1
|
Marques-da-Silva C, Schmidt-Silva C, Bowers C, Charles-Chess NAE, Samuel C, Shiau JC, Park ES, Yuan Z, Kim BH, Kyle DE, Harty JT, MacMicking JD, Kurup SP. Type I interferons induce guanylate-binding proteins and lysosomal defense in hepatocytes to control malaria. Cell Host Microbe 2025; 33:529-544.e9. [PMID: 40168996 DOI: 10.1016/j.chom.2025.03.008] [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: 09/03/2024] [Revised: 01/09/2025] [Accepted: 03/10/2025] [Indexed: 04/03/2025]
Abstract
Plasmodium parasites undergo development and replication within hepatocytes before infecting erythrocytes and initiating clinical malaria. Although type I interferons (IFNs) are known to hinder Plasmodium infection within the liver, the underlying mechanisms remain unclear. Here, we describe two IFN-I-driven hepatocyte antimicrobial programs controlling liver-stage malaria. First, oxidative defense by NADPH oxidases 2 and 4 triggers a pathway of lysosomal fusion with the parasitophorous vacuole (PV) to help clear Plasmodium. Second, guanylate-binding protein (GBP) 1-mediated disruption of the PV activates the caspase-1 inflammasome, inducing pyroptosis to remove infected host cells. Remarkably, both human and mouse hepatocytes enlist these cell-autonomous immune programs to eliminate Plasmodium, with their pharmacologic or genetic inhibition leading to profound malarial susceptibility in vivo. In addition to identifying IFN-I-mediated cell-autonomous immune circuits controlling Plasmodium infection in the hepatocytes, our study also extends the understanding of how non-immune cells are integral to protective immunity against malaria.
Collapse
Affiliation(s)
- Camila Marques-da-Silva
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Clyde Schmidt-Silva
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Carson Bowers
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Nana Appiah Essel Charles-Chess
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Cristina Samuel
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Justine C Shiau
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Eui-Soon Park
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA; Yale Systems Biology Institute, West Haven, CT, USA; Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Zhongyu Yuan
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA; Yale Systems Biology Institute, West Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Bae-Hoon Kim
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA; Yale Systems Biology Institute, West Haven, CT, USA; Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Dennis E Kyle
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - John T Harty
- Department of Pathology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - John D MacMicking
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA; Yale Systems Biology Institute, West Haven, CT, USA; Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Samarchith P Kurup
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA.
| |
Collapse
|
2
|
Boonhok R, Senghoi W, Sangkanu S, Lim CL, Pudla M, Pereira MDL, Wilairatana P, Mahboob T, Rahman MA, Utaisincharoen P, Hiransai P, Nissapatorn V. Acanthamoeba castellanii-Mediated Reduction of Interleukin-1β Secretion and Its Association With Macrophage Autophagy. SCIENTIFICA 2025; 2025:3430892. [PMID: 40109888 PMCID: PMC11922611 DOI: 10.1155/sci5/3430892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Noncanonical autophagy including unconventional protein secretion has been extensively studied. Our work focused on a leaderless IL-1β protein secretion from human macrophage in response to Acanthamoeba castellanii components, Acanthamoeba culture supernatant (CS) and cell lysate (CL), as well as its association with macrophage autophagy. Phorbol 12-myristate 13-acetate (PMA)-induced THP-1 macrophages were treated with Acanthamoeba components of pathogenic (ATCC50739) and nonpathogenic (ATCC30010) strains in vitro. The data showed that Acanthamoeba treatment resulted in low IL-1β secretion from macrophages. In addition, Acanthamoeba CL of both strains was able to upregulate autophagy-related (Atg) protein 8, an autophagy marker, whereas Acanthamoeba CS downregulated Atg8 expression. We further manipulated autophagy and found that autophagy induction by starvation diminished IL-1β secretion while autophagy inhibition by 3-methyladenine (3MA) increased IL-1β secretion. Interestingly, in the presence of Acanthamoeba components either under starvation or 3MA treatment, IL-1β secretion was significantly reduced. Transcriptional expression of other ATG genes, i.e., ATG6, ATG7, and ATG5, were investigated and showed that their mRNA expression was maintained at the basal level under A. castellanii CS or CL treatment. Inflammasome-related genes, NLRP3 and CASPASE1, were upregulated following A. castellanii 50739 CS treatment but not in A. castellanii 50739 CL-treated condition. However, both conditions were able to increase IL-1β mRNA expression. TEM micrographs revealed that 3MA treatment induced the formation of large vacuoles and accumulation of autophagosome at the edge of THP-1 macrophages. However, the number and size of their structures were declined in the presence of A. castellanii 50739 CS with 3MA. Furthermore, immunofluorescence staining demonstrated the association between Atg8/LC3 and IL-1β expression, where downregulation of Atg8 by A. castellanii 50739 CS led to the upregulation of IL-1β. Altogether, the data indicate that Acanthamoeba can manipulate macrophage autophagy, thereby controlling low IL-1β secretion. The expression of autophagy- and inflammasome-related genes also indicates multiple mechanisms in IL-1β secretion in response to Acanthamoeba components. However, further characterization of Atg proteins and investigations into other intracellular pathways or defense mechanisms are needed to fully understand the unconventional secretion of IL-1β in macrophages. This knowledge could eventually lead to the development of innovative therapeutic strategies against Acanthamoeba infection by modulating autophagy or macrophage responses.
Collapse
Affiliation(s)
- Rachasak Boonhok
- Department of Medical Technology, School of Allied Health Sciences, and Research Excellence Center for Innovation and Health Products (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Wilaiwan Senghoi
- Department of Medical Technology, School of Allied Health Sciences, and Center of Excellence Research for Melioidosis and Microorganisms (CERMM), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Suthinee Sangkanu
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla 90112, Thailand
| | - Chooi Ling Lim
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Matsayapan Pudla
- Department of Oral Microbiology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials and Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Tooba Mahboob
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Md Atiar Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Pongsak Utaisincharoen
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Poonsit Hiransai
- Department of Medical Technology, School of Allied Health Sciences, and Center of Excellence in Marijuana, Hemp, and Kratom, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences, Southeast Asia Water Team (SEA Water Team) and World Union for Herbal Drug Discovery (WUHeDD), Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
3
|
Shrikondawar AN, Chennoju K, Ghosh DK, Ranjan A. Mapping of Nuclear Localization Signal in Secreted Liver-Specific Protein 2 of Plasmodium falciparum. ACS Infect Dis 2024; 10:4369-4383. [PMID: 39601512 DOI: 10.1021/acsinfecdis.4c00715] [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] [Indexed: 11/29/2024]
Abstract
The secretory proteome of Plasmodium exhibits differential spatial and functional activity within host cells. Plasmodium secretes proteins that translocate into the human host cell nucleus. Liver-specific protein 2 of Plasmodium falciparum (Pf-LISP2) shows nuclear accumulation in human hepatocytes during the late liver stage of malaria parasite development. However, the nuclear translocation mechanism for Pf-LISP2 remains largely uncharacterized. Here, we identified a classical bipartite nuclear localization signal (NLS) located in the C-terminal region of Pf-LISP2. Phylogenetic analysis revealed that this NLS is unique to Plasmodium falciparum and its close relative Plasmodium reichenowi, suggesting an evolutionary adaptation linked to their shared primate hosts. Functional assays confirmed the NLS's nuclear import activity, as fusion constructs of the Pf-LISP2 NLS with Pf-aldolase (Pf-aldolase-NLS-EGFP) localized exclusively to the nucleus of HepG2 cells. Mutation analysis of key lysine and arginine residues in the bipartite NLS demonstrated that the basic amino acid clusters are essential for nuclear localization. Importin-α/β interaction was found to be crucial for Pf-LISP2 nuclear transport, as coexpression of the NLS constructs with the importin-α/β inhibitor mCherry-Bimax2 significantly blocked nuclear translocation. Specific interactions between the lysine and arginine residues of Pf-LISP2's NLS and the conserved tryptophan and asparagine residues of human importin-α1 facilitate the cytosol-to-nuclear translocation of Pf-LISP2. Additionally, LISP2 lacks any nuclear export signal. These results provide new insights into the mechanisms of nuclear transport in Plasmodium falciparum, potentially contributing to the understanding of its pathogenicity and host-cell interactions during liver-stage infection.
Collapse
Affiliation(s)
- Akshaykumar Nanaji Shrikondawar
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana 500039, India
- Graduate Studies, Regional Centre for Biotechnology, Faridabad, Haryana 121001, India
| | - Kiranmai Chennoju
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana 500039, India
- Graduate Studies, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Debasish Kumar Ghosh
- Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Akash Ranjan
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana 500039, India
| |
Collapse
|
4
|
Nitaramorn N, Kobpornchai P, Tongkrajang N, Chaisri U, Imwong M, Kulkeaw K. Human liver organoids are susceptible to Plasmodium vivax infection. Malar J 2024; 23:368. [PMID: 39639330 PMCID: PMC11622667 DOI: 10.1186/s12936-024-05202-8] [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: 05/15/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The eradication of Plasmodium vivax malaria is complicated due to the presence of hypnozoites, the hidden dormant form of the parasite that is present in the liver. Currently available drug regimens are effective at killing hypnozoites but cause side effects and are difficult to administer. Studies testing drugs for liver-stage malaria remain rare and mainly rely on the use of cancerous or immortalized hepatic cells and primary hepatocytes. METHODS Organoids were used as platform to model liver-stage vivax malaria. Hepatic endoderm cells, endothelial progenitor cells and mesenchymal cells were generated from human induced pluripotent stem cells and self-assembled into liver organoids on top of Matrigel layer. Liver characteristic and maturity were examined through genes and proteins expression of liver markers, and liver functional tests before infected with Plasmodium vivax sporozoites. The infection was then verified by the detection of parasitophorous vacuole membrane proteins, Upregulated in Infectious Sporozoite 4 (UIS4), and blood-stage infection following co-culture with human reticulocytes. RESULTS Generated liver organoids showed upregulation of liver specific transcripts including hepatic nuclear factor 4A (HNF4A), alpha-fetoprotein (AFP), and albumin (ALB) which also confirmed by the protein expression. Furthermore, those organoids resembled mature hepatocytes in terms of albumin secretion, fat and glycogen storage and cytochrome activity. Following invasion of P. vivax sporozoites, PvUIS4 was detected and the hepatic merozoites could develop into ring-stage and early trophozoites in human reticulocytes. Moreover, differential expression patterns of genes involved in lipid and cholesterol synthesis were also detected. CONCLUSIONS Stem cell-derived liver organoids resemble mature liver cells in terms of liver functions and are susceptible to infection with P. vivax sporozoites, paving the way for studies on the mechanism of hypnozoite formation and testing of possible hypnozoitocidal drugs.
Collapse
Affiliation(s)
- Norapat Nitaramorn
- Graduate Program in Biodesign in Medicine, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Porntida Kobpornchai
- Siriraj Integrative Center for Neglected Parasitic Diseases, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Siriraj-Long Read Laboratory, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Nongnat Tongkrajang
- Siriraj Integrative Center for Neglected Parasitic Diseases, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Kasem Kulkeaw
- Siriraj Integrative Center for Neglected Parasitic Diseases, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
- Siriraj-Long Read Laboratory, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
5
|
Silva RCMC, Ribeiro JS, Farias TSDMD, Travassos LH. The role of host autophagy in intracellular protozoan parasites diseases. Arch Biochem Biophys 2024; 761:110186. [PMID: 39455040 DOI: 10.1016/j.abb.2024.110186] [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: 08/28/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Intracellular protozoan parasites are the etiologic agents of important human diseases, like malaria, Chagas disease, toxoplasmosis, and leishmaniasis. Inside host cells, these parasites manipulate the host metabolism and intracellular trafficking for their own benefits and, inevitably, induce several stress response mechanisms. In this review, we discuss autophagy as a stress response mechanism that can be both (i) explored by these intracellular parasites to acquire nutrients and (ii) to restrict parasite proliferation and survival within host cells. We also discuss the immunomodulatory role of autophagy as a strategy to reduce inflammatory-mediated damage, an essential player in the pathophysiology of these parasitic diseases. At last, we propose and discuss several known autophagy modulators as possible pharmaceuticals for adjunctive therapies.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Carlos Chagas Filho Institute of Biophysic, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; State University of Rio de Janeiro, Faculty of Medical Sciences, Campus Cabo Frio, Rio de Janeiro, Brazil
| | - Jhones Sousa Ribeiro
- Laboratory of Immunoreceptors and Signaling, Carlos Chagas Filho Institute of Biophysic, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thalita Santos de Moraes de Farias
- Laboratory of Immunoreceptors and Signaling, Carlos Chagas Filho Institute of Biophysic, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Holanda Travassos
- Laboratory of Immunoreceptors and Signaling, Carlos Chagas Filho Institute of Biophysic, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Marques-da-Silva C, Schmidt-Silva C, Bowers C, Charles-Chess E, Shiau JC, Park ES, Yuan Z, Kim BH, Kyle DE, Harty JT, MacMicking JD, Kurup SP. Type-I IFNs induce GBPs and lysosomal defense in hepatocytes to control malaria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619707. [PMID: 39484443 PMCID: PMC11526971 DOI: 10.1101/2024.10.22.619707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Plasmodium parasites undergo development and replication within the hepatocytes before infecting the erythrocytes and initiating clinical malaria. Although type-I interferons (IFNs) are known to hinder Plasmodium infection within the liver, the underlying mechanisms remain unclear. Here, we describe two IFN-I-driven hepatocyte antimicrobial programs controlling liver-stage malaria. First, oxidative defense by NADPH oxidases 2 and 4 triggers a pathway of lysosomal fusion with the parasitophorous vacuole (PV) to help clear Plasmodium . Second, guanylate-binding protein (GBP) 1 disruption of the PV activates caspase-1 inflammasome, inducing pyroptosis to remove the infected host cells. Remarkably, both human and mouse hepatocytes enlist these cell-autonomous immune programs to eliminate Plasmodium ; their pharmacologic or genetic inhibition led to profound malarial susceptibility, and are essential in vivo . In addition to identifying the IFN-I-mediated cell-autonomous immune circuits controlling Plasmodium infection in the hepatocytes, this study extends our understanding of how non-immune cells are integral to protective immunity against malaria.
Collapse
|
7
|
Marques-da-Silva C, Schmidt-Silva C, Kurup SP. Hepatocytes and the art of killing Plasmodium softly. Trends Parasitol 2024; 40:466-476. [PMID: 38714463 PMCID: PMC11156546 DOI: 10.1016/j.pt.2024.04.004] [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: 02/14/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 05/09/2024]
Abstract
The Plasmodium parasites that cause malaria undergo asymptomatic development in the parenchymal cells of the liver, the hepatocytes, prior to infecting erythrocytes and causing clinical disease. Traditionally, hepatocytes have been perceived as passive bystanders that allow hepatotropic pathogens such as Plasmodium to develop relatively unchallenged. However, now there is emerging evidence suggesting that hepatocytes can mount robust cell-autonomous immune responses that target Plasmodium, limiting its progression to the blood and reducing the incidence and severity of clinical malaria. Here we discuss our current understanding of hepatocyte cell-intrinsic immune responses that target Plasmodium and how these pathways impact malaria.
Collapse
Affiliation(s)
- Camila Marques-da-Silva
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Clyde Schmidt-Silva
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Samarchith P Kurup
- Department of Cellular Biology, University of Georgia, Athens, GA, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA.
| |
Collapse
|
8
|
Bindschedler A, Schmuckli-Maurer J, Buchser S, Fischer TD, Wacker R, Davalan T, Brunner J, Heussler VT. LC3B labeling of the parasitophorous vacuole membrane of Plasmodium berghei liver stage parasites depends on the V-ATPase and ATG16L1. Mol Microbiol 2024; 121:1095-1111. [PMID: 38574236 DOI: 10.1111/mmi.15259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024]
Abstract
The protozoan parasite Plasmodium, the causative agent of malaria, undergoes an obligatory stage of intra-hepatic development before initiating a blood-stage infection. Productive invasion of hepatocytes involves the formation of a parasitophorous vacuole (PV) generated by the invagination of the host cell plasma membrane. Surrounded by the PV membrane (PVM), the parasite undergoes extensive replication. During intracellular development in the hepatocyte, the parasites provoke the Plasmodium-associated autophagy-related (PAAR) response. This is characterized by a long-lasting association of the autophagy marker protein, and ATG8 family member, LC3B with the PVM. LC3B localization at the PVM does not follow the canonical autophagy pathway since upstream events specific to canonical autophagy are dispensable. Here, we describe that LC3B localization at the PVM of Plasmodium parasites requires the V-ATPase and its interaction with ATG16L1. The WD40 domain of ATG16L1 is crucial for its recruitment to the PVM. Thus, we provide new mechanistic insight into the previously described PAAR response targeting Plasmodium liver stage parasites.
Collapse
Affiliation(s)
- Annina Bindschedler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Sophie Buchser
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Tara D Fischer
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Rahel Wacker
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Tim Davalan
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Jessica Brunner
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Volker T Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Ventocilla JA, Tapia LL, Ponce R, Franco A, Leelawong M, Aguiar JC, Baldeviano GC, Wilder BK. Evaluation of naturally acquired immune responses against novel pre-erythrocytic Plasmodium vivax proteins in a low endemic malaria population located in the Peruvian Amazon Basin. Malar J 2024; 23:163. [PMID: 38783317 PMCID: PMC11118720 DOI: 10.1186/s12936-024-04978-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: 12/13/2023] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Plasmodium vivax represents the most geographically widespread human malaria parasite affecting civilian and military populations in endemic areas. Targeting the pre-erythrocytic (PE) stage of the parasite life cycle is especially appealing for developing P. vivax vaccines as it would prevent disease and transmission. Here, naturally acquired immunity to a panel of P. vivax PE antigens was explored, which may facilitate vaccine development and lead to a better understanding of naturally acquired PE immunity. METHODS Twelve P. vivax PE antigens orthologous to a panel of P. falciparum antigens previously identified as highly immunogenic in protected subjects after immunization with radiation attenuated sporozoites (RAS) were used for evaluation of humoral and cellular immunity by ELISA and IFN-γ ELISpot. Samples from P. vivax infected individuals (n = 76) from a low endemic malaria region in the Peruvian Amazon Basin were used. RESULTS In those clinical samples, all PE antigens evaluated showed positive IgG antibody reactivity with a variable prevalence of 58-99% in recently P. vivax diagnosed patients. The magnitude of the IgG antibody response against PE antigens was lower compared with blood stage antigens MSP1 and DBP-II, although antibody levels persisted better for PE antigens (average decrease of 6% for PE antigens and 43% for MSP1, p < 0.05). Higher IgG antibodies was associated with one or more previous malaria episodes only for blood stage antigens (p < 0.001). High IgG responders across PE and blood stage antigens showed significantly lower parasitaemia compared to low IgG responders (median 1,921 vs 4,663 par/µl, p < 0.05). In a subgroup of volunteers (n = 17),positive IFN-γ T cell response by ELISPOT was observed in 35% vs 9-35% against blood stage MSP1 and PE antigens, respectively, but no correlation with IgG responses. CONCLUSIONS These results demonstrate clear humoral and T cell responses against P. vivax PE antigens in individuals naturally infected with P. vivax. These data identify novel attractive PE antigens suitable for use in the potential development and selection of new malaria vaccine candidates which can be used as a part of malaria prevention strategies in civilian and military populations living in P. vivax endemic areas.
Collapse
Affiliation(s)
- Julio A Ventocilla
- Vysnova Partners Inc., Bethesda, USA
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - L Lorena Tapia
- U.S. Naval Medical Research Unit South, Lima-Peru (NAMRU SOUTH), Bellavista, Peru
| | | | | | - Mindy Leelawong
- U.S. Naval Medical Research Unit South, Lima-Peru (NAMRU SOUTH), Bellavista, Peru
- NYC Department of Health and Mental Hygiene, Long Island City, USA
| | | | - G Christian Baldeviano
- U.S. Naval Medical Research Unit South, Lima-Peru (NAMRU SOUTH), Bellavista, Peru
- Bluebird Bio, Inc, Somerville, USA
| | - Brandon K Wilder
- U.S. Naval Medical Research Unit South, Lima-Peru (NAMRU SOUTH), Bellavista, Peru.
- Oregon Health & Science University, Portland, USA.
| |
Collapse
|
10
|
Casanova JL, MacMicking JD, Nathan CF. Interferon- γ and infectious diseases: Lessons and prospects. Science 2024; 384:eadl2016. [PMID: 38635718 DOI: 10.1126/science.adl2016] [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: 10/04/2023] [Accepted: 03/13/2024] [Indexed: 04/20/2024]
Abstract
Infectious diseases continue to claim many lives. Prevention of morbidity and mortality from these diseases would benefit not just from new medicines and vaccines but also from a better understanding of what constitutes protective immunity. Among the major immune signals that mobilize host defense against infection is interferon-γ (IFN-γ), a protein secreted by lymphocytes. Forty years ago, IFN-γ was identified as a macrophage-activating factor, and, in recent years, there has been a resurgent interest in IFN-γ biology and its role in human defense. Here we assess the current understanding of IFN-γ, revisit its designation as an "interferon," and weigh its prospects as a therapeutic against globally pervasive microbial pathogens.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, 75015 Paris, France
| | - John D MacMicking
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, Yale University, West Haven, CT 06477, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Carl F Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
11
|
Aguilera MO, Delgui LR, Reggiori F, Romano PS, Colombo MI. Autophagy as an innate immunity response against pathogens: a Tango dance. FEBS Lett 2024; 598:140-166. [PMID: 38101809 DOI: 10.1002/1873-3468.14788] [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: 08/23/2023] [Revised: 10/18/2023] [Accepted: 10/27/2023] [Indexed: 12/17/2023]
Abstract
Intracellular infections as well as changes in the cell nutritional environment are main events that trigger cellular stress responses. One crucial cell response to stress conditions is autophagy. During the last 30 years, several scenarios involving autophagy induction or inhibition over the course of an intracellular invasion by pathogens have been uncovered. In this review, we will present how this knowledge was gained by studying different microorganisms. We intend to discuss how the cell, via autophagy, tries to repel these attacks with the objective of destroying the intruder, but also how some pathogens have developed strategies to subvert this. These two fates can be compared with a Tango, a dance originated in Buenos Aires, Argentina, in which the partner dancers are in close connection. One of them is the leader, embracing and involving the partner, but the follower may respond escaping from the leader. This joint dance is indeed highly synchronized and controlled, perfectly reflecting the interaction between autophagy and microorganism.
Collapse
Affiliation(s)
- Milton O Aguilera
- Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia-Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Facultad de Odontología, Microbiología, Parasitología e Inmunología, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Laura R Delgui
- Instituto de Histología y Embriología de Mendoza, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro Universitario M5502JMA, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Denmark
- Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Denmark
| | - Patricia S Romano
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora - Instituto de Histología y Embriología de Mendoza, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro Universitario M5502JMA, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
- Facultad de Ciencias Médicas, Centro Universitario M5502JMA, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| | - María I Colombo
- Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia-Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Centro Universitario M5502JMA, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| |
Collapse
|
12
|
Lahree A, Mello-Vieira J, Mota MM. The nutrient games - Plasmodium metabolism during hepatic development. Trends Parasitol 2023; 39:445-460. [PMID: 37061442 DOI: 10.1016/j.pt.2023.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 04/17/2023]
Abstract
Malaria is a febrile illness caused by species of the protozoan parasite Plasmodium and is characterized by recursive infections of erythrocytes, leading to clinical symptoms and pathology. In mammals, Plasmodium parasites undergo a compulsory intrahepatic development stage before infecting erythrocytes. Liver-stage parasites have a metabolic configuration to facilitate the replication of several thousand daughter parasites. Their metabolism is of interest to identify cellular pathways essential for liver infection, to kill the parasite before onset of the disease. In this review, we summarize the current knowledge on nutrient acquisition and biosynthesis by liver-stage parasites mostly generated in murine malaria models, gaps in knowledge, and challenges to create a holistic view of the development and deficiencies in this field.
Collapse
Affiliation(s)
- Aparajita Lahree
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - João Mello-Vieira
- Institute of Biochemistry II, School of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Maria M Mota
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
13
|
Overexpression of hepatocyte EphA2 enhances liver-stage infection by Plasmodium vivax. Sci Rep 2022; 12:21542. [PMID: 36513700 PMCID: PMC9746569 DOI: 10.1038/s41598-022-25281-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
The liver is the first destination of malaria parasites in humans. After reaching the liver by the blood stream, Plasmodium sporozoites cross the liver sinusoid epithelium, enter and exit several hepatocytes, and eventually invade a final hepatocyte host cell. At present, the mechanism of hepatocyte invasion is only partially understood, presenting a key research gap with opportunities for the development of new therapeutics. Recently, human EphA2, a membrane-bound receptor tyrosine kinase, was implicated in hepatocyte infection by the human malaria parasite Plasmodium falciparum and the rodent parasite Plasmodium yoelii, but its role is not known for Plasmodium vivax, a major human parasite whose liver infection poses a specific challenge for malaria treatment and elimination. In this study, the role of EphA2 in P. vivax infection was investigated. It was found that surface expression of several recombinant fragments of EphA2 enhanced the parasite infection rate, thus establishing its role in P. vivax infection. Furthermore, a new permanent cell line (EphA2Extra-HC04) expressing the whole extracellular domain of EphA2 was generated. This cell line supports a higher rate of P. vivax infection and is a valuable tool for P. vivax liver-stage research.
Collapse
|
14
|
Wang Y, Ramos M, Jefferson M, Zhang W, Beraza N, Carding S, Powell PP, Stewart JP, Mayer U, Wileman T. Control of infection by LC3-associated phagocytosis, CASM, and detection of raised vacuolar pH by the V-ATPase-ATG16L1 axis. SCIENCE ADVANCES 2022; 8:eabn3298. [PMID: 36288298 PMCID: PMC9604538 DOI: 10.1126/sciadv.abn3298] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 07/07/2022] [Indexed: 05/29/2023]
Abstract
The delivery of pathogens to lysosomes for degradation provides an important defense against infection. Degradation is enhanced when LC3 is conjugated to endosomes and phagosomes containing pathogens to facilitate fusion with lysosomes. In phagocytic cells, TLR signaling and Rubicon activate LC3-associated phagocytosis (LAP) where stabilization of the NADPH oxidase leads to sustained ROS production and raised vacuolar pH. Raised pH triggers the assembly of the vacuolar ATPase on the vacuole membrane where it binds ATG16L1 to recruit the core LC3 conjugation complex (ATG16L1:ATG5-12). This V-ATPase-ATG16L1 axis is also activated in nonphagocytic cells to conjugate LC3 to endosomes containing extracellular microbes. Pathogens provide additional signals for recruitment of LC3 when they raise vacuolar pH with pore-forming toxins and proteins, phospholipases, or specialized secretion systems. Many microbes secrete virulence factors to inhibit ROS production and/or the V-ATPase-ATG16L1 axis to slow LC3 recruitment and avoid degradation in lysosomes.
Collapse
Affiliation(s)
- Yingxue Wang
- Norwich Medical School, University of East Anglia, Norwich, UK
- Quadram Institute Bioscience, Norwich, UK
| | - Maria Ramos
- Norwich Medical School, University of East Anglia, Norwich, UK
- Quadram Institute Bioscience, Norwich, UK
| | | | - Weijiao Zhang
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | | | | - Penny P. Powell
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - James P. Stewart
- Department of Infection Biology, University of Liverpool, Liverpool, UK
| | - Ulrike Mayer
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia, Norwich, UK
- Quadram Institute Bioscience, Norwich, UK
| |
Collapse
|
15
|
Valenciano AL, Gomez-Lorenzo MG, Vega-Rodríguez J, Adams JH, Roth A. In vitro models for human malaria: targeting the liver stage. Trends Parasitol 2022; 38:758-774. [PMID: 35780012 PMCID: PMC9378454 DOI: 10.1016/j.pt.2022.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
The Plasmodium liver stage represents a vulnerable therapeutic target to prevent disease progression as the parasite resides in the liver before clinical representation caused by intraerythrocytic development. However, most antimalarial drugs target the blood stage of the parasite's life cycle, and the few drugs that target the liver stage are lethal to patients with a glucose-6-phosphate dehydrogenase deficiency. Furthermore, implementation of in vitro liver models to study and develop novel therapeutics against the liver stage of human Plasmodium species remains challenging. In this review, we focus on the progression of in vitro liver models developed for human Plasmodium spp. parasites, provide a brief review on important assay requirements, and lastly present recommendations to improve models to enhance the discovery process of novel preclinical therapeutics.
Collapse
Affiliation(s)
- Ana Lisa Valenciano
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
16
|
Pohl K, Cockburn IA. Innate immunity to malaria: The good, the bad and the unknown. Front Immunol 2022; 13:914598. [PMID: 36059493 PMCID: PMC9437427 DOI: 10.3389/fimmu.2022.914598] [Citation(s) in RCA: 5] [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: 04/07/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
Malaria is the cause of 600.000 deaths annually. However, these deaths represent only a tiny fraction of total malaria cases. Repeated natural infections with the causative agent, Plasmodium sp. parasites, induce protection from severe disease but not sterile immunity. Thus, immunity to Plasmodium is incomplete. Conversely, immunization with attenuated sporozoite stage parasites can induce sterile immunity albeit after multiple vaccinations. These different outcomes are likely to be influenced strongly by the innate immune response to different stages of the parasite lifecycle. Even small numbers of sporozoites can induce a robust proinflammatory type I interferon response, which is believed to be driven by the sensing of parasite RNA. Moreover, induction of innate like gamma-delta cells contributes to the development of adaptive immune responses. Conversely, while blood stage parasites can induce a strong proinflammatory response, regulatory mechanisms are also triggered. In agreement with this, intact parasites are relatively weakly sensed by innate immune cells, but isolated parasite molecules, notably DNA and RNA can induce strong responses. Thus, the innate response to Plasmodium parasite likely represents a trade-off between strong pro-inflammatory responses that may potentiate immunity and regulatory processes that protect the host from cytokine storms that can induce life threatening illness.
Collapse
Affiliation(s)
- Kai Pohl
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University Canberra, Canberra, ACT, Australia
| | - Ian A. Cockburn
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University Canberra, Canberra, ACT, Australia
- *Correspondence: Ian A. Cockburn,
| |
Collapse
|
17
|
Schroeder EA, Chirgwin ME, Derbyshire ER. Plasmodium's fight for survival: escaping elimination while acquiring nutrients. Trends Parasitol 2022; 38:544-557. [PMID: 35534377 PMCID: PMC9187605 DOI: 10.1016/j.pt.2022.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/10/2022] [Accepted: 04/10/2022] [Indexed: 01/08/2023]
Abstract
Plasmodium parasites extensively alter their host hepatocyte to evade host detection and support an unprecedented replication rate. Host cell manipulation includes association with the host early and late endomembrane systems, where Plasmodium accesses nutrients while suppressing cellular immune processes. Early endomembrane organelles provide an opportunity to sequester an abundance of lipids and proteins, but the association with late endomembrane organelles also risks autophagy-mediated elimination. While not all parasites survive, those that do benefit from a plethora of nutrients provided through this pathway. In this review, we discuss recent advances in our understanding of how Plasmodium parasites balance the need for host nutrients while avoiding elimination during the liver stage.
Collapse
Affiliation(s)
- Erin A Schroeder
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | | | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA; Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
18
|
Mancio-Silva L, Gural N, Real E, Wadsworth MH, Butty VL, March S, Nerurkar N, Hughes TK, Roobsoong W, Fleming HE, Whittaker CA, Levine SS, Sattabongkot J, Shalek AK, Bhatia SN. A single-cell liver atlas of Plasmodium vivax infection. Cell Host Microbe 2022; 30:1048-1060.e5. [PMID: 35443155 DOI: 10.1016/j.chom.2022.03.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/31/2022] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
Malaria-causing Plasmodium vivax parasites can linger in the human liver for weeks to years and reactivate to cause recurrent blood-stage infection. Although they are an important target for malaria eradication, little is known about the molecular features of replicative and non-replicative intracellular liver-stage parasites and their host cell dependence. Here, we leverage a bioengineered human microliver platform to culture patient-derived P. vivax parasites for transcriptional profiling. Coupling enrichment strategies with bulk and single-cell analyses, we capture both parasite and host transcripts in individual hepatocytes throughout the course of infection. We define host- and state-dependent transcriptional signatures and identify unappreciated populations of replicative and non-replicative parasites that share features with sexual transmissive forms. We find that infection suppresses the transcription of key hepatocyte function genes and elicits an anti-parasite innate immune response. Our work provides a foundation for understanding host-parasite interactions and reveals insights into the biology of P. vivax dormancy and transmission.
Collapse
Affiliation(s)
- Liliana Mancio-Silva
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France.
| | - Nil Gural
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Eliana Real
- Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France
| | - Marc H Wadsworth
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA
| | - Vincent L Butty
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Niketa Nerurkar
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Travis K Hughes
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok 10400, Thailand
| | - Heather E Fleming
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Charlie A Whittaker
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Stuart S Levine
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok 10400, Thailand
| | - Alex K Shalek
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, MIT, and Harvard, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; The Wyss Institute for Biologically Inspired Engineering Harvard University Boston, MA 02215, USA.
| |
Collapse
|
19
|
Zheng H, Lu X, Li K, Zhu F, Zhao C, Liu T, Ding Y, Fu Y, Zhang K, Zhou T, Dai J, Wu Y, Xu W. ATG Ubiquitination Is Required for Circumsporozoite Protein to Subvert Host Innate Immunity Against Rodent Malaria Liver Stage. Front Immunol 2022; 13:815936. [PMID: 35222391 PMCID: PMC8864237 DOI: 10.3389/fimmu.2022.815936] [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: 11/16/2021] [Accepted: 01/20/2022] [Indexed: 12/02/2022] Open
Abstract
Although exo-erythrocytic forms (EEFs) of liver stage malaria parasite in the parasitophorous vacuole (PV) are encountered with robust host innate immunity, EEFs can still survive and successfully complete the infection of hepatocytes, and the underlying mechanism is largely unknown. Here, we showed that sporozoite circumsporozoite protein (CSP) translocated from the parasitophorous vacuole into the hepatocyte cytoplasm significantly mediated the resistance to the killing of EEFs by interferon-gamma (IFN-γ). Attenuation of IFN-γ-mediated killing of EEFs by CSP was dependent on its ability to reduce the levels of autophagy-related genes (ATGs) in hepatocytes. The ATGs downregulation occurred through its enhanced ubiquitination mediated by E3 ligase NEDD4, an enzyme that was upregulated by CSP when it translocated from the cytoplasm into the nucleus of hepatocytes via its nuclear localization signal (NLS) domain. Thus, we have revealed an unrecognized role of CSP in subverting host innate immunity and shed new light for a prophylaxis strategy against liver-stage infection.
Collapse
Affiliation(s)
- Hong Zheng
- Department of Pathogenic Biology, Army Medical University, Chongqing, China.,The Institute of Immunology, Army Medical University, Chongqing, China.,Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiao Lu
- Department of Pathogenic Biology, Army Medical University, Chongqing, China.,Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Kai Li
- Department of Pathogenic Biology, Army Medical University, Chongqing, China
| | - Feng Zhu
- Department of Pathogenic Biology, Army Medical University, Chongqing, China
| | - Chenhao Zhao
- Department of Pathogenic Biology, Army Medical University, Chongqing, China
| | - Taiping Liu
- Department of Pathogenic Biology, Army Medical University, Chongqing, China
| | - Yan Ding
- Department of Pathogenic Biology, Army Medical University, Chongqing, China
| | - Yong Fu
- Department of Pathogenic Biology, Army Medical University, Chongqing, China
| | - Kun Zhang
- Department of Pathogenic Biology, Army Medical University, Chongqing, China
| | - Taoli Zhou
- Department of Pathogenic Biology, Army Medical University, Chongqing, China
| | - Jigang Dai
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuzhang Wu
- The Institute of Immunology, Army Medical University, Chongqing, China
| | - Wenyue Xu
- Department of Pathogenic Biology, Army Medical University, Chongqing, China.,Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
| |
Collapse
|
20
|
Leleu I, Alloo J, Cazenave PA, Roland J, Pied S. Autophagy Pathways in the Genesis of Plasmodium-Derived Microvesicles: A Double-Edged Sword? Life (Basel) 2022; 12:life12030415. [PMID: 35330166 PMCID: PMC8955828 DOI: 10.3390/life12030415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022] Open
Abstract
Malaria, caused by Plasmodium species (spp.), is a deadly parasitic disease that results in approximately 400,000 deaths per year globally. Autophagy pathways play a fundamental role in the developmental stages of the parasite within the mammalian host. They are also involved in the production of Plasmodium-derived extracellular vesicles (EVs), which play an important role in the infection process, either by providing nutrients for parasite growth or by contributing to the immunopathophysiology of the disease. For example, during the hepatic stage, Plasmodium-derived EVs contribute to parasite virulence by modulating the host immune response. EVs help in evading the different autophagy mechanisms deployed by the host for parasite clearance. During cerebral malaria, on the other hand, parasite-derived EVs promote an astrocyte-mediated inflammatory response, through the induction of a non-conventional host autophagy pathway. In this review, we will discuss the cross-talk between Plasmodium-derived microvesicles and autophagy, and how it influences the outcome of infection.
Collapse
|
21
|
Baindara P, Agrawal S, Franco OL. Host-directed therapies for malaria and tuberculosis: common infection strategies and repurposed drugs. Expert Rev Anti Infect Ther 2022; 20:849-869. [DOI: 10.1080/14787210.2022.2044794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Piyush Baindara
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Missouri, Columbia, MO, USA
| | - Sonali Agrawal
- Immunology Division, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, Uttar Pradesh, India
| | - O. L. Franco
- Proteomics Analysis and Biochemical Center, Catholic University of Brasilia, Brasilia, Brazil; S-Inova Biotech, Catholic University Dom Bosco, Campo Grande, MS, Brazil
| |
Collapse
|
22
|
Glennon EKK, Tongogara T, Primavera VI, Reeder SM, Wei L, Kaushansky A. Elucidating Spatially-Resolved Changes in Host Signaling During Plasmodium Liver-Stage Infection. Front Cell Infect Microbiol 2022; 11:804186. [PMID: 35111697 PMCID: PMC8801743 DOI: 10.3389/fcimb.2021.804186] [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: 10/29/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022] Open
Abstract
Upon transmission to the human host, Plasmodium sporozoites exit the skin, are taken up by the blood stream, and then travel to the liver where they infect and significantly modify a single hepatocyte. Low infection rates within the liver have made proteomic studies of infected hepatocytes challenging, particularly in vivo, and existing studies have been largely unable to consider how protein and phosphoprotein differences are altered at different spatial locations within the heterogeneous liver. Using digital spatial profiling, we characterized changes in host signaling during Plasmodium yoelii infection in vivo without disrupting the liver tissue. Moreover, we measured alterations in protein expression around infected hepatocytes and identified a subset of CD163+ Kupffer cells that migrate towards infected cells during infection. These data offer the first insight into the heterogeneous microenvironment that surrounds the infected hepatocyte and provide insights into how the parasite may alter its milieu to influence its survival and modulate immunity.
Collapse
Affiliation(s)
- Elizabeth K. K. Glennon
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Tinotenda Tongogara
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Grinnell College, Grinnell, IA, United States
| | - Veronica I. Primavera
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Sophia M. Reeder
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Ling Wei
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Alexis Kaushansky
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Brotman Baty Institute for Precision Medicine, Seattle, WA, United States
- *Correspondence: Alexis Kaushansky,
| |
Collapse
|
23
|
Vijayan K, Wei L, Glennon EKK, Mattocks C, Bourgeois N, Staker B, Kaushansky A. Host-targeted Interventions as an Exciting Opportunity to Combat Malaria. Chem Rev 2021; 121:10452-10468. [PMID: 34197083 DOI: 10.1021/acs.chemrev.1c00062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Terminal and benign diseases alike in adults, children, pregnant women, and others are successfully treated by pharmacological inhibitors that target human enzymes. Despite extensive global efforts to fight malaria, the disease continues to be a massive worldwide health burden, and new interventional strategies are needed. Current drugs and vector control strategies have contributed to the reduction in malaria deaths over the past 10 years, but progress toward eradication has waned in recent years. Resistance to antimalarial drugs is a substantial and growing problem. Moreover, targeting dormant forms of the malaria parasite Plasmodium vivax is only possible with two approved drugs, which are both contraindicated for individuals with glucose-6-phosphate dehydrogenase deficiency and in pregnant women. Plasmodium parasites are obligate intracellular parasites and thus have specific and absolute requirements of their hosts. Growing evidence has described these host necessities, paving the way for opportunities to pharmacologically target host factors to eliminate Plasmodium infection. Here, we describe progress in malaria research and adjacent fields and discuss key challenges that remain in implementing host-directed therapy against malaria.
Collapse
Affiliation(s)
| | - Ling Wei
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | | | - Christa Mattocks
- Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Natasha Bourgeois
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Bart Staker
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | - Alexis Kaushansky
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States.,Department of Pediatrics, University of Washington, Seattle, Washington 98105, United States.,Brotman Baty Institute for Precision Medicine, Seattle, Washington 98195, United States
| |
Collapse
|
24
|
De Niz M, Caldelari R, Kaiser G, Zuber B, Heo WD, Heussler VT, Agop-Nersesian C. Hijacking of the host cell Golgi by Plasmodium berghei liver stage parasites. J Cell Sci 2021; 134:jcs252213. [PMID: 34013963 PMCID: PMC8186485 DOI: 10.1242/jcs.252213] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 04/12/2021] [Indexed: 12/28/2022] Open
Abstract
The intracellular lifestyle represents a challenge for the rapidly proliferating liver stage Plasmodium parasite. In order to scavenge host resources, Plasmodium has evolved the ability to target and manipulate host cell organelles. Using dynamic fluorescence-based imaging, we here show an interplay between the pre-erythrocytic stages of Plasmodium berghei and the host cell Golgi during liver stage development. Liver stage schizonts fragment the host cell Golgi into miniaturized stacks, which increases surface interactions with the parasitophorous vacuolar membrane of the parasite. Expression of specific dominant-negative Arf1 and Rab GTPases, which interfere with the host cell Golgi-linked vesicular machinery, results in developmental delay and diminished survival of liver stage parasites. Moreover, functional Rab11a is critical for the ability of the parasites to induce Golgi fragmentation. Altogether, we demonstrate that the structural integrity of the host cell Golgi and Golgi-associated vesicular traffic is important for optimal pre-erythrocytic development of P. berghei. The parasite hijacks the Golgi structure of the hepatocyte to optimize its own intracellular development. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | - Gesine Kaiser
- Institute of Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | - Benoit Zuber
- Institute for Anatomy, University of Bern, CH-3012 Bern, Switzerland
| | - Won Do Heo
- Dept. of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Volker T. Heussler
- Institute of Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | | |
Collapse
|
25
|
Sena-dos-Santos C, Braga-da-Silva C, Marques D, Azevedo dos Santos Pinheiro J, Ribeiro-dos-Santos Â, Cavalcante GC. Unraveling Cell Death Pathways during Malaria Infection: What Do We Know So Far? Cells 2021; 10:479. [PMID: 33672278 PMCID: PMC7926694 DOI: 10.3390/cells10020479] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Malaria is a parasitic disease (caused by different Plasmodium species) that affects millions of people worldwide. The lack of effective malaria drugs and a vaccine contributes to this disease, continuing to cause major public health and socioeconomic problems, especially in low-income countries. Cell death is implicated in malaria immune responses by eliminating infected cells, but it can also provoke an intense inflammatory response and lead to severe malaria outcomes. The study of the pathophysiological role of cell death in malaria in mammalians is key to understanding the parasite-host interactions and design prophylactic and therapeutic strategies for malaria. In this work, we review malaria-triggered cell death pathways (apoptosis, autophagy, necrosis, pyroptosis, NETosis, and ferroptosis) and we discuss their potential role in the development of new approaches for human malaria therapies.
Collapse
Affiliation(s)
- Camille Sena-dos-Santos
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Cíntia Braga-da-Silva
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Diego Marques
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Jhully Azevedo dos Santos Pinheiro
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Ândrea Ribeiro-dos-Santos
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
- Programa de Pós-Graduação em Oncologia e Ciências Médicas, Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66.075-110, Brazil
| | - Giovanna C. Cavalcante
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| |
Collapse
|
26
|
Zhao W, Gao B, Liu C, Zhang B, Shan C, Deng J, Wan Q, Wang X, Zhao R, Gao L, Ao P, Xiao P, Gao H. High pathogenicity island is associated with enhanced autophagy in pathogenic Escherichia coli HPI - infected macrophages. Res Vet Sci 2021; 135:113-120. [PMID: 33465603 DOI: 10.1016/j.rvsc.2021.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 10/22/2022]
Abstract
High pathogenicity island (HPI), which is widely distributed in Escherichia coli (E. coli), can enhance the pathogenicity of E. coli. Thus the HPI positive E. coli could pose a threat to human and animal health. It remains to be elucidated how HPI affects the virulence of pathogenic E. coli. Autophagy is an important mechanism to maintain cellular homeostasis and an innate immunity responses of organisms against pathogens. The interaction between pathogenic E. coli possessing HPI (E. coli HPI) and host autophagy system has not been reported. In this study, it was demonstrated that pathogenic E. coli induced autophagy in 3D4/21 macrophages and HPI was associated with enhanced autophagy through transmission electron microscopy, immunofluorescence and real-time PCR. The PI3K/Akt/mTOR pathway is an important negative regulatory pathway for autophagy. Through detecting the expression of key genes of PI3K/Akt/mTOR pathway, it was speculated that HPI enhanced the inhibition of the signaling pathway stimulated by pathogenic E. coli. Furthermore, HPI inhibited the secretion of IFN-γ, while the presence of HPI did not significantly affect the secretion of IL-1β. This work is the first attempt to explore the interplay between HPI carried by pathogenic E. coli and host cell autophagy. The findings might enable better understanding of the contribution of HPI to pathogenicity.
Collapse
Affiliation(s)
- Weiwei Zhao
- College of food science and technology, Yunnan Agricultural University, Kunming 650201, China
| | - Bin Gao
- College of food science and technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chang Liu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Bo Zhang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chunlan Shan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Jing Deng
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Quan Wan
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Xi Wang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Ru Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Libo Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Pingxing Ao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China.
| | - Hong Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
27
|
Disrupting Plasmodium UIS3-host LC3 interaction with a small molecule causes parasite elimination from host cells. Commun Biol 2020; 3:688. [PMID: 33214643 PMCID: PMC7677311 DOI: 10.1038/s42003-020-01422-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 10/17/2020] [Indexed: 01/12/2023] Open
Abstract
The malaria parasite Plasmodium obligatorily infects and replicates inside hepatocytes surrounded by a parasitophorous vacuole membrane (PVM), which is decorated by the host-cell derived autophagy protein LC3. We have previously shown that the parasite-derived, PVM-resident protein UIS3 sequesters LC3 to avoid parasite elimination by autophagy from hepatocytes. Here we show that a small molecule capable of disrupting this interaction triggers parasite elimination in a host cell autophagy-dependent manner. Molecular docking analysis of more than 20 million compounds combined with a phenotypic screen identified one molecule, C4 (4-{[4-(4-{5-[3-(trifluoromethyl) phenyl]-1,2,4-oxadiazol-3-yl}benzyl)piperazino]carbonyl}benzonitrile), capable of impairing infection. Using biophysical assays, we established that this impairment is due to the ability of C4 to disrupt UIS3-LC3 interaction, thus inhibiting the parasite's ability to evade the host autophagy response. C4 impacts infection in autophagy-sufficient cells without harming the normal autophagy pathway of the host cell. This study, by revealing the disruption of a critical host-parasite interaction without affecting the host's normal function, uncovers an efficient anti-malarial strategy to prevent this deadly disease.
Collapse
|
28
|
Bindschedler A, Wacker R, Egli J, Eickel N, Schmuckli-Maurer J, Franke-Fayard BM, Janse CJ, Heussler VT. Plasmodium berghei sporozoites in nonreplicative vacuole are eliminated by a PI3P-mediated autophagy-independent pathway. Cell Microbiol 2020; 23:e13271. [PMID: 32979009 PMCID: PMC7757174 DOI: 10.1111/cmi.13271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/01/2022]
Abstract
The protozoan parasite Plasmodium, causative agent of malaria, invades hepatocytes by invaginating the host cell plasma membrane and forming a parasitophorous vacuole membrane (PVM). Surrounded by this PVM, the parasite undergoes extensive replication. Parasites inside a PVM provoke the Plasmodium‐associated autophagy‐related (PAAR) response. This is characterised by a long‐lasting association of the autophagy marker protein LC3 with the PVM, which is not preceded by phosphatidylinositol 3‐phosphate (PI3P)‐labelling. Prior to productive invasion, sporozoites transmigrate several cells and here we describe that a proportion of traversing sporozoites become trapped in a transient traversal vacuole, provoking a host cell response that clearly differs from the PAAR response. These trapped sporozoites provoke PI3P‐labelling of the surrounding vacuolar membrane immediately after cell entry, followed by transient LC3‐labelling and elimination of the parasite by lysosomal acidification. Our data suggest that this PI3P response is not only restricted to sporozoites trapped during transmigration but also affects invaded parasites residing in a compromised vacuole. Thus, host cells can employ a pathway distinct from the previously described PAAR response to efficiently recognise and eliminate Plasmodium parasites.
Collapse
Affiliation(s)
- Annina Bindschedler
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Rahel Wacker
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Jessica Egli
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Nina Eickel
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Blandine M Franke-Fayard
- Leiden malaria group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris J Janse
- Leiden malaria group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
29
|
Ghartey-Kwansah G, Adu-Nti F, Aboagye B, Ankobil A, Essuman EE, Opoku YK, Abokyi S, Abu EK, Boampong JN. Autophagy in the control and pathogenesis of parasitic infections. Cell Biosci 2020; 10:101. [PMID: 32944216 PMCID: PMC7487832 DOI: 10.1186/s13578-020-00464-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Background Autophagy has a crucial role in the defense against parasites. The interplay existing between host autophagy and parasites has varied outcomes due to the kind of host cell and microorganism. The presence of autophagic compartments disrupt a significant number of pathogens and are further cleared by xenophagy in an autolysosome. Another section of pathogens have the capacity to outwit the autophagic pathway to their own advantage. Result To comprehend the interaction between pathogens and the host cells, it is significant to distinguish between starvation-induced autophagy and other autophagic pathways. Subversion of host autophagy by parasites is likely due to differences in cellular pathways from those of ‘classical’ autophagy and that they are controlled by parasites in a peculiar way. In xenophagy clearance at the intracellular level, the pathogens are first ubiquitinated before autophagy receptors acknowledgement, followed by labeling with light chain 3 (LC3) protein. The LC3 in LC3-associated phagocytosis (LAP) is added directly into vacuole membrane and functions regardless of the ULK, an initiation complex. The activation of the ULK complex composed of ATG13, FIP200 and ATG101causes the initiation of host autophagic response. Again, the recognition of PAMPs by conserved PRRs marks the first line of defense against pathogens, involving Toll-like receptors (TLRs). These all important immune-related receptors have been reported recently to regulate autophagy. Conclusion In this review, we sum up recent advances in autophagy to acknowledge and understand the interplay between host and parasites, focusing on target proteins for the design of therapeutic drugs. The target host proteins on the initiation of the ULK complex and PRRs-mediated recognition of PAMPs may provide strong potential for the design of therapeutic drugs against parasitic infections.
Collapse
Affiliation(s)
- George Ghartey-Kwansah
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Frank Adu-Nti
- Department of Medical Laboratory Science, Radford University College, Accra, Ghana
| | - Benjamin Aboagye
- Department of Forensic Sciences, College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Amandus Ankobil
- School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana.,Department of Epidemiology and Biostatistics, State University of New York at Albany, New York, USA
| | - Edward Eyipe Essuman
- US Food and Drugs Administration CBER, OBRR, DETTD 10903 New Hampshire Avenue, White Oak, USA
| | - Yeboah Kwaku Opoku
- Department of Biology Education, Faculty of Science, University of Education, Winneba, Ghana
| | - Samuel Abokyi
- Department of Optometry and Vision Science, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana.,School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Emmanuel Kwasi Abu
- Department of Optometry and Vision Science, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Johnson Nyarko Boampong
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
30
|
Coban C. The host targeting effect of chloroquine in malaria. Curr Opin Immunol 2020; 66:98-107. [PMID: 32823144 PMCID: PMC7431399 DOI: 10.1016/j.coi.2020.07.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 01/07/2023]
Abstract
Due to the rapid onset and spread of the COVID-19 pandemic, the treatment of COVID-19 patients by hydroxychloroquine alone or in combination with other drugs has captured a great deal of attention and triggered considerable debate. Historically, the worldwide use of quinoline based-drugs has led to a spectacular reduction in death from malaria. Unfortunately, scientists have been forced to seek alternative drugs to treat malaria due to the emergence of chloroquine-resistant parasites in the 1960s. The repurposing of hydroxychloroquine against viral infections, various types of cancer and autoimmune diseases has been ongoing for more than 70 years, with no clear understanding of its mechanism of action (MOA). Here, we closely examine the MOA of this old but influential drug in and beyond malaria. Better insights into how chloroquine targets the host's cellular and immune responses may help to develop applications against to new pathogens and diseases, and perhaps even restore the clinical utility of chloroquine against malaria.
Collapse
Affiliation(s)
- Cevayir Coban
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo, Japan; Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan.
| |
Collapse
|
31
|
Subramani PA, Vartak-Sharma N, Sreekumar S, Mathur P, Nayer B, Dakhore S, Basavanna SK, Kalappa DM, Krishnamurthy RV, Mukhi B, Mishra P, Yoshida N, Ghosh SK, Shandil R, Narayanan S, Campo B, Hasegawa K, Anvikar AR, Valecha N, Sundaramurthy V. Plasmodium vivax liver stage assay platforms using Indian clinical isolates. Malar J 2020; 19:214. [PMID: 32571333 PMCID: PMC7310233 DOI: 10.1186/s12936-020-03284-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/12/2020] [Indexed: 12/24/2022] Open
Abstract
Background Vivax malaria is associated with significant morbidity and economic loss, and constitutes the bulk of malaria cases in large parts of Asia and South America as well as recent case reports in Africa. The widespread prevalence of vivax is a challenge to global malaria elimination programmes. Vivax malaria control is particularly challenged by existence of dormant liver stage forms that are difficult to treat and are responsible for multiple relapses, growing drug resistance to the asexual blood stages and host-genetic factors that preclude use of specific drugs like primaquine capable of targeting Plasmodium vivax liver stages. Despite an obligatory liver-stage in the Plasmodium life cycle, both the difficulty in obtaining P. vivax sporozoites and the limited availability of robust host cell models permissive to P. vivax infection are responsible for the limited knowledge of hypnozoite formation biology and relapse mechanisms, as well as the limited capability to do drug screening. Although India accounts for about half of vivax malaria cases world-wide, very little is known about the vivax liver stage forms in the context of Indian clinical isolates. Methods To address this, methods were established to obtain infective P. vivax sporozoites from an endemic region in India and multiple assay platforms set up to detect and characterize vivax liver stage forms. Different hepatoma cell lines, including the widely used HCO4 cells, primary human hepatocytes as well as hepatocytes obtained from iPSC’s generated from vivax patients and healthy donors were tested for infectivity with P. vivax sporozoites. Results Both large and small forms of vivax liver stage are detected in these assays, although the infectivity obtained in these platforms are low. Conclusions This study provides a proof of concept for detecting liver stage P. vivax and provide the first characterization of P. vivax liver stage forms from an endemic region in India.
Collapse
Affiliation(s)
- Pradeep A Subramani
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bangalore, 560065, India.,ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India
| | - Neha Vartak-Sharma
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India.,Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advance Studies, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Seetha Sreekumar
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bangalore, 560065, India
| | - Pallavi Mathur
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bangalore, 560065, India
| | - Bhavana Nayer
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India
| | - Sushrut Dakhore
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India
| | - Sowmya K Basavanna
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India
| | - Devaiah M Kalappa
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India
| | | | - Benudhar Mukhi
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India
| | - Priyasha Mishra
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India
| | - Noriko Yoshida
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advance Studies, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Susanta Kumar Ghosh
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India. .,Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | | | | | - Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Kouichi Hasegawa
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India. .,Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advance Studies, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Anupkumar R Anvikar
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, New Delhi, India
| | - Neena Valecha
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, New Delhi, India
| | - Varadharajan Sundaramurthy
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bangalore, 560065, India.
| |
Collapse
|
32
|
Dutour L, Mandonnet É, Nüsse O. [The role of LC3-associated phagocytosis during infection by Listeria monocytogenes]. Med Sci (Paris) 2020; 36:531-533. [PMID: 32452379 DOI: 10.1051/medsci/2020089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Léa Dutour
- M1 Biologie Santé, Université Paris-Saclay, 91405 Orsay, France
| | | | - Oliver Nüsse
- Université Paris-Saclay, CNRS, Institut de chimie physique, UMR8000, 91405 Orsay, France
| |
Collapse
|
33
|
LC3-associated phagocytosis - The highway to hell for phagocytosed microbes. Semin Cell Dev Biol 2020; 101:68-76. [DOI: 10.1016/j.semcdb.2019.04.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/13/2022]
|
34
|
Posfai D, Maher SP, Roesch C, Vantaux A, Sylvester K, Péneau J, Popovici J, Kyle DE, Witkowski B, Derbyshire ER. Plasmodium vivax Liver and Blood Stages Recruit the Druggable Host Membrane Channel Aquaporin-3. Cell Chem Biol 2020; 27:719-727.e5. [PMID: 32330444 PMCID: PMC7303948 DOI: 10.1016/j.chembiol.2020.03.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/07/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
Abstract
Plasmodium vivax infects hepatocytes to form schizonts that cause blood infection, or dormant hypnozoites that can persist for months in the liver before leading to relapsing blood infections. The molecular processes that drive P. vivax schizont and hypnozoite survival remain largely unknown, but they likely involve a rich network of host-pathogen interactions, including those occurring at the host-parasite interface, the parasitophorous vacuole membrane (PVM). Using a recently developed P. vivax liver-stage model system we demonstrate that host aquaporin-3 (AQP3) localizes to the PVM of schizonts and hypnozoites within 5 days after invasion. This recruitment is also observed in P. vivax-infected reticulocytes. Chemical treatment with the AQP3 inhibitor auphen reduces P. vivax liver hypnozoite and schizont burden, and inhibits P. vivax asexual blood-stage growth. These findings reveal a role for AQP3 in P. vivax liver and blood stages and suggest that the protein may be targeted for therapeutic treatment. Host aquaporin-3 (AQP3) is recruited to P. vivax hypnozoites and schizonts The AQP3 inhibitor auphen inhibits P. vivax hypnozoites and schizonts Host AQP3 is recruited to P. vivax-infected erythrocytes derived from patient samples Auphen inhibits blood stages of clinical P. vivax isolates
Collapse
Affiliation(s)
- Dora Posfai
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA
| | - Steven P Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 D.W. Brooks Dr, ste 370, Athens, GE 30602, USA
| | - Camille Roesch
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Amélie Vantaux
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Kayla Sylvester
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA
| | - Julie Péneau
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Jean Popovici
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 D.W. Brooks Dr, ste 370, Athens, GE 30602, USA
| | - Benoît Witkowski
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia.
| | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA; Chemistry Department, Duke University, 124 Science Drive, Durham, NC 27708, USA.
| |
Collapse
|
35
|
Ghartey-Kwansah G, Aboagye B, Adu-Nti F, Opoku YK, Abu EK. Clearing or subverting the enemy: Role of autophagy in protozoan infections. Life Sci 2020; 247:117453. [PMID: 32088215 DOI: 10.1016/j.lfs.2020.117453] [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: 01/02/2020] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/20/2022]
Abstract
The protozoan parasites are evolutionarily divergent, unicellular eukaryotic pathogens representing one of the essential sources of parasitic diseases. These parasites significantly affect the economy and cause public health burdens globally. Protozoan parasites share many cellular features and pathways with their respective host cells. This includes autophagy, a process responsible for self-degradation of the cell's components. There is conservation of the central structural and functional machinery for autophagy in most of the eukaryotic phyla, however, Plasmodium and Toxoplasma possess a decreased number of recognizable autophagy-related proteins (ATG). Plasmodium noticeably lacks clear orthologs of the initiating kinase ATG1/ULK1/2, and both Plasmodium and Toxoplasma lack proteins involved in the nucleation of autophagosomes. These organisms have essential apicoplast, a plastid-like non-photosynthetic organelle, which is an adaptation that is used in penetrating the host cell. Furthermore, available evidence suggests that Leishmania, an intracellular protozoan parasite, induces autophagy in macrophages. The autophagic pathway in Trypanosoma cruzi is activated during metacyclogenesis, a process responsible for the infective forms of parasites. Therefore, numerous pathogens have developed strategies to impair the autophagic mechanism in phagocytes. Regulating autophagy is essential to maintain cellular health as adjustments in the autophagy pathway have been linked to the progression of several physiological and pathological conditions in humans. In this review, we report current advances in autophagy in parasites and their host cells, focusing on the ramifications of these studies in the design of potential anti-protozoan therapeutics.
Collapse
Affiliation(s)
- George Ghartey-Kwansah
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana; College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Benjamin Aboagye
- Department of Forensic Sciences, College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Frank Adu-Nti
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yeboah Kwaku Opoku
- Department of Biology Education, Faculty of Science, University of Education, Winneba, Ghana
| | - Emmanuel Kwasi Abu
- Department of Optometry and Vision Science, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
36
|
van Niekerk G, Meaker C, Engelbrecht AM. Nutritional support in sepsis: when less may be more. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:53. [PMID: 32059698 PMCID: PMC7023788 DOI: 10.1186/s13054-020-2771-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/10/2020] [Indexed: 12/28/2022]
Abstract
Despite sound basis to suspect that aggressive and early administration of nutritional support may hold therapeutic benefits during sepsis, recommendations for nutritional support have been somewhat underwhelming. Current guidelines (ESPEN and ASPEN) recognise a lack of clear evidence demonstrating the beneficial effect of nutritional support during sepsis, raising the question: why, given the perceived low efficacy of nutritionals support, are there no high-quality clinical trials on the efficacy of permissive underfeeding in sepsis? Here, we review clinically relevant beneficial effects of permissive underfeeding, motivating the urgent need to investigate the clinical benefits of delaying nutritional support during sepsis.
Collapse
Affiliation(s)
- Gustav van Niekerk
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa.
| | - Charné Meaker
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
37
|
Keller MD, Torres VJ, Cadwell K. Autophagy and microbial pathogenesis. Cell Death Differ 2020; 27:872-886. [PMID: 31896796 DOI: 10.1038/s41418-019-0481-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a cell biological process that promotes resilience in the face of environmental perturbations. Given that infectious agents represent a major type of environmental threat, it follows that the autophagy pathway is central to the outcome of host-microbe interactions. Detailed molecular studies have revealed intricate ways in which autophagy suppresses or enhances the fitness of infectious agents, particularly intracellular pathogens such as viruses that require the host cell machinery for replication. Findings in animal models have reinforced the importance of these events that occur within individual cells and have extended the role of autophagy to extracellular microbes and immunity at the whole organism level. These functions impact adaptation to bacteria that are part of the gut microbiota, which has implications for the etiology of chronic disorders such as inflammatory bowel disease. Despite major advances in how autophagy regulates inflammatory reactions toward microbes, many challenges remain, including distinguishing autophagy from closely related pathways such as LC3-associated phagocytosis. Here, we review the role of autophagy in microbial pathogenesis at the level of organismal biology. In addition to providing an overview of the prominent function of autophagy proteins in host-microbe interactions, we highlight how observations at the cellular level are informing pathogenesis studies and offer our perspective on the future directions of the field.
Collapse
Affiliation(s)
- Matthew D Keller
- Kimmel Center for Biology and Medicine, Skirball Institute, New York University School of Medicine, New York, NY, 10016, USA.,Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine, Skirball Institute, New York University School of Medicine, New York, NY, 10016, USA. .,Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA. .,Division of Gastroenterology and Hepatology, Department of Medicine, New York University Langone Health, New York, NY, 10016, USA.
| |
Collapse
|
38
|
Mellin R, Boddey JA. Organoids for Liver Stage Malaria Research. Trends Parasitol 2019; 36:158-169. [PMID: 31848118 DOI: 10.1016/j.pt.2019.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 11/30/2022]
Abstract
Plasmodium parasites cause malaria and are maintained between Anopheles mosquitoes and mammalian hosts in a complex life cycle. Malaria parasites occupy tissue niches that can be difficult to access, and models to study them can be challenging to recapitulate experimentally, particularly for Plasmodium species that infect humans. 2D culture models provide extremely beneficial tools to investigate Plasmodium biology but they have limitations. More complex 3D structural networks, such as organoids, have unveiled new avenues for developing more physiological tissue models, and their application to malaria research offers great promise. Here, we review current models for studying Plasmodium infection with a key focus on the obligate pre-erythrocytic stage that culminates in blood infection, causing malaria, and discuss how organoids should fulfil an important and unmet need.
Collapse
Affiliation(s)
- Ronan Mellin
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Victoria, Australia
| | - Justin A Boddey
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Victoria, Australia.
| |
Collapse
|
39
|
Sanches-Vaz M, Temporão A, Luis R, Nunes-Cabaço H, Mendes AM, Goellner S, Carvalho T, Figueiredo LM, Prudêncio M. Trypanosoma brucei infection protects mice against malaria. PLoS Pathog 2019; 15:e1008145. [PMID: 31703103 PMCID: PMC6867654 DOI: 10.1371/journal.ppat.1008145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 11/20/2019] [Accepted: 10/11/2019] [Indexed: 12/22/2022] Open
Abstract
Sleeping sickness and malaria are parasitic diseases with overlapping geographical distributions in sub-Saharan Africa. We hypothesized that the immune response elicited by an infection with Trypanosoma brucei, the etiological agent of sleeping sickness, would inhibit a subsequent infection by Plasmodium, the malaria parasite, decreasing the severity of its associated pathology. To investigate this, we established a new co-infection model in which mice were initially infected with T. brucei, followed by administration of P. berghei sporozoites. We observed that a primary infection by T. brucei significantly attenuates a subsequent infection by the malaria parasite, protecting mice from experimental cerebral malaria and prolonging host survival. We further observed that an ongoing T. brucei infection leads to an accumulation of lymphocyte-derived IFN-γ in the liver, limiting the establishment of a subsequent hepatic infection by P. berghei sporozoites. Thus, we identified a novel host-mediated interaction between two parasitic infections, which may be epidemiologically relevant in regions of Trypanosoma/Plasmodium co-endemicity. Despite the geographical overlap between the parasites that cause sleeping sickness and malaria, the reciprocal impact of a co-infection by T. brucei and Plasmodium had hitherto not been assessed. We hypothesized that the strong immune response elicited by a T. brucei infection could potentially limit the ability of Plasmodium parasites to infect the same host. In this study, we showed that a primary infection by T. brucei significantly attenuates a subsequent infection by the malaria parasite. Importantly, a significant proportion of the co-infected mice do not develop Plasmodium parasitemia, and those few that do, do not display symptoms of severe malaria and survive longer than their singly infected counterparts. We further showed that the prevention or delay in appearance of malaria parasites in the blood results from a dramatic impairment of the preceding liver infection by Plasmodium, which is mediated by the strong immune response mounted against the primary T. brucei infection. Our study provides new insights for a novel inter-pathogen interaction that may bear great epidemiological significance in regions of Trypanosoma/Plasmodium co-endemicity.
Collapse
Affiliation(s)
- Margarida Sanches-Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Adriana Temporão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rafael Luis
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Helena Nunes-Cabaço
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - António M. Mendes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sarah Goellner
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tânia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Luisa M. Figueiredo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- * E-mail: (LMF); (MP)
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- * E-mail: (LMF); (MP)
| |
Collapse
|
40
|
Galais M, Pradel B, Vergne I, Robert-Hebmann V, Espert L, Biard-Piechaczyk M. [LAP (LC3-associated phagocytosis): phagocytosis or autophagy?]. Med Sci (Paris) 2019; 35:635-642. [PMID: 31532375 DOI: 10.1051/medsci/2019129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Phagocytosis and macroautophagy, named here autophagy, are two essential mechanisms of lysosomal degradation of diverse cargos into membrane structures. Both mechanisms are involved in immune regulation and cell survival. However, phagocytosis triggers degradation of extracellular material whereas autophagy engulfs only cytoplasmic elements. Furthermore, activation and maturation of these two processes are different. LAP (LC3-associated phagocytosis) is a form of phagocytosis that uses components of the autophagy pathway. It can eliminate (i) pathogens, (ii) immune complexes, (iii) threatening neighbouring cells, dead or alive, and (iv) cell debris, such as POS (photoreceptor outer segment) and the midbody released at the end of mitosis. Cells have thus optimized their means of elimination of dangerous components by sharing some fundamental elements coming from the two main lysosomal degradation pathways.
Collapse
Affiliation(s)
- Mathilde Galais
- Institut de recherche en infectiologie de Montpellier (IRIM), Université de Montpellier, CNRS, 1919, route de Mende, 34293 Montpellier, France
| | - Baptiste Pradel
- Institut de recherche en infectiologie de Montpellier (IRIM), Université de Montpellier, CNRS, 1919, route de Mende, 34293 Montpellier, France
| | - Isabelle Vergne
- Institut de pharmacologie et de biologie structurale (IPBS), Université de Toulouse, CNRS, UPS, 205, route de Narbonne, 31400 Toulouse, France
| | - Véronique Robert-Hebmann
- Institut de recherche en infectiologie de Montpellier (IRIM), Université de Montpellier, CNRS, 1919, route de Mende, 34293 Montpellier, France
| | - Lucile Espert
- Institut de recherche en infectiologie de Montpellier (IRIM), Université de Montpellier, CNRS, 1919, route de Mende, 34293 Montpellier, France
| | - Martine Biard-Piechaczyk
- Institut de recherche en infectiologie de Montpellier (IRIM), Université de Montpellier, CNRS, 1919, route de Mende, 34293 Montpellier, France
| |
Collapse
|
41
|
LC3-associated phagocytosis: host defense and microbial response. Curr Opin Immunol 2019; 60:81-90. [PMID: 31247378 DOI: 10.1016/j.coi.2019.04.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/16/2019] [Indexed: 12/19/2022]
Abstract
The innate immune system has evolved to recognize diverse microbes and destroy them. At the same time, microbial pathogens undermine immunity to cause disease. Here, we highlight recent advances in understanding an antimicrobial pathway called LC3-associated phagocytosis (LAP), which combines features of autophagy with phagocytosis. Upon phagocytosis, many microbes, including bacteria, fungi, and parasites, are sequestered in an LC3-positive, single-membrane bound compartment, a hallmark of LAP. LAP depends upon NADPH oxidase activity at the incipient phagosome and culminates in lysosomal trafficking and microbial degradation. Most often LAP is an effective host defense, but some pathogens evade LAP or replicate successfully in this microenvironment. Here, we review how LAP targets microbial pathogens and strategies pathogens employ to circumvent LAP.
Collapse
|
42
|
Niklaus L, Agop-Nersesian C, Schmuckli-Maurer J, Wacker R, Grünig V, Heussler VT. Deciphering host lysosome-mediated elimination of Plasmodium berghei liver stage parasites. Sci Rep 2019; 9:7967. [PMID: 31138850 PMCID: PMC6538699 DOI: 10.1038/s41598-019-44449-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 05/13/2019] [Indexed: 02/08/2023] Open
Abstract
Liver stage Plasmodium parasites reside in a parasitophorous vacuole (PV) that associates with lysosomes. It has previously been shown that these organelles can have beneficial as well as harmful effects on the parasite. Yet it is not clear how the association of lysosomes with the parasite is controlled and how interactions with these organelles lead to the antagonistic outcomes. In this study we used advanced imaging techniques to characterize lysosomal interactions with the PV. In host cells harboring successfully developing parasites we observed that these interaction events reach an equilibrium at the PV membrane (PVM). In a population of arrested parasites, this equilibrium appeared to shift towards a strongly increased lysosomal fusion with the PVM witnessed by strong PVM labeling with the lysosomal marker protein LAMP1. This was followed by acidification of the PV and elimination of the parasite. To systematically investigate elimination of arrested parasites, we generated transgenic parasites that express the photosensitizer KillerRed, which leads to parasite killing after activation. Our work provides insights in cellular details of intracellular killing and lysosomal elimination of Plasmodium parasites independent of cells of the immune system.
Collapse
Affiliation(s)
- L Niklaus
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - C Agop-Nersesian
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | | | - R Wacker
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - V Grünig
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - V T Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| |
Collapse
|
43
|
Emran TB, Iyori M, Ono Y, Amelia F, Yusuf Y, Islam A, Alam A, Tamura M, Ogawa R, Matsuoka H, Yamamoto DS, Yoshida S. Baculovirus-Induced Fast-Acting Innate Immunity Kills Liver-Stage Plasmodium. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2441-2451. [PMID: 30209187 DOI: 10.4049/jimmunol.1800908] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/15/2018] [Indexed: 12/14/2022]
Abstract
Baculovirus (BV), an enveloped insect virus with a circular dsDNA genome, possesses unique characteristics that induce strong innate immune responses in mammalian cells. In this study, we show that BV administration in BALB/c mice not only provides complete protection against a subsequent Plasmodium berghei sporozoite infection for up to 7 d after the injection but also eliminates existing liver-stage parasites completely. The elimination of sporozoites by BV was superior to that by primaquine, and this effect occurred in a TLR9-independent manner. At 6 h after BV administration, IFN-α and IFN-γ were robustly produced in the serum, and RNA transcripts of IFN-stimulated genes were markedly upregulated in the liver compared with control mice. The in vivo passive transfer of serum after BV administration effectively eliminated liver-stage parasites, and IFN-α neutralization abolished this effect, indicating that the BV liver-stage parasite-killing mechanism is downstream of the type I IFN signaling pathway. These findings provide evidence that BV-induced, fast-acting innate immunity completely kills liver-stage parasites and, thus, may lead to new malaria drug and vaccine strategies.
Collapse
Affiliation(s)
- Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yuki Ono
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Asrar Alam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Megumi Tamura
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Ryohei Ogawa
- Department of Radiological Sciences, University of Toyama, Toyama 930-0194, Japan; and
| | - Hiroyuki Matsuoka
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0431, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0431, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa 920-1192, Japan;
| |
Collapse
|
44
|
De Niz M, Heussler VT. Rodent malaria models: insights into human disease and parasite biology. Curr Opin Microbiol 2018; 46:93-101. [PMID: 30317152 DOI: 10.1016/j.mib.2018.09.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/29/2018] [Accepted: 09/24/2018] [Indexed: 12/18/2022]
Abstract
The use of rodents as model organisms to study human disease is based on the genetic and physiological similarities between the species. Successful molecular methods to generate transgenic reporter or humanized rodents has rendered rodents as powerful tools for understanding biological processes and host-pathogen interactions relevant to humans. In malaria research, rodent models have been pivotal for the study of liver stages, syndromes arising from blood stages of infection, and malaria transmission to and from the mammalian host. Importantly, many in vivo findings are comparable to pathology observed in humans only when adequate combinations of rodent strains and Plasmodium parasites are used.
Collapse
Affiliation(s)
- Mariana De Niz
- Wellcome Centre for Molecular Parasitology, Glasgow, G12 8TA, UK; Institute for Cell Biology, University of Bern, CH-3012, Switzerland
| | - Volker T Heussler
- Institute for Cell Biology, University of Bern, CH-3012, Switzerland.
| |
Collapse
|
45
|
Gluschko A, Herb M, Wiegmann K, Krut O, Neiss WF, Utermöhlen O, Krönke M, Schramm M. The β 2 Integrin Mac-1 Induces Protective LC3-Associated Phagocytosis of Listeria monocytogenes. Cell Host Microbe 2018; 23:324-337.e5. [PMID: 29544096 DOI: 10.1016/j.chom.2018.01.018] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/26/2016] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
The intracellular pathogen Listeria monocytogenes (L.m.) is targeted by the autophagic machinery, but the molecular mechanisms involved and consequences for anti-listerial immunity remain enigmatic. Here, we demonstrate that L.m. infection of macrophages in vivo exclusively evokes LC3-associated phagocytosis (LAP), but not canonical autophagy, and that targeting of L.m. by LAP is required for anti-listerial immunity. The pathway leading to LAP induction in response to L.m. infection emanates from the β2 integrin Mac-1 (CR3, integrin αMβ2), a receptor recognizing diverse microbial ligands. Interaction of L.m. with Mac-1 induces acid sphingomyelinase-mediated changes in membrane lipid composition that facilitate assembly and activation of the phagocyte NAPDH oxidase Nox2. Nox2-derived reactive oxygen species then trigger LC3 recruitment to L.m.-containing phagosomes by LAP. By promoting fusion of L.m.-containing phagosomes with lysosomes, LAP increases exposure of L.m. to bactericidal acid hydrolases, thereby enhancing anti-listerial activity of macrophages and immunity of mice.
Collapse
Affiliation(s)
- Alexander Gluschko
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Katja Wiegmann
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Oleg Krut
- Microbiology Department, Paul- Ehrlich-Institut, Langen, Germany
| | - Wolfram F Neiss
- Department of Anatomy I, University of Cologne, Cologne, Germany
| | - Olaf Utermöhlen
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany; Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Martin Krönke
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany; Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases, (CECAD), University of Cologne, Cologne, Germany; German Center for Infection Research, Cologne, Germany
| | - Michael Schramm
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany.
| |
Collapse
|
46
|
Evans RJ, Sundaramurthy V, Frickel EM. The Interplay of Host Autophagy and Eukaryotic Pathogens. Front Cell Dev Biol 2018; 6:118. [PMID: 30271774 PMCID: PMC6146372 DOI: 10.3389/fcell.2018.00118] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022] Open
Abstract
For intracellular pathogens, host cells provide a replicative niche, but are also armed with innate defense mechanisms to combat the intruder. Co-evolution of host and pathogens has produced a complex interplay of host-pathogen interactions during infection, with autophagy emerging as a key player in the recent years. Host autophagy as a degradative process is a significant hindrance to intracellular growth of the pathogens, but also can be subverted by the pathogens to provide support such as nutrients. While the role of host cell autophagy in the pathogenesis mechanisms of several bacterial and viral pathogens have been extensively studied, less is known for eukaryotic pathogens. In this review, we focus on the interplay of host autophagy with the eukaryotic pathogens Plasmodium spp, Toxoplasma, Leishmania spp and the fungal pathogens Candida albicans, Aspergillus fumigatus and Cryptococcus neoformans. The differences between these eukaryotic pathogens in terms of the host cell types they infect, infective strategies and the host responses required to defend against them provide an interesting insight into how they respond to and interact with host cell autophagy. Due to the ability to infect multiple host species and cell types during the course of their usually complex lifestyles, autophagy plays divergent roles even for the same pathogen. The scenario is further compounded since many of the eukaryotic pathogens have their own sets of either complete or partial autophagy machinery. Eukaryotic pathogen-autophagy interplay is thus a complex relationship with many novel insights for the basic understanding of autophagy, and potential for clinical relevance.
Collapse
Affiliation(s)
- Robert J. Evans
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, London, United Kingdom
| | | | - Eva-Maria Frickel
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
47
|
Kyrmizi I, Ferreira H, Carvalho A, Figueroa JAL, Zarmpas P, Cunha C, Akoumianaki T, Stylianou K, Deepe GS, Samonis G, Lacerda JF, Campos A, Kontoyiannis DP, Mihalopoulos N, Kwon-Chung KJ, El-Benna J, Valsecchi I, Beauvais A, Brakhage AA, Neves NM, Latge JP, Chamilos G. Calcium sequestration by fungal melanin inhibits calcium-calmodulin signalling to prevent LC3-associated phagocytosis. Nat Microbiol 2018; 3:791-803. [PMID: 29849062 DOI: 10.1038/s41564-018-0167-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 04/23/2018] [Indexed: 11/09/2022]
Abstract
LC3-associated phagocytosis (LAP) is a non-canonical autophagy pathway regulated by Rubicon, with an emerging role in immune homeostasis and antifungal host defence. Aspergillus cell wall melanin protects conidia (spores) from killing by phagocytes and promotes pathogenicity through blocking nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-dependent activation of LAP. However, the signalling regulating LAP upstream of Rubicon and the mechanism of melanin-induced inhibition of this pathway remain incompletely understood. Herein, we identify a Ca2+ signalling pathway that depends on intracellular Ca2+ sources from endoplasmic reticulum, endoplasmic reticulum-phagosome communication, Ca2+ release from phagosome lumen and calmodulin (CaM) recruitment, as a master regulator of Rubicon, the phagocyte NADPH oxidase NOX2 and other molecular components of LAP. Furthermore, we provide genetic evidence for the physiological importance of Ca2+-CaM signalling in aspergillosis. Finally, we demonstrate that Ca2+ sequestration by Aspergillus melanin inside the phagosome abrogates activation of Ca2+-CaM signalling to inhibit LAP. These findings reveal the important role of Ca2+-CaM signalling in antifungal immunity and identify an immunological function of Ca2+ binding by melanin pigments with broad physiological implications beyond fungal disease pathogenesis.
Collapse
Affiliation(s)
- Irene Kyrmizi
- Department of Medicine, University of Crete, Heraklion, Crete, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Crete, Greece
| | - Helena Ferreira
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Julio Alberto Landero Figueroa
- Department of Chemistry, University of Cincinnati/Agilent Technologies Metallomics Center of the Americas, University of Cincinnati, Cincinnati, OH, USA
| | - Pavlos Zarmpas
- Department of Chemistry, University of Crete, Heraklion, Crete, Greece
| | - Cristina Cunha
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Tonia Akoumianaki
- Department of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Kostas Stylianou
- Department of Medicine, University of Crete, Heraklion, Crete, Greece
| | - George S Deepe
- Division of Infectious Diseases, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - George Samonis
- Department of Medicine, University of Crete, Heraklion, Crete, Greece
| | - João F Lacerda
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisbon, Portugal.,Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Lisbon, Portugal
| | - António Campos
- Serviço de Transplantação de Medula Óssea (STMO), Instituto Português de Oncologia do Porto, Porto, Portugal
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, The University of Texas, MD Anderson Cancer Center, Austin, TX, USA
| | | | - Kyung J Kwon-Chung
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jamel El-Benna
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | | | - Anne Beauvais
- Unité des Aspergillus, Institut Pasteur, Paris, France
| | - Axel A Brakhage
- Department of Molecular and Applied Microbiology, Leibniz-Institute for Natural Product Research and Infection Biology (HKI) and Friedrich Schiller University, Jena, Germany
| | - Nuno M Neves
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Georgios Chamilos
- Department of Medicine, University of Crete, Heraklion, Crete, Greece. .,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Crete, Greece.
| |
Collapse
|
48
|
Emran TB, Iyori M, Ono Y, Amelia F, Yusuf Y, Islam A, Alam A, Ogawa R, Matsuoka H, Yamamoto D, Yoshida S. Baculovirus-inducing fast-acting innate immunity kills Plasmodium liver stages.. [DOI: 10.1101/320036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
ABSTRACTBaculovirus (BV), an enveloped insect virus with a circular double-stranded DNA genome, possesses unique characteristics that induce strong innate immune responses in mammalian cells. Here, we show that BV administration not only sterilely protects BALB/c mice for at least 7 days from subsequent Plasmodium berghei sporozoite infection but also eliminates existing liver-stage parasites completely, effects superior to those of primaquine, and does so in a TLR9-independent manner. Six hours post-BV administration, IFN-α and IFN-γ were robustly produced in serum, and RNA transcripts of interferon-stimulated genes were drastically upregulated in the liver. The in vivo passive transfer of post-BV administration serum effectively eliminated liver-stage parasites, and IFN-α neutralization abolished this effect, indicating that the BV liver-stage parasite killing mechanism is downstream of the type I IFN signaling pathway. Our results demonstrate that BV is a potent IFN-inducing prophylactic and therapeutic agent with great potential for further development as a new malaria vaccine and/or anti-hypnozoite drug.
Collapse
|
49
|
Agop-Nersesian C, Niklaus L, Wacker R, Theo Heussler V. Host cell cytosolic immune response during Plasmodium liver stage development. FEMS Microbiol Rev 2018; 42:324-334. [PMID: 29529207 PMCID: PMC5995216 DOI: 10.1093/femsre/fuy007] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/25/2018] [Indexed: 02/07/2023] Open
Abstract
Recent years have witnessed a great gain in knowledge regarding parasite-host cell interactions during Plasmodium liver stage development. It is now an accepted fact that a large percentage of sporozoites invading hepatocytes fail to form infectious merozoites. There appears to be a delicate balance between parasite survival and elimination and we now start to understand why this is so. Plasmodium liver stage parasites replicate within the parasitophorous vacuole (PV), formed during invasion by invagination of the host cell plasma membrane. The main interface between the parasite and hepatocyte is the parasitophorous vacuole membrane (PVM) that surrounds the PV. Recently, it was shown that autophagy marker proteins decorate the PVM of Plasmodium liver stage parasites and eliminate a proportion of them by an autophagy-like mechanism. Successfully developing Plasmodium berghei parasites are initially also labeled but in the course of development, they are able to control this host defense mechanism by shedding PVM material into the tubovesicular network (TVN), an extension of the PVM that releases vesicles into the host cell cytoplasm. Better understanding of the molecular events at the PVM/TVN during parasite elimination could be the basis of new antimalarial measures.
Collapse
Affiliation(s)
- Carolina Agop-Nersesian
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, MA 02118, USA
| | - Livia Niklaus
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, CH-3012 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Rahel Wacker
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, CH-3012 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Volker Theo Heussler
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, CH-3012 Bern, Switzerland
| |
Collapse
|
50
|
Orjuela-Sanchez P, Villa ZH, Moreno M, Tong-Rios C, Meister S, LaMonte GM, Campo B, Vinetz JM, Winzeler EA. Developing Plasmodium vivax Resources for Liver Stage Study in the Peruvian Amazon Region. ACS Infect Dis 2018. [PMID: 29542317 DOI: 10.1021/acsinfecdis.7b00198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To develop new drugs and vaccines for malaria elimination, it will be necessary to discover biological interventions, including small molecules that act against Plasmodium vivax exoerythrocytic forms. However, a robust in vitro culture system for P. vivax is still lacking. Thus, to study exoerythrocytic forms, researchers must have simultaneous access to fresh, temperature-controlled patient blood samples, as well as an anopheline mosquito colony. In addition, researchers must rely on native mosquito species to avoid introducing a potentially dangerous invasive species into a malaria-endemic region. Here, we report an in vitro culture system carried out on site in a malaria-endemic region for liver stage parasites of P. vivax sporozoites obtained from An. darlingi, the main malaria vector in the Americas. P. vivax sporozoites were obtained by dissection of salivary glands from infected An. darlingi mosquitoes and purified by Accudenz density gradient centrifugation. HC04 liver cells were exposed to P. vivax sporozoites and cultured up to 9 days. To overcome low P. vivax patient parasitemias, potentially lower mosquito vectorial capacity, and humid, nonsterile environmental conditions, a new antibiotic cocktail was included in tissue culture to prevent contamination. Culturing conditions supported exoerythrocytic (EEF) P. vivax liver stage growth up to 9 days and allowed for maturation into intrahepatocyte merosomes. Some of the identified small forms were resistant to atovaquone (1 μM) but sensitive to the phosphatidylinositol 4-kinase inhibitor, KDU691 (1 μM). This study reports a field-accessible EEF production process for drug discovery in a malaria-endemic site in which viable P. vivax sporozoites are used for drug studies using hepatocyte infection. Our data demonstrate that the development of meaningful, field-based resources for P. vivax liver stage drug screening and liver stage human malaria experimentation in the Amazon region is feasible.
Collapse
Affiliation(s)
- Pamela Orjuela-Sanchez
- Division of Host-Microbe Systems and Therapeutics, Health Sciences Center for Immunology, Infection and Inflammation, Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| | - Zaira Hellen Villa
- Laboratorio ICEMR-Amazonia, Laboratorios de Investigacion y Desarrollo, Facultad de Ciencias y Filosofia, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porres, Lima, 15102, Peru
| | - Marta Moreno
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| | - Carlos Tong-Rios
- Laboratorio ICEMR-Amazonia, Laboratorios de Investigacion y Desarrollo, Facultad de Ciencias y Filosofia, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porres, Lima, 15102, Peru
| | - Stephan Meister
- Division of Host-Microbe Systems and Therapeutics, Health Sciences Center for Immunology, Infection and Inflammation, Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| | - Gregory M. LaMonte
- Division of Host-Microbe Systems and Therapeutics, Health Sciences Center for Immunology, Infection and Inflammation, Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| | - Brice Campo
- Medicines for Malaria Venture (MMV), International Center Cointrin, Block G, 20 Route de Pre-Bois, POB 1826, Geneva, CH-1215, Switzerland
| | - Joseph M. Vinetz
- Laboratorio ICEMR-Amazonia, Laboratorios de Investigacion y Desarrollo, Facultad de Ciencias y Filosofia, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porres, Lima, 15102, Peru
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
- Instituto de Medicina Tropical “Alexander von Humboldt”, Universidad Peruana Cayetano Heredia, Lima, 15102, Peru
| | - Elizabeth A. Winzeler
- Division of Host-Microbe Systems and Therapeutics, Health Sciences Center for Immunology, Infection and Inflammation, Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| |
Collapse
|