; 26:29-39. [PMID: 21908716 DOI: 10.1096/fj.11-184895] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Leishmania pathogenesis is primarily studied using the disease-inducing promastigote stage of Leishmania major. Despite many efforts, all attempts so far have failed to culture the disease-relevant multiplying amastigote stage of L. major. Here, we established a stably growing axenic L. major amastigote culture system that was characterized genetically, morphologically, and by stage-specific DsRed protein expression. We found parasite stage-specific disease development in resistant C57BL/6 mice. Human neutrophils, as first host cells for promastigotes, do not take up amastigotes. In human macrophages, we observed an amastigote-specific complement receptor 3-mediated, endocytotic entry mechanism, whereas promastigotes are taken up by complement receptor 1-mediated phagocytosis. Promastigote infection of macrophages induced the inflammatory mediators TNF, CCL3, and CCL4, whereas amastigote infection was silent and resulted in significantly increased parasite numbers: from 7.1 ± 1.4 (after 3 h) to 20.1 ± 7.9 parasites/cell (after 96 h). Our study identifies Leishmania stage-specific disease development, host cell preference, entry mechanism, and immune evasion. Since the amastigote stage is the disease-propagating form found in the infected mammalian host, the newly developed L. major axenic cultures will serve as an important tool in better understanding the amastigote-driven immune response in leishmaniasis.
Collapse
Affiliation(s)
- Ulf Alexander Wenzel
- Division of Immunology, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Straße, Langen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Pasieka TJ, Collins L, O'Connor MA, Chen Y, Parker ZM, Berwin BL, Piwnica-Worms DR, Leib DA. Bioluminescent imaging reveals divergent viral pathogenesis in two strains of Stat1-deficient mice, and in αßγ interferon receptor-deficient mice. PLoS One 2011; 6:e24018. [PMID: 21915277 PMCID: PMC3168466 DOI: 10.1371/journal.pone.0024018] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 08/03/2011] [Indexed: 01/14/2023] Open
Abstract
Pivotal components of the IFN response to virus infection include the IFN receptors (IFNR), and the downstream factor signal transducer and activator of transcription 1 (Stat1). Mice deficient for Stat1 and IFNR (Stat1(-/-) and IFNαßγR(-/-) mice) lack responsiveness to IFN and exhibit high sensitivity to various pathogens. Here we examined herpes simplex virus type 1 (HSV-1) pathogenesis in Stat1(-/-) mice and in IFNαßγR(-/-) mice following corneal infection and bioluminescent imaging. Two divergent and paradoxical patterns of infection were observed. Mice with an N-terminal deletion in Stat1 (129Stat1(-/-) (N-term)) had transient infection of the liver and spleen, but succumbed to encephalitis by day 10 post-infection. In stark contrast, infection of IFNαßγR(-/-) mice was rapidly fatal, with associated viremia and fulminant infection of the liver and spleen, with infected infiltrating cells being primarily of the monocyte/macrophage lineage. To resolve the surprising difference between Stat1(-/-) and IFNαßγR(-/-) mice, we infected an additional Stat1(-/-) strain deleted in the DNA-binding domain (129Stat1(-/-) (DBD)). These 129Stat1(-/-) (DBD) mice recapitulated the lethal pattern of liver and spleen infection seen following infection of IFNαßγR(-/-) mice. This lethal pattern was also observed when 129Stat1(-/-) (N-term) mice were infected and treated with a Type I IFN-blocking antibody, and immune cells derived from 129Stat1(-/-) (N-term) mice were shown to be responsive to Type I IFN. These data therefore show significant differences in viral pathogenesis between two commonly-used Stat1(-/-) mouse strains. The data are consistent with the hypothesis that Stat1(-/-) (N-term) mice have residual Type I IFN receptor-dependent IFN responses. Complete loss of IFN signaling pathways allows viremia and rapid viral spread with a fatal infection of the liver. This study underscores the importance of careful comparisons between knockout mouse strains in viral pathogenesis, and may also be relevant to the causation of HSV hepatitis in humans, a rare but frequently fatal infection.
Collapse
Affiliation(s)
- Tracy Jo Pasieka
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Lynne Collins
- BRIGHT Institute, Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Megan A. O'Connor
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Yufei Chen
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Zachary M. Parker
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Brent L. Berwin
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - David R. Piwnica-Worms
- BRIGHT Institute, Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - David A. Leib
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
25
|
Abstract
Leishmania is a genus of protozoan parasites that are transmitted by the bite of phlebotomine sandflies and give rise to a range of diseases (collectively known as leishmaniases) that affect over 150 million people worldwide. Cellular immune mechanisms have a major role in the control of infections with all Leishmania spp. However, as discussed in this Review, recent evidence suggests that each host-pathogen combination evokes different solutions to the problems of parasite establishment, survival and persistence. Understanding the extent of this diversity will be increasingly important in ensuring the development of broadly applicable vaccines, drugs and immunotherapeutic interventions.
Collapse
Affiliation(s)
- Paul Kaye
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, Wentworth Way, UK.
| | | |
Collapse
|
26
|
Coursey TG, Chen PW, Niederkorn JY. Abrogating TNF-α expression prevents bystander destruction of normal tissues during iNOS-mediated elimination of intraocular tumors. Cancer Res 2011; 71:2445-54. [PMID: 21307132 PMCID: PMC3108159 DOI: 10.1158/0008-5472.can-10-2628] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although intraocular tumors reside in an immune privileged site, some tumors are rejected nonetheless. For example, intraocular adenovirus-induced (Ad5E1; adenovirus type 5 early region 1) tumors are rejected in syngeneic C57BL/6 mice by one of two pathways. One pathway leads to extensive necrosis of innocent bystander cells and culminates in destruction of the eye, a condition called phthisis. The second pathway is characterized by piecemeal tumor cell death that rids the eye of the tumor while preserving the architecture and function of the eye. To study the mechanisms of phthisical tumor rejection, we isolated a cell clone-designated clone 2.1 that consistently undergoes rejection in a phthisical manner. CD4(+) T cells and macrophages were required for phthisical rejection of intraocular clone 2.1 tumors and M1 macrophages were involved in mediating tumor rejection. In vitro and in vivo inhibition of iNOS (inducible nitric oxide synthase) abolished macrophage-mediated killing of tumor cells and rejection of intraocular tumors. A role for M1 macrophages was further supported by investigations showing that intraocular tumors grew progressively in IFN-γ KO (knockout) mice. Studies in mice deficient in TNF-α, TNF receptor-1, or TNF receptor-2 revealed that although TNF-α was not needed for tumor rejection, it was required for the development of necrotizing inflammation and phthisis of tumor-bearing eyes. Together, our findings suggest new strategies to successfully eliminate ocular tumors while preserving the integrity of the eye.
Collapse
Affiliation(s)
- Terry G Coursey
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | | | | |
Collapse
|
27
|
Fusion between Leishmania amazonensis and Leishmania major parasitophorous vacuoles: live imaging of coinfected macrophages. PLoS Negl Trop Dis 2010; 4:e905. [PMID: 21151877 PMCID: PMC2998430 DOI: 10.1371/journal.pntd.0000905] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 11/03/2010] [Indexed: 12/14/2022] Open
Abstract
Protozoan parasites of the genus Leishmania alternate between flagellated, elongated extracellular promastigotes found in insect vectors, and round-shaped amastigotes enclosed in phagolysosome-like Parasitophorous Vacuoles (PVs) of infected mammalian host cells. Leishmania amazonensis amastigotes occupy large PVs which may contain many parasites; in contrast, single amastigotes of Leishmania major lodge in small, tight PVs, which undergo fission as parasites divide. To determine if PVs of these Leishmania species can fuse with each other, mouse macrophages in culture were infected with non-fluorescent L. amazonensis amastigotes and, 48 h later, superinfected with fluorescent L. major amastigotes or promastigotes. Fusion was investigated by time-lapse image acquisition of living cells and inferred from the colocalization of parasites of the two species in the same PVs. Survival, multiplication and differentiation of parasites that did or did not share the same vacuoles were also investigated. Fusion of PVs containing L. amazonensis and L. major amastigotes was not found. However, PVs containing L. major promastigotes did fuse with pre-established L. amazonensis PVs. In these chimeric vacuoles, L. major promastigotes remained motile and multiplied, but did not differentiate into amastigotes. In contrast, in doubly infected cells, within their own, unfused PVs metacyclic-enriched L. major promastigotes, but not log phase promastigotes - which were destroyed - differentiated into proliferating amastigotes. The results indicate that PVs, presumably customized by L. major amastigotes or promastigotes, differ in their ability to fuse with L. amazonensis PVs. Additionally, a species-specific PV was required for L. major destruction or differentiation – a requirement for which mechanisms remain unknown. The observations reported in this paper should be useful in further studies of the interactions between PVs to different species of Leishmania parasites, and of the mechanisms involved in the recognition and fusion of PVs. Many non-viral intracellular pathogens lodge within cell vesicles known as “parasitophorous vacuoles” (PVs), which exhibit a variety of pathogen-dependent functional and compositional phenotypes. PVs of the protozoan Leishmania are similar to the digestive organelles known as phagolysosomes. We asked if, in phagocytes infected with two different Leishmania species, would the two parasites be found in the same or in separate vacuoles? Of the species chosen, Leishmania amazonensis develops within large vacuoles which shelter many parasites; in contrast, Leishmania major lodges in small PVs containing one or two parasites. In the present experiments, the species and their life-cycle stages (extracellular promastigotes, and intracellular amastigotes) were distinguished by means of fluorescent markers, and the intracellular localization of the parasites was examined in living cells. We report here that, whereas L. major amastigotes remained within their individual vacuoles, L. major promastigotes were delivered to L. amazonensis vacuoles, in which they survived and multiplied but were unable to differentiate into amastigotes. A species-specific vacuole was thus required for L. major differentiation. The model should be useful in cellular and molecular studies of the biology of these parasites and of their parasitophorous vacuoles.
Collapse
|
28
|
Vinet AF, Fukuda M, Turco SJ, Descoteaux A. The Leishmania donovani lipophosphoglycan excludes the vesicular proton-ATPase from phagosomes by impairing the recruitment of synaptotagmin V. PLoS Pathog 2009; 5:e1000628. [PMID: 19834555 PMCID: PMC2757729 DOI: 10.1371/journal.ppat.1000628] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 09/23/2009] [Indexed: 12/20/2022] Open
Abstract
We recently showed that the exocytosis regulator Synaptotagmin (Syt) V is recruited to the nascent phagosome and remains associated throughout the maturation process. In this study, we investigated the possibility that Syt V plays a role in regulating interactions between the phagosome and the endocytic organelles. Silencing of Syt V by RNA interference revealed that Syt V contributes to phagolysosome biogenesis by regulating the acquisition of cathepsin D and the vesicular proton-ATPase. In contrast, recruitment of cathepsin B, the early endosomal marker EEA1 and the lysosomal marker LAMP1 to phagosomes was normal in the absence of Syt V. As Leishmania donovani promastigotes inhibit phagosome maturation, we investigated their potential impact on the phagosomal association of Syt V. This inhibition of phagolysosome biogenesis is mediated by the virulence glycolipid lipophosphoglycan, a polymer of the repeating Galbeta1,4Manalpha1-PO(4) units attached to the promastigote surface via an unusual glycosylphosphatidylinositol anchor. Our results showed that insertion of lipophosphoglycan into ganglioside GM1-containing microdomains excluded or caused dissociation of Syt V from phagosome membranes. As a consequence, L. donovani promatigotes established infection in a phagosome from which the vesicular proton-ATPase was excluded and which failed to acidify. Collectively, these results reveal a novel function for Syt V in phagolysosome biogenesis and provide novel insight into the mechanism of vesicular proton-ATPase recruitment to maturing phagosomes. We also provide novel findings into the mechanism of Leishmania pathogenesis, whereby targeting of Syt V is part of the strategy used by L. donovani promastigotes to prevent phagosome acidification.
Collapse
Affiliation(s)
- Adrien F. Vinet
- INRS-Institut Armand-Frappier and Centre for Host-Parasite Interactions, Laval, Québec, Canada
| | - Mitsunori Fukuda
- Department of Developmental Biology and Neurosciences, Tohoku University, Sendai, Miyagi, Japan
| | - Salvatore J. Turco
- Department of Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Albert Descoteaux
- INRS-Institut Armand-Frappier and Centre for Host-Parasite Interactions, Laval, Québec, Canada
- * E-mail:
| |
Collapse
|