1
|
Akand SK, Rahman A, Masood M, Tabrez S, Saleem M, Ahmed MZ, Akhter Y, Haque MM, Rub A. hsa-miR-330-5p regulates serine palmitoyltransferase long chain base subunit 1 and augments host protective immune response against Leishmania donovani infection. Arch Microbiol 2025; 207:123. [PMID: 40237871 DOI: 10.1007/s00203-025-04325-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/09/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Leishmaniasis, caused by the protozoan parasites of the genus Leishmania, poses a significant global health challenge, particularly in the resource-limited regions where it causes high mortality. Regardless in the progress of treatment strategies, the emergence of drug resistance and limited efficacy requires the search of novel therapy and therapeutic targets. MicroRNAs, the crucial post-transcriptional regulators of gene expression, play critical roles in host-pathogen interactions. Here, we screened the miRNAs dysregulated during Leishmania donovani infection through literature search. hsa-miR-330-5p, one of the miRNAs which through human KEGG 2021 and Human Cyc 2016 analysis was found to be involved in multiple pathways including sphingolipid signaling pathway. Sphingolipids are important class of lipids involved in different cellular processes and therefore are the targets of many pathogens including Leishmania. hsa-miR-330-5p was found downregulated after 24 h of Leishmania donovani infection in THP-1 derived human macrophages. Target prediction of sphingolipid biosynthetic genes through in silico prediction tools showed 3/ UTR of serine palmitoyltransferase long chain base subunit 1 to be a target of hsa-miR-330-5p. The in silico target prediction of hsa-miR-330-5p was validated by cloning the 3/ UTR target sequence of gene, transfecting and performing luciferase assay in HEK 293 T cell line. Transfection of mimic of hsa-miR-330-5p reduced the luciferase activity which validated the in silico target prediction. Further, mimic of hsa-miR-330-5p inhibited the expression of the target gene, serine palmitoyltransferase long chain base subunit 1 and augmented the expression of pro-inflammatory cytokines in L. donovani infected THP-1 derived macrophages. Mimic of hsa-miR-330-5p also led to a significant reduction in the intracellular parasite burden in both THP-1 derived as well as primary human macrophages. This study has not only identified the sphingolipid biosynthesis regulatory miRNA but will also help in the development of novel and effective treatment strategy against leishmaniasis in future.
Collapse
Affiliation(s)
- Sajjadul Kadir Akand
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Areeba Rahman
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Mohammad Masood
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Shams Tabrez
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
| | - Mohammad Saleem
- Faculty of Dentistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Mohammad Z Ahmed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, 11451, Riyadh, Kingdom of Saudi Arabia
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India
| | - Mohammad Mahfuzul Haque
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Abdur Rub
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India.
| |
Collapse
|
2
|
Lee YT, Senturk M, Guan Y, Wang MC. Bacteria-organelle communication in physiology and disease. J Cell Biol 2024; 223:e202310134. [PMID: 38748249 PMCID: PMC11096858 DOI: 10.1083/jcb.202310134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/03/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
Bacteria, omnipresent in our environment and coexisting within our body, exert dual beneficial and pathogenic influences. These microorganisms engage in intricate interactions with the human body, impacting both human health and disease. Simultaneously, certain organelles within our cells share an evolutionary relationship with bacteria, particularly mitochondria, best known for their energy production role and their dynamic interaction with each other and other organelles. In recent years, communication between bacteria and mitochondria has emerged as a new mechanism for regulating the host's physiology and pathology. In this review, we delve into the dynamic communications between bacteria and host mitochondria, shedding light on their collaborative regulation of host immune response, metabolism, aging, and longevity. Additionally, we discuss bacterial interactions with other organelles, including chloroplasts, lysosomes, and the endoplasmic reticulum (ER).
Collapse
Affiliation(s)
- Yi-Tang Lee
- Waisman Center, University of Wisconsin, Madison, WI, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Integrative Program of Molecular and Biochemical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Mumine Senturk
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Youchen Guan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Meng C. Wang
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| |
Collapse
|
3
|
Carvalho Cabral P, Stegeman SK, Olivier M, Cermakian N. Circadian Regulation of Leishmania Parasite Internalisation in Macrophages and Downstream Cellular Events. Parasite Immunol 2024; 46:e13053. [PMID: 38817112 DOI: 10.1111/pim.13053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024]
Abstract
Leishmania spp. parasites use macrophages as a host cell during infection. As a result, macrophages have a dual role: clearing the parasite as well as acting as host cells. Recently, studies have shown that macrophages harbour circadian clocks, which affect many of their functions such as phagocytosis, receptor expression and cytokine release. Interestingly, Leishmania major infection in hosts was also shown to be under circadian control. Therefore, we decided to investigate what underlies the rhythms of L. major infection within macrophages. Using a culture model of infection of bone marrow-derived macrophages with L. major promastigotes, we show that the parasites are internalised into macrophages with a 24-h variation dependent on a functional circadian clock in the cells. This was associated with a variation in the number of parasites per macrophage. The cell surface expression of parasite receptors was not controlled by the cells' circadian clock. In contrast, the expression of the components of the endocytic pathway, EEA1 and LC3b, varied according to the time of infection. This was paralleled by variations in parasite-induced ROS production as well as cytokine tumour necrosis factor α. In summary, we have uncovered a time-dependent regulation of the internalisation of L. major promastigotes in macrophages, controlled by the circadian clock in these cells, as well as subsequent cellular events in the endocytic pathway, intracellular signalling and cytokine production.
Collapse
Affiliation(s)
| | - Sophia K Stegeman
- Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Martin Olivier
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Nicolas Cermakian
- Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Moreno-Corona NC, de León-Bautista MP, León-Juárez M, Hernández-Flores A, Barragán-Gálvez JC, López-Ortega O. Rab GTPases, Active Members in Antigen-Presenting Cells, and T Lymphocytes. Traffic 2024; 25:e12950. [PMID: 38923715 DOI: 10.1111/tra.12950] [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: 01/07/2024] [Revised: 04/25/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024]
Abstract
Processes such as cell migration, phagocytosis, endocytosis, and exocytosis refer to the intense exchange of information between the internal and external environment in the cells, known as vesicular trafficking. In eukaryotic cells, these essential cellular crosstalks are controlled by Rab GTPases proteins through diverse adaptor proteins like SNAREs complex, coat proteins, phospholipids, kinases, phosphatases, molecular motors, actin, or tubulin cytoskeleton, among others, all necessary for appropriate mobilization of vesicles and distribution of molecules. Considering these molecular events, Rab GTPases are critical components in specific biological processes of immune cells, and many reports refer primarily to macrophages; therefore, in this review, we address specific functions in immune cells, concretely in the mechanism by which the GTPase contributes in dendritic cells (DCs) and, T/B lymphocytes.
Collapse
Affiliation(s)
| | - Mercedes Piedad de León-Bautista
- Escuela de Medicina, Universidad Vasco de Quiroga, Morelia, Mexico
- Human Health, Laboratorio de Enfermedades Infecciosas y Genómica (INEX LAB), Morelia, Mexico
| | - Moises León-Juárez
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | | | - Juan Carlos Barragán-Gálvez
- División de Ciencias Naturales y Exactas, Departamento de Farmacia, Universidad de Guanajuato, Guanajuato, Mexico
| | - Orestes López-Ortega
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institute Necker Enfants Malades, Paris, France
| |
Collapse
|
5
|
Sood C, Verma JK, Basak R, Kapoor A, Gupta S, Mukhopadhyay A. Leishmania highjack host lipid body for its proliferation in macrophages by overexpressing host Rab18 and TRAPPC9 by downregulating miR-1914-3p expression. PLoS Pathog 2024; 20:e1012024. [PMID: 38412149 PMCID: PMC10898768 DOI: 10.1371/journal.ppat.1012024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024] Open
Abstract
Lipids stored in lipid-bodies (LBs) in host cells are potential sources of fatty acids for pathogens. However, the mechanism of recruitment of LBs from the host cells by pathogens to acquire fatty acids is not known. Here, we have found that Leishmania specifically upregulates the expression of host Rab18 and its GEF, TRAPPC9 by downregulating the expression of miR-1914-3p by reducing the level of Dicer in macrophages via their metalloprotease gp63. Our results also show that miR-1914-3p negatively regulates the expression of Rab18 and its GEF in cells. Subsequently, Leishmania containing parasitophorous vacuoles (Ld-PVs) recruit and retain host Rab18 and TRAPPC9. Leishmania infection also induces LB biogenesis in host cells and recruits LBs on Ld-PVs and acquires FLC12-labeled fatty acids from LBs. Moreover, overexpression of miR-1914-3p in macrophages significantly inhibits the recruitment of LBs and thereby suppresses the multiplication of parasites in macrophages as parasites are unable to acquire fatty acids. These results demonstrate a novel mechanism how Leishmania acquire fatty acids from LBs for their growth in macrophages.
Collapse
Affiliation(s)
- Chandni Sood
- National Institute of Immunology, New Delhi, India
| | - Jitender Kumar Verma
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
- National Institute of Immunology, New Delhi, India
| | - Rituparna Basak
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| | - Anjali Kapoor
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| | - Swarnima Gupta
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| | - Amitabha Mukhopadhyay
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
- National Institute of Immunology, New Delhi, India
| |
Collapse
|
6
|
Baert L, Rudy S, Pellisson M, Doll T, Rocchetti R, Kaiser M, Mäser P, Müller M. Induced pluripotent stem cell-derived human macrophages as an infection model for Leishmania donovani. PLoS Negl Trop Dis 2024; 18:e0011559. [PMID: 38166146 PMCID: PMC10786377 DOI: 10.1371/journal.pntd.0011559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/12/2024] [Accepted: 12/19/2023] [Indexed: 01/04/2024] Open
Abstract
The parasite Leishmania donovani is one of the species causing visceral leishmaniasis in humans, a deadly infection claiming up to 40,000 lives each year. The current drugs for leishmaniasis treatment have severe drawbacks and there is an urgent need to find new anti-leishmanial compounds. However, the search for drug candidates is complicated by the intracellular lifestyle of Leishmania. Here, we investigate the use of human induced pluripotent stem cell (iPS)-derived macrophages (iMACs) as host cells for L. donovani. iMACs obtained through embryoid body differentiation were infected with L. donovani promastigotes, and high-content imaging techniques were used to optimize the iMACs seeding density and multiplicity of infection, allowing us to reach infection rates up to 70% five days after infection. IC50 values obtained for miltefosine and amphotericin B using the infected iMACs or mouse peritoneal macrophages as host cells were comparable and in agreement with the literature, showing the potential of iMACs as an infection model for drug screening.
Collapse
Affiliation(s)
- Lore Baert
- Swiss Tropical and Public Health Institute (SwissTPH), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Serena Rudy
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Mélanie Pellisson
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Thierry Doll
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Romina Rocchetti
- Swiss Tropical and Public Health Institute (SwissTPH), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute (SwissTPH), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute (SwissTPH), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Matthias Müller
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
7
|
Wu KC, Condon ND, Hill TA, Reid RC, Fairlie DP, Lim J. Ras-Related Protein Rab5a Regulates Complement C5a Receptor Trafficking, Chemotaxis, and Chemokine Secretion in Human Macrophages. J Innate Immun 2023; 15:468-484. [PMID: 36882040 PMCID: PMC10105068 DOI: 10.1159/000530012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 03/01/2023] [Indexed: 03/09/2023] Open
Abstract
Complement activation and Rab GTPase trafficking are commonly observed in inflammatory responses. Recruitment of innate immune cells to sites of infection or injury and secretion of inflammatory chemokines are promoted by complement component 5a (C5a) that activates the cell surface protein C5a receptor1 (C5aR1). Persistent activation can lead to a myriad of inflammatory and autoimmune diseases. Here, we demonstrate that the mechanism of C5a induced chemotaxis of human monocyte-derived macrophages (HMDMs) and their secretion of inflammatory chemokines are controlled by Rab5a. We find that C5a activation of the G protein coupled receptor C5aR1 expressed on the surface of HMDMs, recruits β-arrestin2 via Rab5a trafficking, then activates downstream phosphatidylinositol 3-kinase (PI3K)/Akt signaling that culminates in chemotaxis and secretion of pro-inflammatory chemokines from HMDMs. High-resolution lattice light-sheet microscopy on live cells showed that C5a activates C5aR1-GFP internalization and colocalization with Rab5a-tdTomato but not with dominant negative mutant Rab5a-S34N-tdTomato in HEK293 cells. We found that Rab5a is significantly upregulated in differentiated HMDMs and internalization of C5aR1 is dependent on Rab5a. Interestingly, while knockdown of Rab5a inhibited C5aR1-mediated Akt phosphorylation, it did not affect C5aR1-mediated ERK1/2 phosphorylation or intracellular calcium mobilization in HMDMs. Functional analysis using transwell migration and µ-slide chemotaxis assays indicated that Rab5a regulates C5a-induced chemotaxis of HMDMs. Further, C5aR1 was found to mediate interaction of Rab5a with β-arrestin2 but not with G proteins in HMDMs. Furthermore, C5a-induced secretion of pro-inflammatory chemokines (CCL2, CCL3) from HMDMs was attenuated by Rab5a or β-arrestin2 knockdown or by pharmacological inhibition with a C5aR1 antagonist or a PI3K inhibitor. These findings reveal a C5a-C5aR1-β-arrestin2-Rab5a-PI3K signaling pathway that regulates chemotaxis and pro-inflammatory chemokine secretion in HMDMs and suggests new ways of selectively modulating C5a-induced inflammatory outputs.
Collapse
Affiliation(s)
- Kai-Chen Wu
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Nicholas D. Condon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy A. Hill
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Robert C. Reid
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David P. Fairlie
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Junxian Lim
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
8
|
Selvapandiyan A, Puri N, Kumar P, Alam A, Ehtesham NZ, Griffin G, Hasnain SE. Zooming in on common immune evasion mechanisms of pathogens in phagolysosomes: potential broad-spectrum therapeutic targets against infectious diseases. FEMS Microbiol Rev 2023; 47:6780197. [PMID: 36309472 DOI: 10.1093/femsre/fuac041] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 01/19/2023] Open
Abstract
The intracellular viral, bacterial, or parasitic pathogens evade the host immune challenges to propagate and cause fatal diseases. The microbes overpower host immunity at various levels including during entry into host cells, phagosome formation, phagosome maturation, phagosome-lysosome fusion forming phagolysosomes, acidification of phagolysosomes, and at times after escape into the cytosol. Phagolysosome is the final organelle in the phagocyte with sophisticated mechanisms to degrade the pathogens. The immune evasion strategies by the pathogens include the arrest of host cell apoptosis, decrease in reactive oxygen species, the elevation of Th2 anti-inflammatory response, avoidance of autophagy and antigen cross-presentation pathways, and escape from phagolysosomal killing. Since the phagolysosome organelle in relation to infection/cure is seldom discussed in the literature, we summarize here the common host as well as pathogen targets manipulated or utilized by the pathogens established in phagosomes and phagolysosomes, to hijack the host immune system for their benefit. These common molecules or pathways can be broad-spectrum therapeutic targets for drug development for intervention against infectious diseases caused by different intracellular pathogens.
Collapse
Affiliation(s)
| | - Niti Puri
- Cellular and Molecular Immunology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pankaj Kumar
- Department of Biochemistry, Jamia Hamdard, New Delhi, 110062, India.,Centre for Tuberculosis Research, Department of Medicine, Johns Hopkins University, Baltimore, MD, 21218, United States
| | - Anwar Alam
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India.,Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India
| | - Nasreen Zafar Ehtesham
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
| | - George Griffin
- Department of Cellular and Molecular Medicine, St. George's University of London, London, SW17 0RE, United Kingdom
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India.,Department of Life Science, School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, 201310, India
| |
Collapse
|
9
|
Jafarzadeh A, Nemati M, Aminizadeh N, Bodhale N, Sarkar A, Jafarzadeh S, Sharifi I, Saha B. Bidirectional cytokine-microRNA control: A novel immunoregulatory framework in leishmaniasis. PLoS Pathog 2022; 18:e1010696. [PMID: 35925884 PMCID: PMC9351994 DOI: 10.1371/journal.ppat.1010696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
As effector innate immune cells and as a host to the protozoan parasite Leishmania, macrophages play a dual role in antileishmanial immunoregulation. The 2 key players in this immunoregulation are the macrophage-expressed microRNAs (miRNAs) and the macrophage-secreted cytokines. miRNAs, as small noncoding RNAs, play vital roles in macrophage functions including cytokines and chemokines production. In the reverse direction, Leishmania-regulated cytokines alter miRNAs expression to regulate the antileishmanial functions of macrophages. The miRNA patterns vary with the time and stage of infection. The cytokine-regulated macrophage miRNAs not only help parasite elimination or persistence but also regulate cytokine production from macrophages. Based on these observations, we propose a novel immunoregulatory framework as a scientific rationale for antileishmanial therapy.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- * E-mail: (AJ); (BS)
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Najmeh Aminizadeh
- Department of Histology, School of Medicine, Islamic Azad University Branch of Kerman, Kerman
| | | | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, Odisha, India
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Bhaskar Saha
- National Centre For Cell Science, Pune, India
- Trident Academy of Creative Technology, Bhubaneswar, Odisha, India
- * E-mail: (AJ); (BS)
| |
Collapse
|
10
|
Díaz E, Febres A, Giammarresi M, Silva A, Vanegas O, Gomes C, Ponte-Sucre A. G Protein-Coupled Receptors as Potential Intercellular Communication Mediators in Trypanosomatidae. Front Cell Infect Microbiol 2022; 12:812848. [PMID: 35651757 PMCID: PMC9149261 DOI: 10.3389/fcimb.2022.812848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Detection and transduction of environmental signals, constitute a prerequisite for successful parasite invasion; i.e., Leishmania transmission, survival, pathogenesis and disease manifestation and dissemination, with diverse molecules functioning as inter-cellular signaling ligands. Receptors [i.e., G protein-coupled receptors (GPCRs)] and their associated transduction mechanisms, well conserved through evolution, specialize in this function. However, canonical GPCR-related signal transduction systems have not been described in Leishmania, although orthologs, with reduced domains and function, have been identified in Trypanosomatidae. These inter-cellular communication means seem to be essential for multicellular and unicellular organism’s survival. GPCRs are flexible in their molecular architecture and may interact with the so-called receptor activity-modifying proteins (RAMPs), which modulate their function, changing GPCRs pharmacology, acting as chaperones and regulating signaling and/or trafficking in a receptor-dependent manner. In the skin, vasoactive- and neuro- peptides released in response to the noxious stimuli represented by the insect bite may trigger parasite physiological responses, for example, chemotaxis. For instance, in Leishmania (V.) braziliensis, sensory [Substance P, SP, chemoattractant] and autonomic [Vasoactive Intestinal Peptide, VIP, and Neuropeptide Y, NPY, chemorepellent] neuropeptides at physiological levels stimulate in vitro effects on parasite taxis. VIP and NPY chemotactic effects are impaired by their corresponding receptor antagonists, suggesting that the stimulated responses might be mediated by putative GPCRs (with essential conserved receptor domains); the effect of SP is blocked by [(D-Pro 2, D-Trp7,9]-Substance P (10-6 M)] suggesting that it might be mediated by neurokinin-1 transmembrane receptors. Additionally, vasoactive molecules like Calcitonin Gene-Related Peptide [CGRP] and Adrenomedullin [AM], exert a chemorepellent effect and increase the expression of a 24 kDa band recognized in western blot analysis by (human-)-RAMP-2 antibodies. In-silico search oriented towards GPCRs-like receptors and signaling cascades detected a RAMP-2-aligned sequence corresponding to Leishmania folylpolyglutamate synthase and a RAMP-3 aligned protein, a hypothetical Leishmania protein with yet unknown function, suggesting that in Leishmania, CGRP and AM activities may be modulated by RAMP- (-2) and (-3) homologs. The possible presence of proteins and molecules potentially involved in GPCRs cascades, i.e., RAMPs, signpost conservation of ancient signaling systems associated with responses, fundamental for cell survival, (i.e., taxis and migration) and may constitute an open field for description of pharmacophores against Leishmania parasites.
Collapse
Affiliation(s)
- Emilia Díaz
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, Caracas, Venezuela
| | - Anthony Febres
- Section of Infectious Diseases, Baylor College of Medicine, TX, United States
| | - Michelle Giammarresi
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, Caracas, Venezuela
| | - Adrian Silva
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, Caracas, Venezuela
| | - Oriana Vanegas
- Pediatric Gastroenterology, University of Iowa, Iowa City, IA, United States
| | - Carlos Gomes
- Royal Berkshire NHS, Foundation Trust, Light House Lab, Bracknell, United Kingdom
| | - Alicia Ponte-Sucre
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, Caracas, Venezuela
- Medical Mission Institute, Würzburg, Germany
- *Correspondence: Alicia Ponte-Sucre,
| |
Collapse
|
11
|
Chang KP, Reynolds JM, Liu Y, He JJ. Leishmaniac Quest for Developing a Novel Vaccine Platform. Is a Roadmap for Its Advances Provided by the Mad Dash to Produce Vaccines for COVID-19? Vaccines (Basel) 2022; 10:vaccines10020248. [PMID: 35214706 PMCID: PMC8874365 DOI: 10.3390/vaccines10020248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 02/04/2023] Open
Abstract
“Bugs as drugs” in medicine encompasses the use of microbes to enhance the efficacy of vaccination, such as the delivery of vaccines by Leishmania—the protozoan etiological agent of leishmaniasis. This novel approach is appraised in light of the successful development of vaccines for Covid-19. All relevant aspects of this pandemic are summarized to provide the necessary framework in contrast to leishmaniasis. The presentation is in a side-by-side matching format with particular emphasis on vaccines. The comparative approach makes it possible to highlight the timeframe of the vaccine workflows condensed by the caveats of pandemic urgency and, at the same time, provides the background of Leishmania behind its use as a vaccine carrier. Previous studies in support of the latter are summarized as follows. Leishmaniasis confers life-long immunity on patients after cure, suggesting the effective vaccination is achievable with whole-cell Leishmania. A new strategy was developed to inactivate these cells in vitro, rendering them non-viable, hence non-disease causing, albeit retaining their immunogenicity and adjuvanticity. This was achieved by installing a dual suicidal mechanism in Leishmania for singlet oxygen (1O2)-initiated inactivation. In vitro cultured Leishmania were genetically engineered for cytosolic accumulation of UV-sensitive uroporphyrin I and further loaded endosomally with a red light-sensitive cationic phthalocyanine. Exposing these doubly dye-loaded Leishmania to light triggers intracellular production of highly reactive but extremely short-lived 1O2, resulting in their rapid and complete inactivation. Immunization of susceptible animals with such inactivated Leishmania elicited immunity to protect them against experimental leishmaniasis. Significantly, the inactivated Leishmania was shown to effectively deliver transgenically add-on ovalbumin (OVA) to antigen-presenting cells (APC), wherein OVA epitopes were processed appropriately for presentation with MHC molecules to activate epitope-specific CD8+ T cells. Application of this approach to deliver cancer vaccine candidates, e.g., enolase-1, was shown to suppress tumor development in mouse models. A similar approach is predicted to elicit lasting immunity against infectious diseases, including complementation of the spike protein-based vaccines in use for COVID-19. This pandemic is devastating, but brings to light the necessity of considering many facets of the disease in developing vaccination programs. Closer collaboration is essential among those in diverse disciplinary areas to provide the roadmap toward greater success in the future. Highlighted herein are several specific issues of vaccinology and new approaches worthy of consideration due to the pandemic.
Collapse
|
12
|
Rashidi S, Mansouri R, Ali-Hassanzadeh M, Ghani E, Barazesh A, Karimazar M, Nguewa P, Carrera Silva EA. Highlighting the interplay of microRNAs from Leishmania parasites and infected-host cells. Parasitology 2021; 148:1434-1446. [PMID: 34218829 PMCID: PMC11010138 DOI: 10.1017/s0031182021001177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/10/2021] [Accepted: 06/27/2021] [Indexed: 02/05/2023]
Abstract
Leishmania parasites, the causative agents of leishmaniasis, are protozoan parasites with the ability to modify the signalling pathway and cell responses of their infected host cells. These parasite strategies alter the host cell environment and conditions favouring their replication, survival and pathogenesis. Since microRNAs (miRNAs) are able to post-transcriptionally regulate gene expression processes, these biomolecules can exert critical roles in controlling Leishmania-host cell interplay. Therefore, the identification of relevant miRNAs differentially expressed in Leishmania parasites as well as in infected cells, which affect the host fitness, could be critical to understand the infection biology, pathogenicity and immune response against these parasites. Accordingly, the current review aims to address the differentially expressed miRNAs in both, the parasite and infected host cells and how these biomolecules change cell signalling and host immune responses during infection. A deep understanding of these processes could provide novel guidelines and therapeutic strategies for managing and treating leishmaniasis.
Collapse
Affiliation(s)
- Sajad Rashidi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Esmaeel Ghani
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afshin Barazesh
- Department of Microbiology and Parasitology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammadreza Karimazar
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paul Nguewa
- University of Navarra, ISTUN Instituto de Salud Tropical, Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), c/Irunlarrea 1, 31008Pamplona, Spain
| | | |
Collapse
|
13
|
Pradhan S, Ghosh S, Hussain S, Paul J, Mukherjee B. Linking membrane fluidity with defective antigen presentation in leishmaniasis. Parasite Immunol 2021; 43:e12835. [PMID: 33756007 DOI: 10.1111/pim.12835] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/11/2022]
Abstract
Hampering-surface presentation of immunogenic peptides by class I/II MHCs is a key strategy opted by several intracellular protozoan pathogens including Leishmania to escape CD8/CD4 mediated host-protective T-cell response. Although Leishmania parasites (LP) primarily hijack/inhibit host lysosomal/proteasomal pathways to hamper antigen-processing/presentation machinery, recent pieces of evidence have linked host-membrane fluidity as a major cause of defective antigen presentation in leishmaniasis. Increased membrane fluidity severely compromised peptide-MHC stability in the lipid raft regions, thereby abrogating T-cell mediated-signalling in the infected host. LP primarily achieves this by quenching host cholesterol, which acts as cementing material in maintaining the membrane fluidity. In this review, we have particularly focused on several strategies opted by LP to hijack-host cholesterol resulting in lipid droplets accumulation around leishmania-containing parasitophorous vacuole favouring intracellular survival of LP. In fact, LP infection can result in altered cholesterol and lipid metabolism in the infected host, thereby favouring the establishment and progression of the infection. From our analysis of two genome-wide transcriptomics data sets of LP infected host, we propose a possible molecular network that connects these interrelated events of altered lipid metabolism with eventual compromised antigen presentation, still existing as a gap in our current understanding of Leishmania infection.
Collapse
Affiliation(s)
- Supratim Pradhan
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur, India
| | - Souradeepa Ghosh
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur, India
| | - Shahbaj Hussain
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur, India
| | - Joydeep Paul
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | | |
Collapse
|
14
|
Iwasaki H, Ichihara Y, Morino K, Lemecha M, Sugawara L, Sawano T, Miake J, Sakurai H, Nishi E, Maegawa H, Imamura T. MicroRNA-494-3p inhibits formation of fast oxidative muscle fibres by targeting E1A-binding protein p300 in human-induced pluripotent stem cells. Sci Rep 2021; 11:1161. [PMID: 33441918 PMCID: PMC7806978 DOI: 10.1038/s41598-020-80742-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/17/2020] [Indexed: 01/29/2023] Open
Abstract
MYOD-induced microRNA-494-3p expression inhibits fast oxidative myotube formation by downregulating myosin heavy chain 2 (MYH2) in human induced pluripotent stem cells (hiPSCs) during skeletal myogenesis. However, the molecular mechanisms regulating MYH2 expression via miR-494-3p remain unknown. Here, using bioinformatic analyses, we show that miR-494-3p potentially targets the transcript of the E1A-binding protein p300 at its 3'-untranslated region (UTR). Myogenesis in hiPSCs with the Tet/ON-myogenic differentiation 1 (MYOD1) gene (MyoD-hiPSCs) was induced by culturing them in doxycycline-supplemented differentiation medium for 7 days. p300 protein expression decreased after transient induction of miR-494-3p during myogenesis. miR-494-3p mimics decreased the levels of p300 and its downstream targets MYOD and MYH2 and myotube formation efficiency. p300 knockdown decreased myotube formation efficiency, MYH2 expression, and basal oxygen consumption rate. The binding of miR-494-3p to the wild type p300 3'-UTR, but not the mutated site, was confirmed using luciferase assay. Overexpression of p300 rescued the miR-494-3p mimic-induced phenotype in MyoD-hiPSCs. Moreover, miR-494-3p mimic reduced the levels of p300, MYOD, and MYH2 in skeletal muscles in mice. Thus, miR-494-3p might modulate MYH2 expression and fast oxidative myotube formation by directly regulating p300 levels during skeletal myogenesis in MyoD-hiPSCs and murine skeletal muscle tissues.
Collapse
Affiliation(s)
- Hirotaka Iwasaki
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan
| | - Yoshinori Ichihara
- Division of Pharmacology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Katsutaro Morino
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Tsukinowa, Seta, Otsu, Shiga, 520-2192, Japan.
| | - Mengistu Lemecha
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Tsukinowa, Seta, Otsu, Shiga, 520-2192, Japan
- Department of Molecular and Cellular Biology, City of Hope, Los Angeles, USA
| | - Lucia Sugawara
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Tsukinowa, Seta, Otsu, Shiga, 520-2192, Japan
| | - Tatsuya Sawano
- Division of Pharmacology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Junichiro Miake
- Division of Pharmacology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Eiichiro Nishi
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan
| | - Hiroshi Maegawa
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Tsukinowa, Seta, Otsu, Shiga, 520-2192, Japan
| | - Takeshi Imamura
- Division of Pharmacology, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
15
|
Tiwari R, Banerjee S, Tyde D, Saha KD, Ethirajan A, Mukherjee N, Chattopadhy S, Pramanik SK, Das A. Redox-Responsive Nanocapsules for the Spatiotemporal Release of Miltefosine in Lysosome: Protection against Leishmania. Bioconjug Chem 2021; 32:245-253. [PMID: 33438999 DOI: 10.1021/acs.bioconjchem.0c00667] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Leishmaniasis, a vector-borne disease, is caused by intracellular parasite Leishmania donovani. Unlike most intracellular pathogens, Leishmania donovani are lodged in parasitophorous vacuoles and replicate within the phagolysosomes in macrophages. Effective vaccines against this disease are still under development, while the efficacy of the available drugs is being questioned owing to the toxicity for nonspecific distribution in human physiology and the reported drug-resistance developed by Leishmania donovani. Thus, a stimuli-responsive nanocarrier that allows specific localization and release of the drug in the lysosome has been highly sought after for addressing two crucial issues, lower drug toxicity and a higher drug efficacy. We report here a unique lysosome targeting polymeric nanocapsules, formed via inverse mini-emulsion technique, for stimuli-responsive release of the drug miltefosine in the lysosome of macrophage RAW 264.7 cell line. A benign polymeric backbone, with a disulfide bonding susceptible to an oxidative cleavage, is utilized for the organelle-specific release of miltefosine. Oxidative rupture of the disulfide bond is induced by intracellular glutathione (GSH) as an endogenous stimulus. Such a stimuli-responsive release of the drug miltefosine in the lysosome of macrophage RAW 264.7 cell line over a few hours helped in achieving an improved drug efficacy by 200 times as compared to pure miltefosine. Such a drug formulation could contribute to a new line of treatment for leishmaniasis.
Collapse
Affiliation(s)
- Rajeshwari Tiwari
- CSIR-Central Salt & Marine Chemicals Research Institute, G.B. Marg, Bhavnagar 364002, Gujarat, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, Uttar Pradesh, India
| | - Saswati Banerjee
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Deepak Tyde
- CSIR-Central Salt & Marine Chemicals Research Institute, G.B. Marg, Bhavnagar 364002, Gujarat, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, Uttar Pradesh, India
| | - Krishna Das Saha
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Anitha Ethirajan
- Institute for Materials Research (IMO), Hasselt University, Wetenschapspark 1, 3590 Diepenbeek, Belgium
| | - Niladri Mukherjee
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | | | - Sumit Kumar Pramanik
- CSIR-Central Salt & Marine Chemicals Research Institute, G.B. Marg, Bhavnagar 364002, Gujarat, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, Uttar Pradesh, India
| | - Amitava Das
- Indian Institute of Science Education and Research Kolkata; Mohanpur 741246, West Bengal, India
| |
Collapse
|
16
|
Pei X, Liu J, Liu M, Zhou H, Wang X, Fan H. Quantitative proteomics revealed modulation of macrophages by MetQ gene of Streptococcus suis serotype 2. AMB Express 2020; 10:195. [PMID: 33125582 PMCID: PMC7599288 DOI: 10.1186/s13568-020-01131-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 10/18/2020] [Indexed: 11/10/2022] Open
Abstract
Streptococcus suis serotype 2 (SS2) is a serious zoonotic pathogen; it can lead to symptoms of streptococcal toxic shock syndrome (STSS) in humans and sepsis in pigs, and poses a great threat to public health. The SS2 MetQ gene deletion strain has attenuated antiphagocytosis, although the mechanism of antiphagocytosis and pathogenesis of MetQ in SS2 has remained unclear. In this study, stable isotope labeling by amino acids in cell culture (SILAC) based liquid chromatography–mass spectrometry (LC–MS) and subsequent bioinformatics analysis was used to determine differentially expressed proteins of RAW264.7 cells infected with △MetQ and ZY05719. Proteomic results were verified by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting for selected proteins. Further research was focused mainly on immune system processes related to downregulated proteins, such as Src and Ccl9, and actin cytoskeleton and endocytosis related upregulated proteins, like Pstpip1 and Ppp1r9b. The proteomic results in this study shed light on the mechanism of antiphagocytosis and innate immunity of macrophages infected with △MetQ and ZY05719, which might provide novel targets to prevent or control the infection of SS2.
Collapse
|
17
|
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
|
18
|
Wang L, Li C, Zhang X, Yang M, Wei S, Huang Y, Qin Q, Wang S. The Small GTPase Rab5c Exerts Bi-Function in Singapore Grouper Iridovirus Infections and Cellular Responses in the Grouper, Epinephelus coioides. Front Immunol 2020; 11:2133. [PMID: 33013900 PMCID: PMC7495150 DOI: 10.3389/fimmu.2020.02133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/06/2020] [Indexed: 02/04/2023] Open
Abstract
The small GTPase Rab5 is one of the master regulators of vesicular trafficking that participates in early stages of the endocytic pathway, such as endocytosis and endosome maturation. Three Rab5 isoforms (a, b, and c) share high sequence identity, and exhibit complex functions. However, the role of Rab5c in virus infection and cellular immune responses remains poorly understood. In this study, based on the established virus-cell infection model, Singapore grouper iridovirus (SGIV)-infected grouper spleen (GS) cells, we investigated the role of Rab5c in virus infection and host immune responses. Rab5c was cloned from the orange-spotted grouper, Epinephelus coioides, and termed EcRab5c. EcRab5c encoded a 220-amino-acid polypeptide, showing 99% and 91% identity to Anabas testudineus, and Homo sapiens, respectively. Confocal imaging showed that EcRab5c localized as punctate structures in the cytoplasm. However, a constitutively active (CA) EcRab5c mutant led to enlarged vesicles, while a dominant negative (DN) EcRab5c mutant reduced vesicle structures. EcRab5c expression levels were significantly increased after SGIV infection. EcRab5c knockdown, or CA/DN EcRab5c overexpression significantly inhibited SGIV infection. Using single-particle imaging analysis, we further observed that EcRab5c disruption impaired crucial events at the early stage of SGIV infection, including virus binding, entry, and transport from early to late endosomes, at the single virus level. Furthermore, it is the first time to investigate that EcRab5c is required in autophagy. Equally, EcRab5c positively regulated interferon-related factors and pro-inflammatory cytokines. In summary, these data showed that EcRab5c exerted a bi-functional role on iridovirus infection and host immunity in fish, which furthers our understanding of virus and host immune interactions.
Collapse
Affiliation(s)
- Liqun Wang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Chen Li
- College of Fisheries, Henan Normal University, Xinxiang, China
| | - Xinyue Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Min Yang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Shina Wei
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Youhua Huang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Qiwei Qin
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shaowen Wang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
19
|
Rastogi R, Kapoor A, Verma JK, Ansari I, Sood C, Kumar K, Mukhopadhyay A. Rab5b function is essential to acquire heme from hemoglobin endocytosis for survival of Leishmania. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118868. [PMID: 33011192 DOI: 10.1016/j.bbamcr.2020.118868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 09/02/2020] [Accepted: 09/18/2020] [Indexed: 01/22/2023]
Abstract
Previously, we showed that Rab5a and Rab5b differentially regulate fluid-phase and receptor-mediated endocytosis in Leishmania, respectively. To unequivocally demonstrate the role of Rab5b in hemoglobin endocytosis in Leishmania, we generated null-mutants of Rab5b parasites by sequentially replacing both copies of LdRab5b with the hygromycin and neomycin resistance gene cassettes. LdRab5b-/- null-mutant parasite was confirmed by qPCR analysis of genomic DNA using LdRab5b specific primers. LdRab5b-/- cells showed severe growth defect indicating essential function of LdRab5b in parasite. To characterize the role of Rab5b in Hb endocytosis in parasites, LdRab5b-/- cells were rescued by exogenous addition of hemin in growth medium. Our results showed that LdRab5b-/- cells are relatively smaller in size. Ultrastructural analysis revealed the presence of relatively enlarged flagellar pocket and bigger intracellular vesicles in these cells in comparison to control cells. Both promastigotes and amastigotes of Rab5b null-mutant parasites were unable to internalize Hb but fluid phase endocytosis of different markers was not affected. However, complementation of LdRab5b:WT in LdRab5b-/- cells (LdRab5b-/-:pRab5b:WT) rescued Hb internalization in these cells. Interestingly, LdRab5b-/- cells showed significantly less Hb-receptor on cell surface in comparison to control cells indicating a block in HbR trafficking. Finally, we showed that LdRab5b-/- parasites can infect the macrophages but are unable to survive after 96 h of infection in comparison to control cells. However, supplementation of hemin in the growth medium significantly rescued LdRab5b-/-Leishmania survival in macrophage indicating that LdRab5b function is essential for the acquisition of heme from internalized Hb for the survival of Leishmania.
Collapse
Affiliation(s)
- Ruchir Rastogi
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Anjali Kapoor
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India; National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Jitender Kumar Verma
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
| | - Irshad Ansari
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Chandni Sood
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Kamal Kumar
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
| | - Amitabha Mukhopadhyay
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India.
| |
Collapse
|
20
|
Haldar AK, Nigam U, Yamamoto M, Coers J, Goyal N. Guanylate Binding Proteins Restrict Leishmania donovani Growth in Nonphagocytic Cells Independent of Parasitophorous Vacuolar Targeting. mBio 2020; 11:e01464-20. [PMID: 32723921 PMCID: PMC7387799 DOI: 10.1128/mbio.01464-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 02/05/2023] Open
Abstract
Interferon (IFN)-inducible guanylate binding proteins (GBPs) play important roles in host defense against many intracellular pathogens that reside within pathogen-containing vacuoles (PVs). For instance, members of the GBP family translocate to PVs occupied by the protozoan pathogen Toxoplasma and facilitate PV disruption and lytic parasite killing. While the GBP defense program targeting Toxoplasma has been studied in some detail, the role of GBPs in host defense to other protozoan pathogens is poorly characterized. Here, we report a critical role for both mouse and human GBPs in the cell-autonomous immune response against the vector-borne parasite Leishmania donovani Although L. donovani can infect both phagocytic and nonphagocytic cells, it predominantly replicates inside professional phagocytes. The underlying basis for this cell type tropism is unclear. Here, we demonstrate that GBPs restrict growth of L. donovani in both mouse and human nonphagocytic cells. GBP-mediated restriction of L. donovani replication occurs via a noncanonical pathway that operates independent of detectable translocation of GBPs to L. donovan-containing vacuoles (LCVs). Instead of promoting the lytic destruction of PVs, as reported for GBP-mediated killing of Toxoplasma in phagocytic cells, GBPs facilitate the delivery of L. donovani into autolysosomal-marker-positive compartments in mouse embryonic fibroblasts as well as the human epithelial cell line A549. Together our results show that GBPs control a novel cell-autonomous host defense program, which renders nonphagocytic cells nonpermissible for efficient Leishmania replication.IMPORTANCE The obligate intracellular parasite Leishmania causes the disease leishmaniasis, which is transmitted to mammalian hosts, including humans, via the sandfly vector. Following the bite-induced breach of the skin barrier, Leishmania is known to live and replicate predominantly inside professional phagocytes. Although Leishmania is also able to infect nonphagocytic cells, nonphagocytic cells support limited parasitic replication for unknown reasons. In this study, we show that nonphagocytic cells possess an intrinsic property to restrict Leishmania growth. Our study defines a novel role for a family of host defense proteins, the guanylate binding proteins (GBPs), in antileishmanial immunity. Mechanistically, our data indicate that GBPs facilitate the delivery of Leishmania into antimicrobial autolysosomes, thereby enhancing parasite clearance in nonphagocytic cells. We propose that this GBP-dependent host defense program makes nonphagocytic cells an inhospitable host cell type for Leishmania growth.
Collapse
Affiliation(s)
- Arun Kumar Haldar
- Division of Biochemistry, Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, India
| | - Utsav Nigam
- Division of Biochemistry, Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, India
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Neena Goyal
- Division of Biochemistry, Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, India
| |
Collapse
|
21
|
Batista MF, Nájera CA, Meneghelli I, Bahia D. The Parasitic Intracellular Lifestyle of Trypanosomatids: Parasitophorous Vacuole Development and Survival. Front Cell Dev Biol 2020; 8:396. [PMID: 32587854 PMCID: PMC7297907 DOI: 10.3389/fcell.2020.00396] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/29/2020] [Indexed: 12/21/2022] Open
Abstract
The trypanosomatid (protozoan) parasites Trypanosoma cruzi and Leishmania spp. are causative agents of Chagas disease and Leishmaniasis, respectively. They display high morphological plasticity, are capable of developing in both invertebrate and vertebrate hosts, and are the only trypanosomatids that can survive and multiply inside mammalian host cells. During internalization by host cells, these parasites are lodged in "parasitophorous vacuoles" (PVs) comprised of host cell endolysosomal system components. PVs effectively shelter parasites within the host cell. PV development and maturation (acidification, acquisition of membrane markers, and/or volumetric expansion) precede parasite escape from the vacuole and ultimately from the host cell, which are key determinants of infective burden and persistence. PV biogenesis varies, depending on trypanosomatid species, in terms of morphology (e.g., size), biochemical composition, and parasite-mediated processes that coopt host cell machinery. PVs play essential roles in the intracellular development (i.e., morphological differentiation and/or multiplication) of T. cruzi and Leishmania spp. They are of great research interest as potential gateways for drug delivery systems and other therapeutic strategies for suppression of parasite multiplication and control of the large spectrum of diseases caused by these trypanosomatids. This mini-review focuses on mechanisms of PV biogenesis, and processes whereby PVs of T. cruzi and Leishmania spp. promote parasite persistence within and dissemination among mammalian host cells.
Collapse
Affiliation(s)
| | | | | | - Diana Bahia
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
22
|
Baek KH, Piel L, Rosazza T, Prina E, Späth GF, No JH. Infectivity and Drug Susceptibility Profiling of Different Leishmania-Host Cell Combinations. Pathogens 2020; 9:pathogens9050393. [PMID: 32443883 PMCID: PMC7281264 DOI: 10.3390/pathogens9050393] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/18/2020] [Accepted: 05/18/2020] [Indexed: 11/16/2022] Open
Abstract
Protozoan parasites of the genus Leishmania are the causative agents of leishmaniasis, a spectrum of a disease that threatens public health worldwide. Although next-generation therapeutics are urgently needed, the early stage of the drug discovery process is hampered by very low hit rates from intracellular Leishmania phenotypic high-throughput screenings. Designing and applying a physiologically relevant in vitro assay is therefore in high demand. In this study, we characterized the infectivity, morphology, and drug susceptibility of different Leishmania and host cell infection combinations. Primary bone marrow-derived macrophage (BMDM) and differentiated human acute monocytic leukemia (THP-1) cells were infected with amastigote or promastigote forms of Leishmania amazonensis and Leishmania donovani. Regardless of host cell types, amastigotes were generally well phagocytosed and showed high infectivity, whereas promastigotes, especially those of L. donovani, had predominantly remained in the extracellular space. In the drug susceptibility test, miltefosine and sodium stibogluconate (SSG) showed varying ranges of activity with 14 and >10-fold differences in susceptibility, depending on the host-parasite pairs, indicating the importance of assay conditions for evaluating antileishmanial activity. Overall, our results suggest that combinations of Leishmania species, infection forms, and host cells must be carefully optimized to evaluate the activity of potential therapeutic compounds against Leishmania.
Collapse
Affiliation(s)
- Kyung-Hwa Baek
- Leishmania Research Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do 13488, Korea;
| | - Laura Piel
- Institut Pasteur, Unité de Parasitologie Moléculaire et Signalisation, 75015 Paris, France; (L.P.); (T.R.); (E.P.); (G.F.S.)
| | - Thibault Rosazza
- Institut Pasteur, Unité de Parasitologie Moléculaire et Signalisation, 75015 Paris, France; (L.P.); (T.R.); (E.P.); (G.F.S.)
| | - Eric Prina
- Institut Pasteur, Unité de Parasitologie Moléculaire et Signalisation, 75015 Paris, France; (L.P.); (T.R.); (E.P.); (G.F.S.)
| | - Gerald F. Späth
- Institut Pasteur, Unité de Parasitologie Moléculaire et Signalisation, 75015 Paris, France; (L.P.); (T.R.); (E.P.); (G.F.S.)
| | - Joo Hwan No
- Leishmania Research Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do 13488, Korea;
- Correspondence: ; Tel.: +82-31-8018-8210
| |
Collapse
|
23
|
da Silva Vieira T, Arango Duque G, Ory K, Gontijo CM, Soares RP, Descoteaux A. Leishmania braziliensis: Strain-Specific Modulation of Phagosome Maturation. Front Cell Infect Microbiol 2019; 9:319. [PMID: 31555609 PMCID: PMC6743224 DOI: 10.3389/fcimb.2019.00319] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022] Open
Abstract
Leishmania (Viannia) braziliensis is responsible for the largest number of American tegumentary leishmaniasis (ATL) in Brazil. ATL can present several clinical forms including typical (TL) and atypical (AL) cutaneous and mucocutaneous (ML) lesions. To identify parasite and host factors potentially associated with these diverse clinical manifestations, we first surveyed the expression of two virulence-associated glycoconjugates, lipophosphoglycan (LPG) and the metalloprotease GP63 by a panel of promastigotes of Leishmania braziliensis (L. braziliensis) strains isolated from patients with different clinical manifestations of ATL and from the sand fly vector. We observed a diversity of expression patterns for both LPG and GP63, which may be related to strain-specific polymorphisms. Interestingly, we noted that GP63 activity varies from strain to strain, including the ability to cleave host cell molecules. We next evaluated the ability of promastigotes from these L. braziliensis strains to modulate phagolysosome biogenesis in bone marrow-derived macrophages (BMM), by assessing phagosomal recruitment of the lysosome-associated membrane protein 1 (LAMP-1) and intraphagosomal acidification. Whereas, three out of six L. braziliensis strains impaired the phagosomal recruitment of LAMP-1, only the ML strain inhibited phagosome acidification to the same extent as the L. donovani strain that was used as a positive control. While decreased phagosomal recruitment of LAMP-1 correlated with higher LPG levels, decreased phagosomal acidification correlated with higher GP63 levels. Finally, we observed that the ability to infect and replicate within host cells did not fully correlate with the inhibition of phagosome maturation. Collectively, our results revealed a diversity of strain-specific phenotypes among L. braziliensis isolates, consistent with the high genetic diversity within Leishmania populations.
Collapse
Affiliation(s)
- Tamara da Silva Vieira
- Fundação Oswaldo Cruz - FIOCRUZ, Centro de Pesquisas René Rachou, Belo Horizonte, Brazil.,INRS - Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Guillermo Arango Duque
- INRS - Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Kévin Ory
- INRS - Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada.,Université de Rennes 1, CHU Rennes, INSERM, Rennes, France
| | - Celia Maria Gontijo
- Fundação Oswaldo Cruz - FIOCRUZ, Centro de Pesquisas René Rachou, Belo Horizonte, Brazil
| | - Rodrigo Pedro Soares
- Fundação Oswaldo Cruz - FIOCRUZ, Centro de Pesquisas René Rachou, Belo Horizonte, Brazil
| | - Albert Descoteaux
- INRS - Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| |
Collapse
|
24
|
The host cell secretory pathway mediates the export of Leishmania virulence factors out of the parasitophorous vacuole. PLoS Pathog 2019; 15:e1007982. [PMID: 31356625 PMCID: PMC6687203 DOI: 10.1371/journal.ppat.1007982] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 08/08/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022] Open
Abstract
To colonize phagocytes, Leishmania subverts microbicidal processes through components of its surface coat that include lipophosphoglycan and the GP63 metalloprotease. How these virulence glycoconjugates are shed, exit the parasitophorous vacuole (PV), and traffic within host cells is poorly understood. Here, we show that lipophosphoglycan and GP63 are released from the parasite surface following phagocytosis and redistribute to the endoplasmic reticulum (ER) of macrophages. Pharmacological disruption of the trafficking between the ER and the Golgi hindered the exit of these molecules from the PV and dampened the cleavage of host proteins by GP63. Silencing by RNA interference of the soluble N-ethylmaleimide-sensitive-factor attachment protein receptors Sec22b and syntaxin-5, which regulate ER-Golgi trafficking, identified these host proteins as components of the machinery that mediates the spreading of Leishmania effectors within host cells. Our findings unveil a mechanism whereby a vacuolar pathogen takes advantage of the host cell's secretory pathway to promote egress of virulence factors beyond the PV. Leishmania promastigotes are internalized by phagocytes into a highly modified phagosome that promotes parasite growth and differentiation into the amastigote form. To survive in the phagosome, Leishmania employs surface-bound glycoconjugates such as the GP63 metalloprotease and lipophosphoglycan to subvert the phagosome’s microbicidal potential. In particular, GP63 cleaves host cell vesicle fusion molecules that regulate phagosomal processes ranging from antigen cross-presentation to cytokine secretion. Unlike apicomplexan parasites and bacteria, Leishmania does not inject its virulence-associated glycoconjugates across the phagosome membrane. We found that post-phagocytosis, Leishmania co-opts the host cell secretory pathway to promote the egress of its virulence factors out of the phagosome. Importantly, chemical and genetic inhibition of endoplasmic reticulum (ER) to Golgi transport hindered the redistribution of GP63 and lipophosphoglycan, thereby impeding the cleavage of GP63 target Synaptotagmin XI. Notably, knockdown ER/ERGIC-resident membrane fusion regulators Sec22b and syntaxin-5 revealed that these host molecules were essential to the phagosomal egress of Leishmania virulence factors. These findings provide new insight into how Leishmania sabotages the host cell endomembrane system for its own benefit.
Collapse
|
25
|
Hu ZQ, Rao CL, Tang ML, zhang Y, Lu XX, Chen JG, Mao C, Deng L, Li Q, Mao XH. Rab32 GTPase, as a direct target of miR-30b/c, controls the intracellular survival of Burkholderia pseudomallei by regulating phagosome maturation. PLoS Pathog 2019; 15:e1007879. [PMID: 31199852 PMCID: PMC6594657 DOI: 10.1371/journal.ppat.1007879] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 06/26/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023] Open
Abstract
Burkholderia pseudomallei is a gram-negative, facultative intracellular bacterium, which causes a disease known as melioidosis. Professional phagocytes represent a crucial first line of innate defense against invading pathogens. Uptake of pathogens by these cells involves the formation of a phagosome that matures by fusing with early and late endocytic vesicles, resulting in killing of ingested microbes. Host Rab GTPases are central regulators of vesicular trafficking following pathogen phagocytosis. However, it is unclear how Rab GTPases interact with B. pseudomallei to regulate the transport and maturation of bacterial-containing phagosomes. Here, we showed that the host Rab32 plays an important role in mediating antimicrobial activity by promoting phagosome maturation at an early phase of infection with B. pseudomallei. And we demonstrated that the expression level of Rab32 is increased through the downregulation of the synthesis of miR-30b/30c in B. pseudomallei infected macrophages. Subsequently, we showed that B. pseudomallei resides temporarily in Rab32-positive compartments with late endocytic features. And Rab32 enhances phagosome acidification and promotes the fusion of B. pseudomallei-containing phagosomes with lysosomes to activate cathepsin D, resulting in restricted intracellular growth of B. pseudomallei. Additionally, Rab32 mediates phagosome maturation depending on its guanosine triphosphate/guanosine diphosphate (GTP/GDP) binding state. Finally, we report the previously unrecognized role of miR-30b/30c in regulating B. pseudomallei-containing phagosome maturation by targeting Rab32 in macrophages. Altogether, we provide a novel insight into the host immune-regulated cellular pathway against B. pseudomallei infection is partially dependent on Rab32 trafficking pathway, which regulates phagosome maturation and enhances the killing of this bacterium in macrophages.
Collapse
Affiliation(s)
- Zhi-qiang Hu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Cheng-long Rao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Meng-ling Tang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu zhang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiao-xue Lu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian-gao Chen
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chan Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ling Deng
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xu-hu Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory & Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
26
|
Shadab M, Das S, Banerjee A, Sinha R, Asad M, Kamran M, Maji M, Jha B, Deepthi M, Kumar M, Tripathi A, Kumar B, Chakrabarti S, Ali N. RNA-Seq Revealed Expression of Many Novel Genes Associated With Leishmania donovani Persistence and Clearance in the Host Macrophage. Front Cell Infect Microbiol 2019; 9:17. [PMID: 30805314 PMCID: PMC6370631 DOI: 10.3389/fcimb.2019.00017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 01/17/2019] [Indexed: 12/14/2022] Open
Abstract
Host- as well as parasite-specific factors are equally crucial in allowing either the Leishmania parasites to dominate, or host macrophages to resist infection. To identify such factors, we infected murine peritoneal macrophages with either the virulent (vAG83) or the non-virulent (nvAG83) parasites of L. donovani. Then, through dual RNA-seq, we simultaneously elucidated the transcriptomic changes occurring both in the host and the parasites. Through Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of the differentially expressed (DE) genes, we showed that the vAG83-infected macrophages exhibit biased anti-inflammatory responses compared to the macrophages infected with the nvAG83. Moreover, the vAG83-infected macrophages displayed suppression of many important cellular processes, including protein synthesis. Further, through protein-protein interaction study, we showed significant downregulation in the expression of many hubs and hub-bottleneck genes in macrophages infected with vAG83 as compared to nvAG83. Cell signaling study showed that these two parasites activated the MAPK and PI3K-AKT signaling pathways differentially in the host cells. Through gene ontology analyses of the parasite-specific genes, we discovered that the genes for virulent factors and parasite survival were significantly upregulated in the intracellular amastigotes of vAG83. In contrast, genes involved in the immune stimulations, and those involved in negative regulation of the cell cycle and transcriptional regulation, were upregulated in the nvAG83. Collectively, these results depicted a differential regulation in the host and the parasite-specific molecules during in vitro persistence and clearance of the parasites.
Collapse
Affiliation(s)
- Mohammad Shadab
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Sonali Das
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Anindyajit Banerjee
- Structural Biology and Bio-Informatics Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Roma Sinha
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Mohammad Asad
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Mohd Kamran
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Mithun Maji
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Baijayanti Jha
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Makaraju Deepthi
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | | | | | - Bipin Kumar
- Nucleome Informatics Pvt. Ltd., Hyderabad, India
| | - Saikat Chakrabarti
- Structural Biology and Bio-Informatics Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
27
|
Druggable Targets in Cyclic Nucleotide Signaling Pathways in Apicomplexan Parasites and Kinetoplastids against Disabling Protozoan Diseases in Humans. Int J Mol Sci 2019; 20:ijms20010138. [PMID: 30609697 PMCID: PMC6337498 DOI: 10.3390/ijms20010138] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 12/19/2018] [Accepted: 12/24/2018] [Indexed: 12/20/2022] Open
Abstract
Cell signaling in eukaryotes is an evolutionarily conserved mechanism to respond and adapt to various environmental changes. In general, signal sensation is mediated by a receptor which transfers the signal to a cascade of effector proteins. The cyclic nucleotides 3′,5′-cyclic adenosine monophosphate (cAMP) and 3′,5′-cyclic guanosine monophosphate (cGMP) are intracellular messengers mediating an extracellular stimulus to cyclic nucleotide-dependent kinases driving a change in cell function. In apicomplexan parasites and kinetoplastids, which are responsible for a variety of neglected, tropical diseases, unique mechanisms of cyclic nucleotide signaling are currently identified. Collectively, cyclic nucleotides seem to be essential for parasitic proliferation and differentiation. However, there is no a genomic evidence for canonical G-proteins in these parasites while small GTPases and secondary effector proteins with structural differences to host orthologues occur. Database entries encoding G-protein-coupled receptors (GPCRs) are still without functional proof. Instead, signals from the parasite trigger GPCR-mediated signaling in the host during parasite invasion and egress. The role of cyclic nucleotide signaling in the absence of G-proteins and GPCRs, with a particular focus on small GTPases in pathogenesis, is reviewed here. Due to the absence of G-proteins, apicomplexan parasites and kinetoplastids may use small GTPases or their secondary effector proteins and host canonical G-proteins during infection. Thus, the feasibility of targeting cyclic nucleotide signaling pathways in these parasites, will be an enormous challenge for the identification of selective, pharmacological inhibitors since canonical host proteins also contribute to pathogenesis.
Collapse
|
28
|
Zheng Y. Suppression of mouse miRNA-222-3p in response to Echinococcus multilocularis infection. Int Immunopharmacol 2018; 64:252-255. [PMID: 30212751 DOI: 10.1016/j.intimp.2018.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 11/13/2022]
Abstract
During Echinococcus multilocularis infection, serum miR-222-3p is dramatically downregulated, but its role is yet to be established. Here the expression of miR-222-3p in the spleen of infected mice was shown to be significantly decreased in response to parasite infection (p < 0.05). Using RAW264.7 macrophages, it was further demonstrated that E. multilocularis crude antigens significantly inhibited miR-222-3p expression (p < 0.01). In macrophages transfected with a miR-222-3p inhibitor, NO secretion was moderately decreased compared with the control (p < 0.05). Although all the pro- and anti-inflammatory cytokine genes tested kept constant in expression, four key genes involved in the LPS/TLR4 signaling pathway were significantly down- or up-regulated in transfected cells (p < 0.05), including CD14, TLR4, TICAM2 and AP-1. These results suggest that downregulated miR-222-3p is capable of modulating macrophage immune functions, possibly contributing to the pathogenesis during E. multilocularis infection.
Collapse
Affiliation(s)
- Yadong Zheng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| |
Collapse
|
29
|
Martínez-López M, Soto M, Iborra S, Sancho D. Leishmania Hijacks Myeloid Cells for Immune Escape. Front Microbiol 2018; 9:883. [PMID: 29867798 PMCID: PMC5949370 DOI: 10.3389/fmicb.2018.00883] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/17/2018] [Indexed: 12/23/2022] Open
Abstract
Protozoan parasites of the Leishmania genus are the causative agents of leishmaniasis, a group of neglected tropical diseases whose clinical manifestations vary depending on the infectious Leishmania species but also on host factors. Recognition of the parasite by host myeloid immune cells is a key to trigger an effective Leishmania-specific immunity. However, the parasite is able to persist in host myeloid cells by evading, delaying and manipulating host immunity in order to escape host resistance and ensure its transmission. Neutrophils are first in infiltrating infection sites and could act either favoring or protecting against infection, depending on factors such as the genetic background of the host or the parasite species. Macrophages are the main host cells where the parasites grow and divide. However, macrophages are also the main effector population involved in parasite clearance. Parasite elimination by macrophages requires the priming and development of an effector Th1 adaptive immunity driven by specific subtypes of dendritic cells. Herein, we will provide a comprehensive outline of how myeloid cells regulate innate and adaptive immunity against Leishmania, and the mechanisms used by the parasites to promote their evasion and sabotage. Understanding the interactions between Leishmania and the host myeloid cells may lead to the development of new therapeutic approaches and improved vaccination to leishmaniases, an important worldwide health problem in which current therapeutic or preventive approaches are limited.
Collapse
Affiliation(s)
- María Martínez-López
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain
| | - Manuel Soto
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Salvador Iborra
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain.,Department of Immunology, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain
| |
Collapse
|