1
|
Fan YM, Zhang QQ, Pan M, Hou ZF, Fu L, Xu X, Huang SY. Toxoplasma gondii Sustains Survival by Regulating Cholesterol Biosynthesis and Uptake via SREBP2 Activation. J Lipid Res 2024:100684. [PMID: 39490926 DOI: 10.1016/j.jlr.2024.100684] [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: 07/24/2024] [Revised: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024] Open
Abstract
Toxoplasma gondii (T. gondii) is an obligate intracellular parasite that cannot biosynthesize cholesterol via the mevalonate pathway, it sources this lipid from its host. We discovered that T. gondii infection upregulated the expression of host cholesterol synthesis related genes HMG-CoA reductase(HMGCR), squalene epoxidase (SQLE) and dehydrocholesterol reductase-7 (DHCR7), and increased the uptake pathway gene low-density lipoprotein receptor (LDLR). We found a protein, sterol regulatory element binding protein 2 (SREBP2), which is the key protein regulating the host cholesterol synthesis and uptake during T. gondii infection. T. gondii induced a dose-dependent nuclear translocation of SREBP2. Knockdown SREBP2 reduced T. gondii-induced cholesterol biosynthesis and uptake. Consequently, the parasite's ability to acquire cholesterol was significantly diminished, impairing its invasion, replication, and bradyzoites development. Interfering cholesterol metabolism using AY9944 effectively inhibited T. gondii replication. In summary, SREBP2 played an important role in T. gondii infection in vitro, serving as a potential target for regulating T. gondii-induced cholesterol metabolism, offering insights into the prevention and treatment of toxoplasmosis.
Collapse
Affiliation(s)
- Yi-Min Fan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu Province 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, PR China
| | - Qing-Qi Zhang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu Province 225009, PR China
| | - Ming Pan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu Province 225009, PR China
| | - Zhao-Feng Hou
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu Province 225009, PR China
| | - Lizhi Fu
- Chongqing Academy of Animal Sciences, Chongqing, PR China
| | - Xiulong Xu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu Province 225009, PR China
| | - Si-Yang Huang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu Province 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, PR China; Chongqing Academy of Animal Sciences, Chongqing, PR China.
| |
Collapse
|
2
|
He TY, Li YT, Liu ZD, Cheng H, Bao YF, Zhang JL. Lipid metabolism: the potential targets for toxoplasmosis treatment. Parasit Vectors 2024; 17:111. [PMID: 38448975 PMCID: PMC10916224 DOI: 10.1186/s13071-024-06213-9] [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: 10/21/2023] [Accepted: 02/23/2024] [Indexed: 03/08/2024] Open
Abstract
Toxoplasmosis is a zoonosis caused by Toxoplasma gondii (T. gondii). The current treatment for toxoplasmosis remains constrained due to the absence of pharmaceutical interventions. Thus, the pursuit of more efficient targets is of great importance. Lipid metabolism in T. gondii, including fatty acid metabolism, phospholipid metabolism, and neutral lipid metabolism, assumes a crucial function in T. gondii because those pathways are largely involved in the formation of the membranous structure and cellular processes such as division, invasion, egress, replication, and apoptosis. The inhibitors of T. gondii's lipid metabolism can directly lead to the disturbance of various lipid component levels and serious destruction of membrane structure, ultimately leading to the death of the parasites. In this review, the specific lipid metabolism pathways, correlative enzymes, and inhibitors of lipid metabolism of T. gondii are elaborated in detail to generate novel ideas for the development of anti-T. gondii drugs that target the parasites' lipid metabolism.
Collapse
Affiliation(s)
- Tian-Yi He
- Health Science Center, Ningbo University, Ningbo, China
| | - Ye-Tian Li
- Health Science Center, Ningbo University, Ningbo, China
| | - Zhen-Di Liu
- Health Science Center, Ningbo University, Ningbo, China
| | - Hao Cheng
- Health Science Center, Ningbo University, Ningbo, China
| | - Yi-Feng Bao
- Health Science Center, Ningbo University, Ningbo, China
| | - Ji-Li Zhang
- Health Science Center, Ningbo University, Ningbo, China.
| |
Collapse
|
3
|
He K, Wang Q, Gao X, Tang T, Ding H, Long S. Transcriptomic and metabolomic analyses reveal the essential nature of Rab1B in Toxoplasma gondii. Parasit Vectors 2023; 16:409. [PMID: 37941035 PMCID: PMC10634116 DOI: 10.1186/s13071-023-06030-6] [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: 09/13/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND The protozoan parasite Toxoplasma gondii encodes a dozen Rab proteins, which are parts of the small GTPase superfamily and regulate intracellular membrane trafficking. Our previous study showed that depletion of Rab1B caused severe defects regarding parasite growth and morphological structure, yet early defects of endocytic trafficking and vesicle sorting to the rhoptry in T. gondii are not expected to have a strong effect. To understand this discrepancy, we performed an integrated analysis at the level of transcriptomics and metabolomics. METHODS In the study, tetracycline-inducible TATi/Ty-Rab1B parasite line treated with ATc at three different time points (0, 18 and 24 h) was used. We first observed the morphological changes caused by Rab1B depletion via transmission electron technology. Then, high-throughput transcriptome along with non-targeted metabolomics were performed to analyze the RNA expression and metabolite changes in the Rab1B-depleted parasite. The essential nature of Rab1B in the parasite was revealed by the integrated omics approach. RESULTS Transmission electron micrographs showed a strong disorganization of endo-membranes in the Rab1B-depleted parasites. Our deep analysis of transcriptome and metabolome identified 2181 and 2374 differentially expressed genes (DEGs) and 30 and 83 differentially expressed metabolites (DEMs) at 18 and 24 h of induction in the tetracycline-inducible parasite line, respectively. These DEGs included key genes associated with crucial organelles that contain the rhoptry, microneme, endoplasmic reticulum and Golgi apparatus. The analysis of qRT-PCR verified some of the key DEGs identified by RNA-Seq, supporting that the key vesicular regulator Rab1B was involved in biogenesis of multiple parasite organelles. Functional enrichment analyses revealed pathways related to central carbon metabolisms and lipid metabolisms, such as the TCA cycle, glycerophospholipid metabolism and fatty acid biosynthesis and elongation. Further correlation analysis of the major DEMs and DEGs supported the role of Rab1B in biogenesis of fatty acids (e.g. myrisoleic acid and oleic acid) (R > 0.95 and P < 0.05), which was consistent with the scavenging role in biotin via the endocytic process. CONCLUSIONS Rab1B played an important role in parasite growth and morphology, which was supported by the replication assay and transmission electron microscopy observation. Our multi-omics analyses provided detailed insights into the overall impact on the parasite upon depletion of the protein. These analyses reinforced the role of Rab1B in the endocytic process, which has an impact on fatty acid biogenesis and the TCA cycle. Taken together, these findings contribute to our understanding of a key vesicular regulator, Rab1B, on parasite metabolism and morphological formation in T. gondii.
Collapse
Affiliation(s)
- Kai He
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
- National KeyLaboratory of Veterinary Public Health Safety, School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Qiangqiang Wang
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
- National KeyLaboratory of Veterinary Public Health Safety, School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xuwen Gao
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
- National KeyLaboratory of Veterinary Public Health Safety, School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Tao Tang
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
- National KeyLaboratory of Veterinary Public Health Safety, School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Huiyong Ding
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
- National KeyLaboratory of Veterinary Public Health Safety, School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Shaojun Long
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
- National KeyLaboratory of Veterinary Public Health Safety, School of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
4
|
Asady B, Sampels V, Romano JD, Levitskaya J, Lige B, Khare P, Le A, Coppens I. Function and regulation of a steroidogenic CYP450 enzyme in the mitochondrion of Toxoplasma gondii. PLoS Pathog 2023; 19:e1011566. [PMID: 37651449 PMCID: PMC10499268 DOI: 10.1371/journal.ppat.1011566] [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: 01/05/2023] [Revised: 09/13/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
As an obligate intracellular parasite, Toxoplasma gondii must import essential nutrients from the host cell into the parasitophorous vacuole. We previously reported that the parasite scavenges cholesterol from host endocytic organelles for incorporation into membranes and storage as cholesteryl esters in lipid droplets. In this study, we have investigated whether Toxoplasma utilizes cholesterol as a precursor for the synthesis of metabolites, such as steroids. In mammalian cells, steroidogenesis occurs in mitochondria and involves membrane-bound type I cytochrome P450 oxidases that are activated through interaction with heme-binding proteins containing a cytochrome b5 domain, such as members of the membrane-associated progesterone receptor (MAPR) family. Our LC-MS targeted lipidomics detect selective classes of hormone steroids in Toxoplasma, with a predominance for anti-inflammatory hydroxypregnenolone species, deoxycorticosterone and dehydroepiandrosterone. The genome of Toxoplasma contains homologs encoding a single type I CYP450 enzyme (we named TgCYP450mt) and a single MAPR (we named TgMAPR). We showed that TgMAPR is a hemoprotein with conserved residues in a heme-binding cytochrome b5 domain. Both TgCYP450 and TgMAPR localize to the mitochondrion and show interactions in in situ proximity ligation assays. Genetic ablation of cyp450mt is not tolerated by Toxoplasma; we therefore engineered a conditional knockout strain and showed that iΔTgCYP450mt parasites exhibit growth impairment in cultured cells. Parasite strains deficient for mapr could be generated; however, ΔTgMAPR parasites suffer from poor global fitness, loss of plasma membrane integrity, aberrant mitochondrial cristae, and an abnormally long S-phase in their cell cycle. Compared to wild-type parasites, iΔTgCYP450mt and ΔTgMAPR lost virulence in mice and metabolomics studies reveal that both mutants have reduced levels of steroids. These observations point to a steroidogenic pathway operational in the mitochondrion of a protozoan that involves an evolutionary conserved TgCYP450mt enzyme and its binding partner TgMAPR.
Collapse
Affiliation(s)
- Beejan Asady
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Vera Sampels
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Julia D. Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jelena Levitskaya
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Bao Lige
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Pratik Khare
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Anne Le
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
5
|
Velásquez ZD, Rojas-Barón L, Larrazabal C, Salierno M, Gärtner U, Pervizaj-Oruqaj L, Herold S, Hermosilla C, Taubert A. Neospora caninum Infection Triggers S-phase Arrest and Alters Nuclear Characteristics in Primary Bovine Endothelial Host Cells. Front Cell Dev Biol 2022; 10:946335. [PMID: 36111335 PMCID: PMC9469085 DOI: 10.3389/fcell.2022.946335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Abstract
Neospora caninum represents a major cause of abortive disease in bovines and small ruminants worldwide. As a typical obligate intracellular apicomplexan parasite, N. caninum needs to modulate its host cell for successful replication. In the current study, we focused on parasite-driven interference with host cell cycle progression. By performing DNA content-based cell cycle phase analyses in N. caninum-infected primary bovine umbilical vein endothelial cells (BUVEC), a parasite-driven S-phase arrest was detected at both 24 and 32 h p. i., being paralleled by fewer host cells experiencing the G0/G1 cell cycle phase. When analyzing S-subphases, proliferation cell nuclear antigen (per PCNA)-based experiments showed a reduced population of BUVEC in the late S-phase. Analyses on key molecules of cell cycle regulation documented a significant alteration of cyclin A2 and cyclin B1 abundance in N. caninum-infected host endothelial cells, thereby confirming irregularities in the S-phase and S-to-G2/M-phase transition. In line with cell cycle alterations, general nuclear parameters revealed smaller nuclear sizes and morphological abnormalities of BUVEC nuclei within the N. caninum-infected host cell layer. The latter observations were also confirmed by transmission electron microscopy (TEM) and by analyses of lamin B1 as a marker of nuclear lamina, which illustrated an inhomogeneous nuclear lamin B1 distribution, nuclear foldings, and invaginations, thereby reflecting nuclear misshaping. Interestingly, the latter finding applied to both non-infected and infected host cells within parasitized BUVEC layer. Additionally, actin detection indicated alterations in the perinuclear actin cap formation since typical nucleo-transversal filaments were consistently lacking in N. caninum-infected BUVEC, as also documented by significantly decreased actin-related intensities in the perinuclear region. These data indicate that N. caninum indeed alters host cell cycle progression and severely affects the host cell nuclear phenotype in primary bovine endothelial host cells. In summary, these findings add novel data on the complex N. caninum-specific modulation of host cell and nucleus, thereby demonstrating clear differences in cell cycle progression modulation driven by other closely related apicomplexans like Toxoplasma gondii and Besnotia besnoiti.
Collapse
Affiliation(s)
- Zahady D. Velásquez
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
- *Correspondence: Zahady D. Velásquez,
| | - Lisbeth Rojas-Barón
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Camilo Larrazabal
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Marcelo Salierno
- Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Ulrich Gärtner
- Institute of Anatomy and Cell Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Learta Pervizaj-Oruqaj
- Department of Medicine V Internal Medicine Infectious Diseases and Infection Control Universities of Giessen and Marburg Lung Center (UGMLC) Member of the German Center for Lung Research (DZL) Justus-Liebig University Giessen, Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
- Excellence Cluster Cardipulmonary Institute (CPI), Giessen, Germany
| | - Susanne Herold
- Department of Medicine V Internal Medicine Infectious Diseases and Infection Control Universities of Giessen and Marburg Lung Center (UGMLC) Member of the German Center for Lung Research (DZL) Justus-Liebig University Giessen, Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
- Excellence Cluster Cardipulmonary Institute (CPI), Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
6
|
Shunmugam S, Arnold CS, Dass S, Katris NJ, Botté CY. The flexibility of Apicomplexa parasites in lipid metabolism. PLoS Pathog 2022; 18:e1010313. [PMID: 35298557 PMCID: PMC8929637 DOI: 10.1371/journal.ppat.1010313] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Apicomplexa are obligate intracellular parasites responsible for major human infectious diseases such as toxoplasmosis and malaria, which pose social and economic burdens around the world. To survive and propagate, these parasites need to acquire a significant number of essential biomolecules from their hosts. Among these biomolecules, lipids are a key metabolite required for parasite membrane biogenesis, signaling events, and energy storage. Parasites can either scavenge lipids from their host or synthesize them de novo in a relict plastid, the apicoplast. During their complex life cycle (sexual/asexual/dormant), Apicomplexa infect a large variety of cells and their metabolic flexibility allows them to adapt to different host environments such as low/high fat content or low/high sugar levels. In this review, we discuss the role of lipids in Apicomplexa parasites and summarize recent findings on the metabolic mechanisms in host nutrient adaptation.
Collapse
Affiliation(s)
- Serena Shunmugam
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Christophe-Sébastien Arnold
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Sheena Dass
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Nicholas J. Katris
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Cyrille Y. Botté
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| |
Collapse
|
7
|
Fader Kaiser CM, Romano PS, Vanrell MC, Pocognoni CA, Jacob J, Caruso B, Delgui LR. Biogenesis and Breakdown of Lipid Droplets in Pathological Conditions. Front Cell Dev Biol 2022; 9:826248. [PMID: 35198567 PMCID: PMC8860030 DOI: 10.3389/fcell.2021.826248] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/22/2021] [Indexed: 12/17/2022] Open
Abstract
Lipid droplets (LD) have long been considered as mere fat drops; however, LD have lately been revealed to be ubiquitous, dynamic and to be present in diverse organelles in which they have a wide range of key functions. Although incompletely understood, the biogenesis of eukaryotic LD initiates with the synthesis of neutral lipids (NL) by enzymes located in the endoplasmic reticulum (ER). The accumulation of NL leads to their segregation into nanometric nuclei which then grow into lenses between the ER leaflets as they are further filled with NL. The lipid composition and interfacial tensions of both ER and the lenses modulate their shape which, together with specific ER proteins, determine the proneness of LD to bud from the ER toward the cytoplasm. The most important function of LD is the buffering of energy. But far beyond this, LD are actively integrated into physiological processes, such as lipid metabolism, control of protein homeostasis, sequestration of toxic lipid metabolic intermediates, protection from stress, and proliferation of tumours. Besides, LD may serve as platforms for pathogen replication and defense. To accomplish these functions, from biogenesis to breakdown, eukaryotic LD have developed mechanisms to travel within the cytoplasm and to establish contact with other organelles. When nutrient deprivation occurs, LD undergo breakdown (lipolysis), which begins with the LD-associated members of the perilipins family PLIN2 and PLIN3 chaperone-mediated autophagy degradation (CMA), a specific type of autophagy that selectively degrades a subset of cytosolic proteins in lysosomes. Indeed, PLINs CMA degradation is a prerequisite for further true lipolysis, which occurs via cytosolic lipases or by lysosome luminal lipases when autophagosomes engulf portions of LD and target them to lysosomes. LD play a crucial role in several pathophysiological processes. Increased accumulation of LD in non-adipose cells is commonly observed in numerous infectious diseases caused by intracellular pathogens including viral, bacterial, and parasite infections, and is gradually recognized as a prominent characteristic in a variety of cancers. This review discusses current evidence related to the modulation of LD biogenesis and breakdown caused by intracellular pathogens and cancer.
Collapse
Affiliation(s)
- Claudio M Fader Kaiser
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Patricia S Romano
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - M Cristina Vanrell
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Cristian A Pocognoni
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Julieta Jacob
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Benjamín Caruso
- Instituto de Investigaciones Biologicas y Tecnologicas, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Laura R Delgui
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| |
Collapse
|
8
|
de Souza G, Silva RJ, Milián ICB, Rosini AM, de Araújo TE, Teixeira SC, Oliveira MC, Franco PS, da Silva CV, Mineo JR, Silva NM, Ferro EAV, Barbosa BF. Cyclooxygenase (COX)-2 modulates Toxoplasma gondii infection, immune response and lipid droplets formation in human trophoblast cells and villous explants. Sci Rep 2021; 11:12709. [PMID: 34135407 PMCID: PMC8209052 DOI: 10.1038/s41598-021-92120-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/04/2021] [Indexed: 01/01/2023] Open
Abstract
Congenital toxoplasmosis is represented by the transplacental passage of Toxoplasma gondii from the mother to the fetus. Our studies demonstrated that T. gondii developed mechanisms to evade of the host immune response, such as cyclooxygenase (COX)-2 and prostaglandin E2 (PGE2) induction, and these mediators can be produced/stored in lipid droplets (LDs). The aim of this study was to evaluate the role of COX-2 and LDs during T. gondii infection in human trophoblast cells and villous explants. Our data demonstrated that COX-2 inhibitors decreased T. gondii replication in trophoblast cells and villous. In BeWo cells, the COX-2 inhibitors induced an increase of pro-inflammatory cytokines (IL-6 and MIF), and a decrease in anti-inflammatory cytokines (IL-4 and IL-10). In HTR-8/SVneo cells, the COX-2 inhibitors induced an increase of IL-6 and nitrite and decreased IL-4 and TGF-β1. In villous explants, the COX-2 inhibitors increased MIF and decreased TNF-α and IL-10. Furthermore, T. gondii induced an increase in LDs in BeWo and HTR-8/SVneo, but COX-2 inhibitors reduced LDs in both cells type. We highlighted that COX-2 is a key factor to T. gondii proliferation in human trophoblast cells, since its inhibition induced a pro-inflammatory response capable of controlling parasitism and leading to a decrease in the availability of LDs, which are essentials for parasite growth.
Collapse
Affiliation(s)
- Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Rafaela José Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Iliana Claudia Balga Milián
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Thádia Evelyn de Araújo
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Mário Cézar Oliveira
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Priscila Silva Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Claudio Vieira da Silva
- Laboratory of Trypanosomatids, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - José Roberto Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil.
| |
Collapse
|
9
|
Onguka O, Babin BM, Lakemeyer M, Foe IT, Amara N, Terrell SM, Lum KM, Cieplak P, Niphakis MJ, Long JZ, Bogyo M. Toxoplasma gondii serine hydrolases regulate parasite lipid mobilization during growth and replication within the host. Cell Chem Biol 2021; 28:1501-1513.e5. [PMID: 34043961 DOI: 10.1016/j.chembiol.2021.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/09/2021] [Accepted: 05/04/2021] [Indexed: 12/24/2022]
Abstract
The intracellular protozoan parasite Toxoplasma gondii must scavenge cholesterol and other lipids from the host to facilitate intracellular growth and replication. Enzymes responsible for neutral lipid synthesis have been identified but there is no evidence for enzymes that catalyze lipolysis of cholesterol esters and esterified lipids. Here, we characterize several T. gondii serine hydrolases with esterase and thioesterase activities that were previously thought to be depalmitoylating enzymes. We find they do not cleave palmitoyl thiol esters but rather hydrolyze short-chain lipid esters. Deletion of one of the hydrolases results in alterations in levels of multiple lipids species. We also identify small-molecule inhibitors of these hydrolases and show that treatment of parasites results in phenotypic defects reminiscent of parasites exposed to excess cholesterol or oleic acid. Together, these data characterize enzymes necessary for processing lipids critical for infection and highlight the potential for targeting parasite hydrolases for therapeutic applications.
Collapse
Affiliation(s)
- Ouma Onguka
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brett M Babin
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Markus Lakemeyer
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ian T Foe
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Neri Amara
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stephanie M Terrell
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Kenneth M Lum
- Lundbeck La Jolla Research Center, San Diego, CA 92121, USA
| | - Piotr Cieplak
- Infectious & Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | | | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
10
|
Gubbels MJ, Coppens I, Zarringhalam K, Duraisingh MT, Engelberg K. The Modular Circuitry of Apicomplexan Cell Division Plasticity. Front Cell Infect Microbiol 2021; 11:670049. [PMID: 33912479 PMCID: PMC8072463 DOI: 10.3389/fcimb.2021.670049] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/22/2021] [Indexed: 12/31/2022] Open
Abstract
The close-knit group of apicomplexan parasites displays a wide variety of cell division modes, which differ between parasites as well as between different life stages within a single parasite species. The beginning and endpoint of the asexual replication cycles is a 'zoite' harboring the defining apical organelles required for host cell invasion. However, the number of zoites produced per division round varies dramatically and can unfold in several different ways. This plasticity of the cell division cycle originates from a combination of hard-wired developmental programs modulated by environmental triggers. Although the environmental triggers and sensors differ between species and developmental stages, widely conserved secondary messengers mediate the signal transduction pathways. These environmental and genetic input integrate in division-mode specific chromosome organization and chromatin modifications that set the stage for each division mode. Cell cycle progression is conveyed by a smorgasbord of positively and negatively acting transcription factors, often acting in concert with epigenetic reader complexes, that can vary dramatically between species as well as division modes. A unique set of cell cycle regulators with spatially distinct localization patterns insert discrete check points which permit individual control and can uncouple general cell cycle progression from nuclear amplification. Clusters of expressed genes are grouped into four functional modules seen in all division modes: 1. mother cytoskeleton disassembly; 2. DNA replication and segregation (D&S); 3. karyokinesis; 4. zoite assembly. A plug-and-play strategy results in the variety of extant division modes. The timing of mother cytoskeleton disassembly is hard-wired at the species level for asexual division modes: it is either the first step, or it is the last step. In the former scenario zoite assembly occurs at the plasma membrane (external budding), and in the latter scenario zoites are assembled in the cytoplasm (internal budding). The number of times each other module is repeated can vary regardless of this first decision, and defines the modes of cell division: schizogony, binary fission, endodyogeny, endopolygeny.
Collapse
Affiliation(s)
- Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Kourosh Zarringhalam
- Department of Mathematics, University of Massachusetts Boston, Boston, MA, United States
| | - Manoj T. Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| | - Klemens Engelberg
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| |
Collapse
|
11
|
Tavares VDS, de Castro MV, Souza RDSO, Gonçalves IKA, Lima JB, Borges VDM, Araújo-Santos T. Lipid droplets of protozoan parasites: survival and pathogenicity. Mem Inst Oswaldo Cruz 2021; 116:e210270. [PMID: 35195194 PMCID: PMC8851939 DOI: 10.1590/0074-02760210270] [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: 08/11/2021] [Accepted: 12/14/2021] [Indexed: 12/04/2022] Open
Abstract
Lipid droplets (LDs; lipid bodies) are intracellular sites of lipid storage and metabolism present in all cell types. Eukaryotic LDs are involved in eicosanoid production during several inflammatory conditions, including infection by protozoan parasites. In parasites, LDs play a role in the acquisition of cholesterol and other neutral lipids from the host. The number of LDs increases during parasite differentiation, and the biogenesis of these organelles use specific signaling pathways involving protein kinases. In addition, LDs are important in cellular protection against lipotoxicity. Recently, these organelles have been implicated in eicosanoid and specialised lipid metabolism. In this article, we revise the main functions of protozoan parasite LDs and discuss future directions in the comprehension of these organelles in the context of pathogen virulence.
Collapse
Affiliation(s)
| | | | | | | | - Jonilson Berlink Lima
- Universidade Federal do Oeste da Bahia, Brasil; Fundação Oswaldo Cruz-Fiocruz, Brasil
| | | | - Théo Araújo-Santos
- Universidade Federal do Oeste da Bahia, Brasil; Fundação Oswaldo Cruz-Fiocruz, Brasil
| |
Collapse
|
12
|
Asady B, Dick CF, Ehrenman K, Sahu T, Romano JD, Coppens I. A single Na+-Pi cotransporter in Toxoplasma plays key roles in phosphate import and control of parasite osmoregulation. PLoS Pathog 2021; 16:e1009067. [PMID: 33383579 PMCID: PMC7817038 DOI: 10.1371/journal.ppat.1009067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 01/20/2021] [Accepted: 10/14/2020] [Indexed: 11/22/2022] Open
Abstract
Inorganic ions such as phosphate, are essential nutrients required for a broad spectrum of cellular functions and regulation. During infection, pathogens must obtain inorganic phosphate (Pi) from the host. Despite the essentiality of phosphate for all forms of life, how the intracellular parasite Toxoplasma gondii acquires Pi from the host cell is still unknown. In this study, we demonstrated that Toxoplasma actively internalizes exogenous Pi by exploiting a gradient of Na+ ions to drive Pi uptake across the plasma membrane. The Na+-dependent phosphate transport mechanism is electrogenic and functionally coupled to a cipargarmin sensitive Na+-H+-ATPase. Toxoplasma expresses one transmembrane Pi transporter harboring PHO4 binding domains that typify the PiT Family. This transporter named TgPiT, localizes to the plasma membrane, the inward buds of the endosomal organelles termed VAC, and many cytoplasmic vesicles. Upon Pi limitation in the medium, TgPiT is more abundant at the plasma membrane. We genetically ablated the PiT gene, and ΔTgPiT parasites are impaired in importing Pi and synthesizing polyphosphates. Interestingly, ΔTgPiT parasites accumulate 4-times more acidocalcisomes, storage organelles for phosphate molecules, as compared to parental parasites. In addition, these mutants have a reduced cell volume, enlarged VAC organelles, defects in calcium storage and a slightly alkaline pH. Overall, these mutants exhibit severe growth defects and have reduced acute virulence in mice. In survival mode, ΔTgPiT parasites upregulate several genes, including those encoding enzymes that cleave or transfer phosphate groups from phosphometabolites, transporters and ions exchangers localized to VAC or acidocalcisomes. Taken together, these findings point to a critical role of TgPiT for Pi supply for Toxoplasma and also for protection against osmotic stresses. Inorganic phosphate (Pi) is indispensable for the biosynthesis of key cellular components, and is involved in many metabolic and signaling pathways. Transport across the plasma membrane is the first step in the utilization of Pi. The import mechanism of Pi by the intracellular parasite Toxoplasma is unknown. We characterized a transmembrane, high-affinity Na+-Pi cotransporter, named TgPiT, expressed by the parasite at the plasma membrane for Pi uptake. Interestingly, TgPiT is also localized to inward buds of the endosomal VAC organelles and some cytoplasmic vesicles. Loss of TgPiT results in a severe reduction in Pi internalization and polyphosphate levels, but stimulation of the biogenesis of phosphate-enriched acidocalcisomes. ΔTgPiT parasites have a shrunken cell body, enlarged VAC organelles, poor release of stored calcium and a mildly alkaline pH, suggesting a role for TgPiT in the maintenance of overall ionic homeostasis. ΔTgPiT parasites are poorly infectious in vitro and in mice. The mutant appears to partially cope with the absence of TgPiT by up-regulating genes coding for ion transporters and enzymes catalyzing phosphate group transfer. Our data highlight a scenario in which the role of TgPiT in Pi and Na+ transport is functionally coupled with osmoregulation activities central to sustain Toxoplasma survival.
Collapse
Affiliation(s)
- Beejan Asady
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore Maryland, United States of America
| | - Claudia F. Dick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore Maryland, United States of America
| | - Karen Ehrenman
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore Maryland, United States of America
| | - Tejram Sahu
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore Maryland, United States of America
| | - Julia D. Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore Maryland, United States of America
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore Maryland, United States of America
- * E-mail:
| |
Collapse
|
13
|
Structural, Functional, and Metabolic Alterations in Human Cerebrovascular Endothelial Cells during Toxoplasma gondii Infection and Amelioration by Verapamil In Vitro. Microorganisms 2020; 8:microorganisms8091386. [PMID: 32927732 PMCID: PMC7564162 DOI: 10.3390/microorganisms8091386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Toxoplasma gondii (T. gondii), the causative agent of toxoplasmosis, is a frequent cause of brain infection. Despite its known ability to invade the brain, there is still a dire need to better understand the mechanisms by which this parasite interacts with and crosses the blood–brain barrier (BBB). The present study revealed structural and functional changes associated with infection and replication of T. gondii within human brain microvascular endothelial cells (BMECs) in vitro. T. gondii proliferated within the BMECs and disrupted the integrity of the cerebrovascular barrier through diminishing the cellular viability, disruption of the intercellular junctions and increasing permeability of the BMEC monolayer, as well as altering lipid homeostasis. Proton nuclear magnetic resonance (1H NMR)-based metabolomics combined with multivariate data analysis revealed profiles that can be attributed to infection and variations in the amounts of certain metabolites (e.g., amino acids, fatty acids) in the extracts of infected compared to control cells. Notably, treatment with the Ca2+ channel blocker verapamil rescued BMEC barrier integrity and restricted intracellular replication of the tachyzoites regardless of the time of treatment application (i.e., prior to infection, early- and late-infection). This study provides new insights into the structural and functional changes that accompany T. gondii infection of the BMECs, and sheds light upon the ability of verapamil to inhibit the parasite proliferation and to ameliorate the adverse effects caused by T. gondii infection.
Collapse
|
14
|
Gubbels MJ, Keroack CD, Dangoudoubiyam S, Worliczek HL, Paul AS, Bauwens C, Elsworth B, Engelberg K, Howe DK, Coppens I, Duraisingh MT. Fussing About Fission: Defining Variety Among Mainstream and Exotic Apicomplexan Cell Division Modes. Front Cell Infect Microbiol 2020; 10:269. [PMID: 32582569 PMCID: PMC7289922 DOI: 10.3389/fcimb.2020.00269] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022] Open
Abstract
Cellular reproduction defines life, yet our textbook-level understanding of cell division is limited to a small number of model organisms centered around humans. The horizon on cell division variants is expanded here by advancing insights on the fascinating cell division modes found in the Apicomplexa, a key group of protozoan parasites. The Apicomplexa display remarkable variation in offspring number, whether karyokinesis follows each S/M-phase or not, and whether daughter cells bud in the cytoplasm or bud from the cortex. We find that the terminology used to describe the various manifestations of asexual apicomplexan cell division emphasizes either the number of offspring or site of budding, which are not directly comparable features and has led to confusion in the literature. Division modes have been primarily studied in two human pathogenic Apicomplexa, malaria-causing Plasmodium spp. and Toxoplasma gondii, a major cause of opportunistic infections. Plasmodium spp. divide asexually by schizogony, producing multiple daughters per division round through a cortical budding process, though at several life-cycle nuclear amplifications stages, are not followed by karyokinesis. T. gondii divides by endodyogeny producing two internally budding daughters per division round. Here we add to this diversity in replication mechanisms by considering the cattle parasite Babesia bigemina and the pig parasite Cystoisospora suis. B. bigemina produces two daughters per division round by a “binary fission” mechanism whereas C. suis produces daughters through both endodyogeny and multiple internal budding known as endopolygeny. In addition, we provide new data from the causative agent of equine protozoal myeloencephalitis (EPM), Sarcocystis neurona, which also undergoes endopolygeny but differs from C. suis by maintaining a single multiploid nucleus. Overall, we operationally define two principally different division modes: internal budding found in cyst-forming Coccidia (comprising endodyogeny and two forms of endopolygeny) and external budding found in the other parasites studied (comprising the two forms of schizogony, binary fission and multiple fission). Progressive insights into the principles defining the molecular and cellular requirements for internal vs. external budding, as well as variations encountered in sexual stages are discussed. The evolutionary pressures and mechanisms underlying apicomplexan cell division diversification carries relevance across Eukaryota.
Collapse
Affiliation(s)
- Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Caroline D Keroack
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Sriveny Dangoudoubiyam
- Department of Veterinary Science, Gluck Equine Research Center, University of Kentucky, Lexington, KY, United States
| | - Hanna L Worliczek
- Department of Biology, Boston College, Chestnut Hill, MA, United States.,Institute of Parasitology, University of Veterinary Medicine, Vienna, Austria
| | - Aditya S Paul
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Ciara Bauwens
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Brendan Elsworth
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, United States.,School of Biosciences, University of Melbourne, Melbourne, VIC, Australia
| | - Klemens Engelberg
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Daniel K Howe
- Department of Veterinary Science, Gluck Equine Research Center, University of Kentucky, Lexington, KY, United States
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
| |
Collapse
|
15
|
Kluck GEG, Wendt CHC, Imperio GED, Araujo MFC, Atella TC, da Rocha I, Miranda KR, Atella GC. Plasmodium Infection Induces Dyslipidemia and a Hepatic Lipogenic State in the Host through the Inhibition of the AMPK-ACC Pathway. Sci Rep 2019; 9:14695. [PMID: 31604978 PMCID: PMC6789167 DOI: 10.1038/s41598-019-51193-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/12/2019] [Indexed: 12/18/2022] Open
Abstract
Malaria is a major parasitic disease of humans and is a health public problem that affects more than 100 countries. In 2017, it caused nearly half a million deaths out of 219 million infections. Malaria is caused by the protozoan parasites of the genus Plasmodium and is transmitted by female mosquitoes of the genus Anopheles. Once in the bloodstream, Plasmodium merozoites invade erythrocytes and proliferate until the cells lyses and release new parasites that invade other erythrocytes. Remarkably, they can manipulate the vertebrate host's lipid metabolism pathways, since they cannot synthesize lipid classes that are essential for their development and replication. In this study, we show that mice infected with Plasmodium chabaudi present a completely different plasma profile from control mice, with marked hyperproteinemia, hypertriglyceridemia, hypoglycemia, and hypocholesterolemia. In addition, white adipose and hepatic tissue and analyses from infected animals revealed the accumulation of triacylglycerol in both tissues and free fatty acids and free cholesterol in the liver. Hepatic mRNA and protein expression of key enzymes and transcription factors involved in lipid metabolism were also altered by P. chabaudi infection, leading to a lipogenic state. The enzyme 5' AMP-activated protein kinase (AMPK), a master regulator of cell energetic metabolism, was also modulated by the parasite, which reduced AMPK phosphorylation levels upon infection. Pretreatment with metformin for 21 days followed by infection with P. chabaudi was effective in preventing infection of mice and also lowered the hepatic accumulation of lipids while activating AMPK. Together, these results provide new and important information on the specific molecular mechanisms induced by the malaria parasite to regulate hepatic lipid metabolism in order to facilitate its development, proliferation, and lifespan in its vertebrate host.
Collapse
Affiliation(s)
- George Eduardo Gabriel Kluck
- Laboratory of Lipid and Lipoproteins Biochemistry, Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camila Hübner Costabile Wendt
- Laboratory of Cellular Ultrastructure Hertha Meyer, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guinever Eustaquio do Imperio
- Laboratory of Translational Endocrinology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Fernanda Carvalho Araujo
- Laboratory of Lipid and Lipoproteins Biochemistry, Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tainá Correa Atella
- Laboratory of Comparative Neurobiology and Development, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isabella da Rocha
- Laboratory of Lipid and Lipoproteins Biochemistry, Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kildare Rocha Miranda
- Laboratory of Translational Endocrinology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Georgia Correa Atella
- Laboratory of Lipid and Lipoproteins Biochemistry, Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Silva LMR, Lütjohann D, Hamid P, Velasquez ZD, Kerner K, Larrazabal C, Failing K, Hermosilla C, Taubert A. Besnoitia besnoiti infection alters both endogenous cholesterol de novo synthesis and exogenous LDL uptake in host endothelial cells. Sci Rep 2019; 9:6650. [PMID: 31040348 PMCID: PMC6491585 DOI: 10.1038/s41598-019-43153-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/12/2019] [Indexed: 02/07/2023] Open
Abstract
Besnoitia besnoiti, an apicomplexan parasite of cattle being considered as emergent in Europe, replicates fast in host endothelial cells during acute infection and is in considerable need for energy, lipids and other building blocks for offspring formation. Apicomplexa are generally considered as defective in cholesterol synthesis and have to scavenge cholesterol from their host cells for successful replication. Therefore, we here analysed the influence of B. besnoiti on host cellular endogenous cholesterol synthesis and on sterol uptake from exogenous sources. GC-MS-based profiling of cholesterol-related sterols revealed enhanced cholesterol synthesis rates in B. besnoiti-infected cells. Accordingly, lovastatin and zaragozic acid treatments diminished tachyzoite production. Moreover, increased lipid droplet contents and enhanced cholesterol esterification was detected and inhibition of the latter significantly blocked parasite proliferation. Furthermore, artificial increase of host cellular lipid droplet disposability boosted parasite proliferation. Interestingly, lectin-like oxidized low density lipoprotein receptor 1 expression was upregulated in infected endothelial hostcells, whilst low density lipoproteins (LDL) receptor was not affected by parasite infection. However, exogenous supplementations with non-modified and acetylated LDL both boosted B. besnoiti proliferation. Overall, current data show that B. besnoiti simultaneously exploits both, endogenous cholesterol biosynthesis and cholesterol uptake from exogenous sources, during asexual replication.
Collapse
Affiliation(s)
- Liliana M R Silva
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany.
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics Bonn, Laboratory for Special Lipid Diagnostics/Center Internal Medicine/Building 26/UG 68, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
| | - Penny Hamid
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany.,Department of Parasitology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Jl. Fauna No. 2 Karangmalang, 55281, Yogyakarta, Indonesia
| | - Zahady D Velasquez
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany
| | - Katharina Kerner
- Institute for Hygiene and Infectious Diseases of Animals, Justus-Liebig-University, Giessen, Frankfurter Str. 85-89, D-35392, Germany
| | - Camilo Larrazabal
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany
| | - Klaus Failing
- Unit for Biomathematics and Data Processing, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Frankfurter Str. 95, D-35392, Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany
| |
Collapse
|
17
|
Novel Approaches To Kill Toxoplasma gondii by Exploiting the Uncontrolled Uptake of Unsaturated Fatty Acids and Vulnerability to Lipid Storage Inhibition of the Parasite. Antimicrob Agents Chemother 2018; 62:AAC.00347-18. [PMID: 30061287 DOI: 10.1128/aac.00347-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/21/2018] [Indexed: 12/17/2022] Open
Abstract
Toxoplasma gondii, an obligate intracellular parasite replicating in mammalian cells within a parasitophorous vacuole (PV), is an avid scavenger of lipids retrieved from the host cell. Following lipid uptake, this parasite stores excess lipids in lipid droplets (LD). Here, we examined the lipid storage capacities of Toxoplasma upon supplementation of the culture medium with various fatty acids at physiological concentrations. Supplemental unsaturated fatty acids (oleate [OA], palmitoleate, linoleate) accumulate in large LD and impair parasite replication, whereas saturated fatty acids (palmitate, stearate) neither stimulate LD formation nor impact growth. Examination of parasite growth defects with 0.4 mM OA revealed massive lipid deposits outside LD, indicating enzymatic inadequacies for storing neutral lipids in LD in response to the copious salvage of OA. Toxoplasma exposure to 0.5 mM OA led to irreversible growth arrest and lipid-induced damage, confirming a major disconnect between fatty acid uptake and the parasite's cellular lipid requirements. The importance of neutral lipid synthesis and storage to avoid lipotoxicity was further highlighted by the selective vulnerability of Toxoplasma, both the proliferative and the encysted forms, to subtoxic concentrations of the acyl coenzyme A:diacylglycerol acyltransferase 1 (DGAT1) pharmacological inhibitor T863. T863-treated parasites did not form LD but instead built up large membranous structures within the cytoplasm, which suggests improper channeling and management of the excess lipid. Dual addition of OA and T863 to infected cells intensified the deterioration of the parasite. Overall, our data pinpoint Toxoplasma DGAT as a promising drug target for the treatment of toxoplasmosis that would not incur the risk of toxicity for mammalian cells.
Collapse
|
18
|
Functional Analyses of a Putative, Membrane-Bound, Peroxisomal Protein Import Mechanism from the Apicomplexan Protozoan Toxoplasma gondii. Genes (Basel) 2018; 9:genes9090434. [PMID: 30158461 PMCID: PMC6162456 DOI: 10.3390/genes9090434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 01/28/2023] Open
Abstract
Peroxisomes are central to eukaryotic metabolism, including the oxidation of fatty acids—which subsequently provide an important source of metabolic energy—and in the biosynthesis of cholesterol and plasmalogens. However, the presence and nature of peroxisomes in the parasitic apicomplexan protozoa remains controversial. A survey of the available genomes revealed that genes encoding peroxisome biogenesis factors, so-called peroxins (Pex), are only present in a subset of these parasites, the coccidia. The basic principle of peroxisomal protein import is evolutionarily conserved, proteins harbouring a peroxisomal-targeting signal 1 (PTS1) interact in the cytosol with the shuttling receptor Pex5 and are then imported into the peroxisome via the membrane-bound protein complex formed by Pex13 and Pex14. Surprisingly, whilst Pex5 is clearly identifiable, Pex13 and, perhaps, Pex14 are apparently absent from the coccidian genomes. To investigate the functionality of the PTS1 import mechanism in these parasites, expression of Pex5 from the model coccidian Toxoplasma gondii was shown to rescue the import defect of Pex5-deleted Saccharomyces cerevisiae. In support of these data, green fluorescent protein (GFP) bearing the enhanced (e)PTS1 known to efficiently localise to peroxisomes in yeast, localised to peroxisome-like bodies when expressed in Toxoplasma. Furthermore, the PTS1-binding domain of Pex5 and a PTS1 ligand from the putatively peroxisome-localised Toxoplasma sterol carrier protein (SCP2) were shown to interact in vitro. Taken together, these data demonstrate that the Pex5–PTS1 interaction is functional in the coccidia and indicate that a nonconventional peroxisomal import mechanism may operate in the absence of Pex13 and Pex14.
Collapse
|
19
|
Trypanosoma cruzi epimastigotes store cholesteryl esters in lipid droplets after cholesterol endocytosis. Mol Biochem Parasitol 2018; 224:6-16. [PMID: 30016698 DOI: 10.1016/j.molbiopara.2018.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/05/2018] [Accepted: 07/10/2018] [Indexed: 11/21/2022]
Abstract
The Chagas disease agent Trypanosoma cruzi proliferates in the insect vector as highly endocytic epimastigotes that store nutrients, including lipids in reservosomes (lysosome related compartments). Although nutrient storage is important for epimastigote transformation into infective metacyclics, the epimastigote lipid droplets (LDs) remain uncharacterized. Here, we characterized the epimastigote LDs and examined their relationship with the endocytic pathway. Fluorescence microscopy using BODIPY showed that LDs have high neutral lipid content and harbor Rab18, differently from other lipid-rich organelles (such as reservosomes). Using transmission electron microscopy (TEM), we observed a close relationship between LDs and the endoplasmic reticulum, mitochondria and glycosomes. We developed a reproducible protocol to isolate LDs, and showed (by HTPLC and GC/MS analyses) that they have 89% neutral lipids and 11% phospholipids, which are likely to form the LD monolayer seen by TEM. The LD neutral lipids were mostly sterols, although triacylglycerol, diacylglycerol, monoacylglycerol and free fatty acids (FFA) were also found. Endocytosis of 3H-labeled cholesterol-BSA showed that internalized cholesterol is stored in LDs mostly in the cholesteryl ester form. Together, these results suggest that exogenous cholesterol internalized by endocytosis reaches the reservosomes and is then stored into LDs after esterification.
Collapse
|
20
|
Taubert A, Silva LMR, Velásquez ZD, Larrazabal C, Lütjohann D, Hermosilla C. Modulation of cholesterol-related sterols during Eimeria bovis macromeront formation and impact of selected oxysterols on parasite development. Mol Biochem Parasitol 2018; 223:1-12. [PMID: 29909067 DOI: 10.1016/j.molbiopara.2018.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/12/2018] [Accepted: 06/12/2018] [Indexed: 11/16/2022]
Abstract
Obligate intracellular apicomplexan parasites are considered as deficient in cholesterol biosynthesis and scavenge cholesterol from their host cell in a parasite-specific manner. Compared to fast proliferating apicomplexan species producing low numbers of merozoites per host cell, (e. g. Toxoplasma gondii), the macromeront-forming protozoa Eimeria bovis is in extraordinary need for cholesterol for offspring production (≥ 170,000 merozoites I/macromeront). Interestingly, optimized in vitro E. bovis merozoite I production occurs under low foetal calf serum (FCS, 1.2%) supplementation. To analyze the impact of extensive E. bovis proliferation on host cellular sterol metabolism we here compared the sterol profiles of E. bovis-infected primary endothelial host cells grown under optimized (1.2% FCS) and non-optimized (10% FCS) cell culture conditions. Therefore, several sterols indicating endogenous de novo cholesterol synthesis, cholesterol conversion and sterol uptake (phytosterols) were analyzed via GC-MS-based approaches. Overall, significantly enhanced levels of phytosterols were detected in both FCS conditions indicating infection-triggered sterol uptake from extracellular sources as a major pathway of sterol acquisition. Interestingly, a simultaneous induction of endogenous cholesterol synthesis based on increased levels of distinct cholesterol precursors was only observed in case of optimized parasite proliferation indicating a parasite proliferation-dependent effect. Considering side-chain oxysterols, 25 hydroxycholesterol levels were selectively found increased in E. bovis-infected host cells, while 24 hydroxycholesterol and 27 hydroxycholesterol contents were not significantly altered by infection. Exogenous treatments with 25 hydroxycholesterol, 27 hydroxycholesterol, and 7 ketocholesterol revealed significant adverse effects on E. bovis intracellular development. Thus, the number and size of developing macromeronts and merozoite I production was significantly reduced indicating that these oxysterols bear direct or indirect antiparasitic properties. Overall, the current data indicate parasite-driven changes in the host cellular sterol profile reflecting the huge demand of E. bovis for cholesterol during macromeront formation and its versatility in the acquisition of cholesterol sources.
Collapse
Affiliation(s)
- A Taubert
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| | - L M R Silva
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| | - Z D Velásquez
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| | - C Larrazabal
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| | - D Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Clinics of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany.
| | - C Hermosilla
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| |
Collapse
|
21
|
Vallochi AL, Teixeira L, Oliveira KDS, Maya-Monteiro CM, Bozza PT. Lipid Droplet, a Key Player in Host-Parasite Interactions. Front Immunol 2018; 9:1022. [PMID: 29875768 PMCID: PMC5974170 DOI: 10.3389/fimmu.2018.01022] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/24/2018] [Indexed: 12/18/2022] Open
Abstract
Lipid droplets (lipid bodies, LDs) are dynamic organelles that have important roles in regulating lipid metabolism, energy homeostasis, cell signaling, membrane trafficking, and inflammation. LD biogenesis, composition, and functions are highly regulated and may vary according to the stimuli, cell type, activation state, and inflammatory environment. Increased cytoplasmic LDs are frequently observed in leukocytes and other cells in a number of infectious diseases. Accumulating evidence reveals LDs participation in fundamental mechanisms of host-pathogen interactions, including cell signaling and immunity. LDs are sources of eicosanoid production, and may participate in different aspects of innate signaling and antigen presentation. In addition, intracellular pathogens evolved mechanisms to subvert host metabolism and may use host LDs, as ways of immune evasion and nutrients source. Here, we review mechanisms of LDs biogenesis and their contributions to the infection progress, and discuss the latest discoveries on mechanisms and pathways involving LDs roles as regulators of the immune response to protozoan infection.
Collapse
Affiliation(s)
- Adriana Lima Vallochi
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | | | | | - Patricia T. Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Basyoni MMA, Fouad SA, Amer MF, Amer AF, Ismail DI. Atorvastatin: In-Vivo Synergy with Metronidazole as Anti- Blastocystis Therapy. THE KOREAN JOURNAL OF PARASITOLOGY 2018; 56:105-112. [PMID: 29742864 PMCID: PMC5976012 DOI: 10.3347/kjp.2018.56.2.105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/04/2018] [Accepted: 04/08/2018] [Indexed: 11/23/2022]
Abstract
Blastocystis is an enteric Straminopile in tropical, subtropical and developing countries. Metronidazole has been a chemotheraputic for blastocystosis. Failures in its regimens were reported and necessitate new studies searching for alternative therapeutic agents. Aim of current study is to investigate potential effects of Atorvastatin (AVA) compared to the conventional chemotherapeutic MTZ in experimentally Blastocystis-infected mice. Anti-Blastocystis efficacy of AVA was evaluated parasitologically, histopathologically and by transmission electron microscopy using MTZ (10 mg/kg) as a control. Therapeutic efficacy of AVA was apparently dose-dependent. Regimens of AVA (20 and 40 mg/kg) proved effective against Blastocystis infections with high reduction in Blastocystis shedding (93.4–97.9%) compared to MTZ (79.3%). The highest reductions (98.1% and 99.4%) were recorded in groups of combination treatments AVA 20–40 mg/kg and MTZ 10 mg/kg. Blastocystis was nearly eradicated by the 20th day post infection. Genotype analysis revealed that genotype I was most susceptible, genotype III was less. Histopathologic and ultrastructural studies revealed apoptotic changes in Blastocystis and significant improvement of intestinal histopathological changes more remarkable in combinational therapy groups. Thus, the present study offers AVA as a potential candidate for Blastocystis therapy combined with MTZ.
Collapse
Affiliation(s)
- Maha M A Basyoni
- Medical Parasitology Department, Faculty of Medicine, Cairo University, Egypt
| | - Shawky A Fouad
- Internal Medicine Department, Faculty of Medicine, Cairo University, Egypt
| | - Marwa F Amer
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Egypt
| | | | | |
Collapse
|
23
|
Ren J, Franklin ET, Xia Y. Uncovering Structural Diversity of Unsaturated Fatty Acyls in Cholesteryl Esters via Photochemical Reaction and Tandem Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2017; 28:1432-1441. [PMID: 28417305 PMCID: PMC5483228 DOI: 10.1007/s13361-017-1639-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 05/09/2023]
Abstract
Mass spectrometry analysis of cholesteryl esters (CEs) faces several challenges, with one of them being the determination of the carbon-carbon double bond (C=C) locations within unsaturated fatty acyl chains. Paternὸ-Büchi (PB) reaction, a photochemical reaction based on the addition of acetone to C=C, is capable of C=C location determination when coupled with tandem mass spectrometry (MS/MS). In this study, the PB reaction conditions were tailored for CEs and subsequent nanoelectrospray ionization (nanoESI). A solvent system containing acetone/methanol/dichloromethane/water (40/30/20/10, volume ratios) and 100 μM LiOH was determined to be optimal, resulting in reasonable PB reaction yield (~30%) and good ionization efficiency (forming lithium adduct of CEs). Collision-induced dissociation (CID) of the PB reaction products produced characteristic fragment ions of CE together with those modified by the PB reactions, such as lithiated fatty acyl ([FA + Li]+) and its PB product ([FA - PB + Li]+). MS3 CID of [FA - PB + Li]+ led to abundant C=C diagnostic ion formation, which was used for C=C location determination and isomer quantitation. A PB-MS3 CID approach was developed and applied for CE analysis from human plasma. A series of unsaturated CEs was identified with specific C=C locations within fatty acyl chains. Absolute quantitation for each CE species was achieved including coexisting C=C location isomers, such as Δ9 and Δ11 isomers of CE 18:1 and ω-6 and ω-3 isomers of CE 18:3. These results show that PB-MS/MS is useful in uncovering structural diversity of CEs due to unsaturation in fatty acyls, which is often undetected from current lipid analysis approach. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Jia Ren
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907-2084, USA
| | - Elissia T Franklin
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907-2084, USA
| | - Yu Xia
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907-2084, USA.
| |
Collapse
|
24
|
Nolan SJ, Romano JD, Coppens I. Host lipid droplets: An important source of lipids salvaged by the intracellular parasite Toxoplasma gondii. PLoS Pathog 2017; 13:e1006362. [PMID: 28570716 PMCID: PMC5469497 DOI: 10.1371/journal.ppat.1006362] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 06/13/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022] Open
Abstract
Toxoplasma is an obligate intracellular parasite that replicates in mammalian cells within a parasitophorous vacuole (PV) that does not fuse with any host organelles. One mechanism developed by the parasite for nutrient acquisition is the attraction of host organelles to the PV. Here, we examined the exploitation of host lipid droplets (LD), ubiquitous fat storage organelles, by Toxoplasma. We show that Toxoplasma replication is reduced in host cells that are depleted of LD, or impaired in TAG lipolysis or fatty acid catabolism. In infected cells, the number of host LD and the expression of host LD-associated genes (ADRP, DGAT2), progressively increase until the onset of parasite replication. Throughout infection, the PV are surrounded by host LD. Toxoplasma is capable of accessing lipids stored in host LD and incorporates these lipids into its own membranes and LD. Exogenous addition of oleic acid stimulates LD biogenesis in the host cell and results in the overaccumulation of neutral lipids in very large LD inside the parasite. To access LD-derived lipids, Toxoplasma intercepts and internalizes within the PV host LD, some of which remaining associated with Rab7, which become wrapped by an intravacuolar network of membranes (IVN). Mutant parasites impaired in IVN formation display diminished capacity of lipid uptake from host LD. Moreover, parasites lacking an IVN-localized phospholipase A2 are less proficient in salvaging lipids from host LD in the PV, suggesting a major contribution of the IVN for host LD processing in the PV and, thus lipid content release. Interestingly, gavage of parasites with lipids unveils, for the first time, the presence in Toxoplasma of endocytic-like structures containing lipidic material originating from the PV lumen. This study highlights the reliance of Toxoplasma on host LD for its intracellular development and the parasite’s capability in scavenging neutral lipids from host LD. Toxoplasma is an obligate intracellular pathogen that multiplies in mammalian cells within a specialized compartment, named the parasitophorous vacuole (PV). While the vacuole does not fuse with host organelles, the parasite scavenges nutrients, including lipids, from these compartments. Present in all mammalian cells, lipid droplets (LD) are dynamic structures that store neutral lipids. Whether Toxoplasma targets host LD for their nutritional content remains to be investigated. We demonstrate that the parasite relies on host LD lipids and their lipolytic enzymatic activities to grow. Toxoplasma salvages lipids from host LD, which surround the PV and, at least partially, accesses these lipids by intercepting and engulfing within the PV host Rab7-associated LD. In the PV lumen, a parasite lipase releases lipids from host LD, thus making them available to the parasite. Exogenous addition of fatty acids stimulates host LD biogenesis and results in the accumulation of enlarged LD containing neutral lipids in Toxoplasma. This study highlights the ability of Toxoplasma to scavenge and store lipids from host LD. Interestingly, exposure of Toxoplasma to excess lipids reveals, for the first time, coated invaginations of the parasite’s plasma membrane and cytoplasmic vesicles containing lipids originating from the PV lumen, potentially involved in endocytosis.
Collapse
Affiliation(s)
- Sabrina J. Nolan
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Julia D. Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
25
|
Lipid Body Organelles within the Parasite Trypanosoma cruzi: A Role for Intracellular Arachidonic Acid Metabolism. PLoS One 2016; 11:e0160433. [PMID: 27490663 PMCID: PMC4973985 DOI: 10.1371/journal.pone.0160433] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 07/19/2016] [Indexed: 12/19/2022] Open
Abstract
Most eukaryotic cells contain varying amounts of cytosolic lipidic inclusions termed lipid bodies (LBs) or lipid droplets (LDs). In mammalian cells, such as macrophages, these lipid-rich organelles are formed in response to host-pathogen interaction during infectious diseases and are sites for biosynthesis of arachidonic acid (AA)-derived inflammatory mediators (eicosanoids). Less clear are the functions of LBs in pathogenic lower eukaryotes. In this study, we demonstrated that LBs, visualized by light microscopy with different probes and transmission electron microscopy (TEM), are produced in trypomastigote forms of the parasite Trypanosoma cruzi, the causal agent of Chagas' disease, after both host interaction and exogenous AA stimulation. Quantitative TEM revealed that LBs from amastigotes, the intracellular forms of the parasite, growing in vivo have increased size and electron-density compared to LBs from amastigotes living in vitro. AA-stimulated trypomastigotes released high amounts of prostaglandin E2 (PGE2) and showed PGE2 synthase expression. Raman spectroscopy demonstrated increased unsaturated lipid content and AA incorporation in stimulated parasites. Moreover, both Raman and MALDI mass spectroscopy revealed increased AA content in LBs purified from AA-stimulated parasites compared to LBs from unstimulated group. By using a specific technique for eicosanoid detection, we immunolocalized PGE2 within LBs from AA-stimulated trypomastigotes. Altogether, our findings demonstrate that LBs from the parasite Trypanosoma cruzi are not just lipid storage inclusions but dynamic organelles, able to respond to host interaction and inflammatory events and involved in the AA metabolism. Acting as sources of PGE2, a potent immunomodulatory lipid mediator that inhibits many aspects of innate and adaptive immunity, newly-formed parasite LBs may be implicated with the pathogen survival in its host.
Collapse
|
26
|
In Vivo Biotinylation of the Toxoplasma Parasitophorous Vacuole Reveals Novel Dense Granule Proteins Important for Parasite Growth and Pathogenesis. mBio 2016; 7:mBio.00808-16. [PMID: 27486190 PMCID: PMC4981711 DOI: 10.1128/mbio.00808-16] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite that invades host cells and replicates within a unique parasitophorous vacuole. To maintain this intracellular niche, the parasite secretes an array of dense granule proteins (GRAs) into the nascent parasitophorous vacuole. These GRAs are believed to play key roles in vacuolar remodeling, nutrient uptake, and immune evasion while the parasite is replicating within the host cell. Despite the central role of GRAs in the Toxoplasma life cycle, only a subset of these proteins have been identified, and many of their roles have not been fully elucidated. In this report, we utilize the promiscuous biotin ligase BirA* to biotinylate GRA proteins secreted into the vacuole and then identify those proteins by affinity purification and mass spectrometry. Using GRA-BirA* fusion proteins as bait, we have identified a large number of known and candidate GRAs and verified localization of 13 novel GRA proteins by endogenous gene tagging. We proceeded to functionally characterize three related GRAs from this group (GRA38, GRA39, and GRA40) by gene knockout. While Δgra38 and Δgra40 parasites showed no altered phenotype, disruption of GRA39 results in slow-growing parasites that contain striking lipid deposits in the parasitophorous vacuole, suggesting a role in lipid regulation that is important for parasite growth. In addition, parasites lacking GRA39 showed dramatically reduced virulence and a lower tissue cyst burden in vivo. Together, the findings from this work reveal a partial vacuolar proteome of T. gondii and identify a novel GRA that plays a key role in parasite replication and pathogenesis. Most intracellular pathogens reside inside a membrane-bound vacuole within their host cell that is extensively modified by the pathogen to optimize intracellular growth and avoid host defenses. In Toxoplasma, this vacuole is modified by a host of secretory GRA proteins, many of which remain unidentified. Here we demonstrate that in vivo biotinylation of proximal and interacting proteins using the promiscuous biotin ligase BirA* is a powerful approach to rapidly identify vacuolar GRA proteins. We further demonstrate that one factor identified by this approach, GRA39, plays an important role in the ability of the parasite to replicate within its host cell and cause disease.
Collapse
|
27
|
Fernández C, Jaimes J, Ortiz MC, Ramírez JD. Host and Toxoplasma gondii genetic and non-genetic factors influencing the development of ocular toxoplasmosis: A systematic review. INFECTION GENETICS AND EVOLUTION 2016; 44:199-209. [PMID: 27389360 DOI: 10.1016/j.meegid.2016.06.053] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022]
Abstract
Toxoplasmosis is a cosmopolitan infection caused by the apicomplexan parasite Toxoplasma gondii. This infectious disease is widely distributed across the world where cats play an important role in its spread. The symptomatology caused by this parasite is diverse but the ocular affectation emerges as the most important clinical phenotype. Therefore, we conducted a systematic review of the current knowledge of ocular toxoplasmosis from the genetic diversity of the pathogen towards the treatment available for this infection. This review represents an update to the scientific community regarding the genetic diversity of the parasite, the genetic factors of the host, the molecular pathogenesis and its association with disease, the available diagnostic tools and the available treatment of patients undergoing ocular toxoplamosis. This review will be an update for the scientific community in order to encourage researchers to deploy cutting-edge investigation across this field.
Collapse
Affiliation(s)
- Carolina Fernández
- Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia; Grupo de Investigaciones Microbiológicas - UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Jesús Jaimes
- Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia; Grupo de Investigaciones Microbiológicas - UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - María Camila Ortiz
- Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia; Grupo de Investigaciones Microbiológicas - UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Grupo de Investigaciones Microbiológicas - UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
28
|
Toledo DAM, D'Avila H, Melo RCN. Host Lipid Bodies as Platforms for Intracellular Survival of Protozoan Parasites. Front Immunol 2016; 7:174. [PMID: 27199996 PMCID: PMC4853369 DOI: 10.3389/fimmu.2016.00174] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 04/19/2016] [Indexed: 12/31/2022] Open
Abstract
Pathogens induce several changes in the host cell signaling and trafficking mechanisms in order to evade and manipulate the immune response. One prominent pathogen-mediated change is the formation of lipid-rich organelles, termed lipid bodies (LBs) or lipid droplets, in the host cell cytoplasm. Protozoan parasites, which contribute expressively to the burden of infectious diseases worldwide, are able to induce LB genesis in non-immune and immune cells, mainly macrophages, key players in the initial resistance to the infection. Under host–parasite interaction, LBs not only accumulate in the host cytoplasm but also relocate around and move into parasitophorous vacuoles. There is increasing evidence that protozoan parasites may target host-derived LBs either for gaining nutrients or for escaping the host immune response. Newly formed, parasite-induced LBs may serve as lipid sources for parasite growth and also produce inflammatory mediators that potentially act in the host immune response deactivation. In this mini review, we summarize current knowledge on the formation and role of host LBs as sites exploited by intracellular protozoan parasites as a strategy to maintain their own survival.
Collapse
Affiliation(s)
- Daniel A M Toledo
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences (ICB), Federal University of Juiz de Fora (UFJF) , Juiz de Fora, Minas Gerais , Brazil
| | - Heloísa D'Avila
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences (ICB), Federal University of Juiz de Fora (UFJF) , Juiz de Fora, Minas Gerais , Brazil
| | - Rossana C N Melo
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences (ICB), Federal University of Juiz de Fora (UFJF) , Juiz de Fora, Minas Gerais , Brazil
| |
Collapse
|
29
|
Li Y, Shah-Simpson S, Okrah K, Belew AT, Choi J, Caradonna KL, Padmanabhan P, Ndegwa DM, Temanni MR, Corrada Bravo H, El-Sayed NM, Burleigh BA. Transcriptome Remodeling in Trypanosoma cruzi and Human Cells during Intracellular Infection. PLoS Pathog 2016; 12:e1005511. [PMID: 27046031 PMCID: PMC4821583 DOI: 10.1371/journal.ppat.1005511] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 02/28/2016] [Indexed: 01/22/2023] Open
Abstract
Intracellular colonization and persistent infection by the kinetoplastid protozoan parasite, Trypanosoma cruzi, underlie the pathogenesis of human Chagas disease. To obtain global insights into the T. cruzi infective process, transcriptome dynamics were simultaneously captured in the parasite and host cells in an infection time course of human fibroblasts. Extensive remodeling of the T. cruzi transcriptome was observed during the early establishment of intracellular infection, coincident with a major developmental transition in the parasite. Contrasting this early response, few additional changes in steady state mRNA levels were detected once mature T. cruzi amastigotes were formed. Our findings suggest that transcriptome remodeling is required to establish a modified template to guide developmental transitions in the parasite, whereas homeostatic functions are regulated independently of transcriptomic changes, similar to that reported in related trypanosomatids. Despite complex mechanisms for regulation of phenotypic expression in T. cruzi, transcriptomic signatures derived from distinct developmental stages mirror known or projected characteristics of T. cruzi biology. Focusing on energy metabolism, we were able to validate predictions forecast in the mRNA expression profiles. We demonstrate measurable differences in the bioenergetic properties of the different mammalian-infective stages of T. cruzi and present additional findings that underscore the importance of mitochondrial electron transport in T. cruzi amastigote growth and survival. Consequences of T. cruzi colonization for the host include dynamic expression of immune response genes and cell cycle regulators with upregulation of host cholesterol and lipid synthesis pathways, which may serve to fuel intracellular T. cruzi growth. Thus, in addition to the biological inferences gained from gene ontology and functional enrichment analysis of differentially expressed genes in parasite and host, our comprehensive, high resolution transcriptomic dataset provides a substantially more detailed interpretation of T. cruzi infection biology and offers a basis for future drug and vaccine discovery efforts. In-depth knowledge of the functional processes governing host colonization and transmission of pathogenic microorganisms is essential for the advancement of effective intervention strategies. This study focuses on Trypanosoma cruzi, the vector-borne protozoan parasite responsible for human Chagas disease and the leading cause of infectious myocarditis worldwide. To gain global insights into the biology of this parasite and its interaction with mammalian host cells, we have exploited a deep-sequencing approach to generate comprehensive, high-resolution transcriptomic maps for mammalian-infective stages of T. cruzi with the simultaneous interrogation of the human host cell transcriptome across an infection time course. We demonstrate that the establishment of intracellular T. cruzi infection in mammalian host cells is accompanied by extensive remodeling of the parasite and host cell transcriptomes. Despite the lack of transcriptional control mechanisms in trypanosomatids, our analyses identified functionally-enriched processes within sets of developmentally-regulated transcripts in T. cruzi that align with known or predicted biological features of the parasite. The novel insights into the biology of intracellular T. cruzi infection and the regulation of amastigote development gained in this study establish a unique foundation for functional network analyses that will be instrumental in providing functional links between parasite dependencies and host functional pathways that have the potential to be exploited for intervention.
Collapse
Affiliation(s)
- Yuan Li
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Sheena Shah-Simpson
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Kwame Okrah
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - A Trey Belew
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Jungmin Choi
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Kacey L Caradonna
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Prasad Padmanabhan
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - David M Ndegwa
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - M Ramzi Temanni
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Héctor Corrada Bravo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Najib M El-Sayed
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America.,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Barbara A Burleigh
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
30
|
Pszenny V, Ehrenman K, Romano JD, Kennard A, Schultz A, Roos DS, Grigg ME, Carruthers VB, Coppens I. A Lipolytic Lecithin:Cholesterol Acyltransferase Secreted by Toxoplasma Facilitates Parasite Replication and Egress. J Biol Chem 2015; 291:3725-46. [PMID: 26694607 DOI: 10.1074/jbc.m115.671974] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Indexed: 11/06/2022] Open
Abstract
The protozoan parasite Toxoplasma gondii develops within a parasitophorous vacuole (PV) in mammalian cells, where it scavenges cholesterol. When cholesterol is present in excess in its environment, the parasite expulses this lipid into the PV or esterifies it for storage in lipid bodies. Here, we characterized a unique T. gondii homologue of mammalian lecithin:cholesterol acyltransferase (LCAT), a key enzyme that produces cholesteryl esters via transfer of acyl groups from phospholipids to the 3-OH of free cholesterol, leading to the removal of excess cholesterol from tissues. TgLCAT contains a motif characteristic of serine lipases "AHSLG" and the catalytic triad consisting of serine, aspartate, and histidine (SDH) from LCAT enzymes. TgLCAT is secreted by the parasite, but unlike other LCAT enzymes it is cleaved into two proteolytic fragments that share the residues of the catalytic triad and need to be reassembled to reconstitute enzymatic activity. TgLCAT uses phosphatidylcholine as substrate to form lysophosphatidylcholine that has the potential to disrupt membranes. The released fatty acid is transferred to cholesterol, but with a lower transesterification activity than mammalian LCAT. TgLCAT is stored in a subpopulation of dense granule secretory organelles, and following secretion, it localizes to the PV and parasite plasma membrane. LCAT-null parasites have impaired growth in vitro, reduced virulence in animals, and exhibit delays in egress from host cells. Parasites overexpressing LCAT show increased virulence and faster egress. These observations demonstrate that TgLCAT influences the outcome of an infection, presumably by facilitating replication and egress depending on the developmental stage of the parasite.
Collapse
Affiliation(s)
- Viviana Pszenny
- From the Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, the Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Karen Ehrenman
- From the Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205
| | - Julia D Romano
- From the Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205
| | - Andrea Kennard
- the Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Aric Schultz
- the Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, and
| | - David S Roos
- the Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Michael E Grigg
- the Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Vern B Carruthers
- the Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, and
| | - Isabelle Coppens
- From the Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205,
| |
Collapse
|
31
|
Hamid PH, Hirzmann J, Kerner K, Gimpl G, Lochnit G, Hermosilla CR, Taubert A. Eimeria bovis infection modulates endothelial host cell cholesterol metabolism for successful replication. Vet Res 2015; 46:100. [PMID: 26395984 PMCID: PMC4579583 DOI: 10.1186/s13567-015-0230-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/04/2015] [Indexed: 11/10/2022] Open
Abstract
During first merogony Eimeria bovis forms large macromeronts in endothelial host cells containing >120 000 merozoites I. During multiplication, large amounts of cholesterol are indispensable for the enormous offspring membrane production. Cholesterol auxotrophy was proven for other apicomplexan parasites. Consequently they scavenge cholesterol from their host cell apparently in a parasite-specific manner. We here analyzed the influence of E. bovis infection on endothelial host cell cholesterol metabolism and found considerable differences to other coccidian parasites. Overall, free cholesterol significantly accumulated in E. bovis infected host cells. Furthermore, a striking increase of lipid droplet formation was observed within immature macromeronts. Artificial host cell lipid droplet enrichment significantly improved E. bovis merozoite I production confirming the key role of lipid droplet contents for optimal parasite proliferation. The transcription of several genes being involved in both, cholesterol de novo biosynthesis and low density lipoprotein-(LDL) mediated uptake, was significantly up-regulated at a time in infected cells suggesting a simultaneous exploitation of these two cholesterol acquisition pathways. E. bovis scavenges LDL-derived cholesterol apparently through significantly increased levels of surface LDL receptor abundance and LDL binding to infected cells. Consequently, LDL supplementation significantly improved parasite replication. The up-regulation of the oxidized LDL receptor 1 furthermore identified this scavenger receptor as a key molecule in parasite-triggered LDL uptake. Moreover, cellular cholesterol processing was altered in infected cells as indicated by up-regulation of cholesterol-25-hydroxylase and sterol O-acyltransferase. Overall, these results show that E. bovis considerably exploits the host cell cholesterol metabolism to guarantee its massive intracellular growth and replication.
Collapse
Affiliation(s)
- Penny H Hamid
- Institute of Parasitology, Biomedical Research Centre, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany.
| | - Joerg Hirzmann
- Institute of Parasitology, Biomedical Research Centre, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany.
| | - Katharina Kerner
- Institute for Hygiene and Infectious Diseases of Animals, Justus LiebigUniversity Giessen, Frankfurter Str. 85-89, D-35392, Giessen, Germany.
| | - Gerald Gimpl
- Institute of Pharmacy and Biochemistry, Department of Biochemistry, Johann-Joachim-Becherweg 30, D-55099, Mainz, Germany.
| | - Guenter Lochnit
- Institute of Biochemistry, Justus Liebig University Giessen, Friedrichstr. 24, D-35392, Giessen, Germany.
| | - Carlos R Hermosilla
- Institute of Parasitology, Biomedical Research Centre, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany.
| | - Anja Taubert
- Institute of Parasitology, Biomedical Research Centre, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany.
| |
Collapse
|
32
|
Khadka M, Salem M, Leblond JD. Sterol Composition and Biosynthetic Genes of Vitrella brassicaformis
, a Recently Discovered Chromerid: Comparison to Chromera velia
and Phylogenetic Relationship with Apicomplexan Parasites. J Eukaryot Microbiol 2015; 62:786-98. [DOI: 10.1111/jeu.12237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 11/29/2022]
Affiliation(s)
- Manoj Khadka
- Department of Biology; Middle Tennessee State University; PO Box 60 Murfreesboro Tennessee 37132
| | - Mohamed Salem
- Department of Biology; Middle Tennessee State University; PO Box 60 Murfreesboro Tennessee 37132
| | - Jeffrey D. Leblond
- Department of Biology; Middle Tennessee State University; PO Box 60 Murfreesboro Tennessee 37132
- Ecology and Evolution Group; Middle Tennessee State University; PO Box 60 Murfreesboro Tennessee 37132
| |
Collapse
|
33
|
Pereira MG, Visbal G, Salgado LT, Vidal JC, Godinho JLP, De Cicco NNT, Atella GC, de Souza W, Cunha-e-Silva N. Trypanosoma cruzi Epimastigotes Are Able to Manage Internal Cholesterol Levels under Nutritional Lipid Stress Conditions. PLoS One 2015; 10:e0128949. [PMID: 26068009 PMCID: PMC4466137 DOI: 10.1371/journal.pone.0128949] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 05/02/2015] [Indexed: 11/19/2022] Open
Abstract
Trypanosoma cruzi epimastigotes store high amounts of cholesterol and cholesteryl esters in reservosomes. These unique organelles are responsible for cellular digestion by providing substrates for homeostasis and parasite differentiation. Here we demonstrate that under nutritional lipid stress, epimastigotes preferentially mobilized reservosome lipid stocks, instead of lipid bodies, leading to the consumption of parasite cholesterol reservoirs and production of ergosterol. Starved epimastigotes acquired more LDL-NBD-cholesterol by endocytosis and distributed the exogenous cholesterol to their membranes faster than control parasites. Moreover, the parasites were able to manage internal cholesterol levels, alternating between consumption and accumulation. With normal lipid availability, parasites esterified cholesterol exhibiting an ACAT-like activity that was sensitive to Avasimibe in a dose-dependent manner. This result also implies that exogenous cholesterol has a role in lipid reservoirs in epimastigotes.
Collapse
Affiliation(s)
- Miria Gomes Pereira
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Laboratório de Bioquímica de Lipídios e Lipoproteínas, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Gonzalo Visbal
- Instituto Nacional de Metrologia, Qualidade e Tecnologia—INMETRO, Rio de Janeiro, Brasil
| | - Leonardo T. Salgado
- Instituto de Pesquisas Jardim Botânico do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Juliana Cunha Vidal
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Joseane L. P. Godinho
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Nuccia N. T. De Cicco
- Laboratório de Bioquímica de Lipídios e Lipoproteínas, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Geórgia C. Atella
- Laboratório de Bioquímica de Lipídios e Lipoproteínas, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Metrologia, Qualidade e Tecnologia—INMETRO, Rio de Janeiro, Brasil
| | - Narcisa Cunha-e-Silva
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- * E-mail:
| |
Collapse
|
34
|
Neospora caninum Recruits Host Cell Structures to Its Parasitophorous Vacuole and Salvages Lipids from Organelles. EUKARYOTIC CELL 2015; 14:454-73. [PMID: 25750213 DOI: 10.1128/ec.00262-14] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/24/2015] [Indexed: 12/21/2022]
Abstract
Toxoplasma gondii and Neospora caninum, which cause the diseases toxoplasmosis and neosporosis, respectively, are two closely related apicomplexan parasites. They have similar heteroxenous life cycles and conserved genomes and share many metabolic features. Despite these similarities, T. gondii and N. caninum differ in their transmission strategies and zoonotic potential. Comparative analyses of the two parasites are important to identify the unique biological features that underlie the basis of host preference and pathogenicity. T. gondii and N. caninum are obligate intravacuolar parasites; in contrast to T. gondii, events that occur during N. caninum infection remain largely uncharacterized. We examined the capability of N. caninum (Liverpool isolate) to interact with host organelles and scavenge nutrients in comparison to that of T. gondii (RH strain). N. caninum reorganizes the host microtubular cytoskeleton and attracts endoplasmic reticulum (ER), mitochondria, lysosomes, multivesicular bodies, and Golgi vesicles to its vacuole though with some notable differences from T. gondii. For example, the host ER gathers around the N. caninum parasitophorous vacuole (PV) but does not physically associate with the vacuolar membrane; the host Golgi apparatus surrounds the N. caninum PV but does not fragment into ministacks. N. caninum relies on plasma lipoproteins and scavenges cholesterol from NPC1-containing endocytic organelles. This parasite salvages sphingolipids from host Golgi Rab14 vesicles that it sequesters into its vacuole. Our data highlight a remarkable degree of conservation in the intracellular infection program of N. caninum and T. gondii. The minor differences between the two parasites related to the recruitment and rearrangement of host organelles around their vacuoles likely reflect divergent evolutionary paths.
Collapse
|
35
|
Soupene E, Wang D, Kuypers FA. Remodeling of host phosphatidylcholine by Chlamydia acyltransferase is regulated by acyl-CoA binding protein ACBD6 associated with lipid droplets. Microbiologyopen 2015; 4:235-251. [PMID: 25604091 PMCID: PMC4398506 DOI: 10.1002/mbo3.234] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/25/2014] [Accepted: 12/01/2014] [Indexed: 12/25/2022] Open
Abstract
The bacterial human pathogen Chlamydia trachomatis invades cells as an infectious elementary body (EB). The EB is internalized into a vacuole that is hidden from the host defense mechanism, and is modified to sustain the development of the replicative reticulate body (RB). Inside this parasitophorous compartment, called the inclusion, the pathogen survives supported by an active exchange of nutrients and proteins with the host cell. We show that host lipids are scavenged and modified into bacterial-specific lipids by the action of a shared human-bacterial acylation mechanism. The bacterial acylating enzymes for the essential lipids 1-acyl-sn-glycerol 3-phosphate and 1-acyl-sn-phosphatidylcholine were identified as CT453 and CT775, respectively. Bacterial CT775 was found to be associated with lipid droplets (LDs). During the development of C. trachomatis, the human acyl-CoA carrier hACBD6 was recruited to cytosolic LDs and translocated into the inclusion. hACBD6 protein modulated the activity of CT775 in an acyl-CoA dependent fashion and sustained the activity of the bacterial acyltransferase by buffering the concentration of acyl-CoAs. We propose that disruption of the binding activity of the acyl-CoA carrier might represent a new drug-target to prevent growth of C. trachomatis.
Collapse
Affiliation(s)
- Eric Soupene
- Children's Hospital Oakland Research Institute, Oakland, California USA
| | - Derek Wang
- Children's Hospital Oakland Research Institute, Oakland, California USA
| | - Frans A Kuypers
- Children's Hospital Oakland Research Institute, Oakland, California USA
| |
Collapse
|
36
|
Differential inhibition of host cell cholesterol de novo biosynthesis and processing abrogates Eimeria bovis intracellular development. Parasitol Res 2014; 113:4165-76. [DOI: 10.1007/s00436-014-4092-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/22/2014] [Indexed: 10/24/2022]
|
37
|
Mota LAM, Roberto Neto J, Monteiro VG, Lobato CSS, Oliveira MAD, Cunha MD, D'Ávila H, Seabra SH, Bozza PT, DaMatta RA. Culture of mouse peritoneal macrophages with mouse serum induces lipid bodies that associate with the parasitophorous vacuole and decrease their microbicidal capacity against Toxoplasma gondii. Mem Inst Oswaldo Cruz 2014; 109:767-74. [PMID: 25317704 PMCID: PMC4238769 DOI: 10.1590/0074-0276140119] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 06/26/2014] [Indexed: 11/22/2022] Open
Abstract
Lipid bodies [lipid droplets (LBs)] are lipid-rich organelles involved in lipid metabolism, signalling and inflammation. Recent findings suggest a role for LBs in host response to infection; however, the potential functions of this organelle in Toxoplasma gondii infection and how it alters macrophage microbicidal capacity during infection are not well understood. Here, we investigated the role of host LBs in T. gondii infection in mouse peritoneal macrophages in vitro. Macrophages cultured with mouse serum (MS) had higher numbers of LBs than those cultured in foetal bovine serum and can function as a model to study the role of LBs during intracellular pathogen infection. LBs were found in association with the parasitophorous vacuole, suggesting that T. gondii may benefit from this lipid source. Moreover, increased numbers of macrophage LBs correlated with high prostaglandin E2 (PGE2) production and decreased nitric oxide (NO) synthesis. Accordingly, LB-enriched macrophages cultured with MS were less efficient at controlling T. gondii growth. Treatment of macrophages cultured with MS with indomethacin, an inhibitor of PGE2 production, increased the microbicidal capacity against T. gondii. Collectively, these results suggest that culture with MS caused a decrease in microbicidal activity of macrophages against T. gondii by increasing PGE2 while lowering NO production.
Collapse
Affiliation(s)
- Laura Azeredo Miranda Mota
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - João Roberto Neto
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Verônica Gomes Monteiro
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Caroliny Samary Silva Lobato
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Marco Antonio de Oliveira
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Maura da Cunha
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Heloisa D'Ávila
- Laboratório de Imunofarmacologia, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brasil
| | - Sérgio Henrique Seabra
- Laboratório de Tecnologia em Bioquímica e Microscopia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
| | | | - Renato Augusto DaMatta
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| |
Collapse
|
38
|
Elsheikha HM, Alkurashi M, Kong K, Zhu XQ. Metabolic footprinting of extracellular metabolites of brain endothelium infected with Neospora caninum in vitro. BMC Res Notes 2014; 7:406. [PMID: 24973017 PMCID: PMC4105892 DOI: 10.1186/1756-0500-7-406] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 06/13/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The survival of the intracellular protozoan parasite Neospora caninum depends on its ability to adapt to changing metabolic conditions of the host cell. Thus, defining cellular and metabolic changes in affected target tissues may aid in delineating pathogenetic mechanism. We undertook this study to assess the metabolic response of human brain microvascular endothelial cells (HBMECs) to N. caninum infection in vitro. METHODS HBMECs were exposed to N. caninum infection and the cytotoxic effects of infection were analyzed by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazoliumbromidin (MTT) assay and lactate dehydrogenase (LDH) release assay. Metabolic footprinting of the extracellular metabolites of parasite-infected and non-infected culture supernatant was determined by using targeted (Randox RX Imola clinical chemistry analyser) and unbiased RS (Raman microspectroscopy) approaches. RESULTS The MTT assay did not reveal any cytotoxic effect of N. caninum challenge on host cell viability. Measurement of LDH activity showed that N. caninum significantly induced loss of cell membrane integrity in a time-dependent and dose-dependent manner compared to control cells. Targeted biochemical analysis revealed that beta hydroxybutyrate, pyruvate, ATP, total protein, non-esterified fatty acids, and triglycerides are significantly different in infected cells compared to controls. RS-based footprinting with principal component analysis (PCA) were able to correctly distinguish extracellular metabolites obtained from infected and control cultures, and revealed infection-related spectral signatures at 865 cm-1, 984 cm-1, 1046 cm-1, and 1420 cm-1, which are attributed to variations in the content of lipids and nucleic acids in infected cultures. CONCLUSIONS The changing pattern of extracellular metabolites suggests that HBMECs are target of metabolic alterations in N. caninum infection, which seem to reflect the changing metabolic state of infected cells and constitute a level of information exchange that host and parasite use to coordinate activities.
Collapse
Affiliation(s)
- Hany M Elsheikha
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Sutton Bonington Campus, Leicestershire LE12 5RD, UK.
| | | | | | | |
Collapse
|
39
|
Ihara F, Nishikawa Y. Starvation of low-density lipoprotein-derived cholesterol induces bradyzoite conversion in Toxoplasma gondii. Parasit Vectors 2014; 7:248. [PMID: 24885547 PMCID: PMC4046157 DOI: 10.1186/1756-3305-7-248] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 05/19/2014] [Indexed: 11/10/2022] Open
Abstract
Background Lacking enzymes for sterol synthesis, the intracellular protozoan Toxoplasma gondii scavenges cholesterol from host cells to multiply. T. gondii has a complex life cycle consisting of two asexual stages; the proliferative stage (tachyzoite), and the latent stage characterized by tissue cysts (bradyzoite). In vitro, bradyzoite development can be induced by mimicking host immune response stressors through treatment with IFN-γ, heat shock, nitric oxide, and high pH. However, the extent to which host nutrients contribute to stage conversion in T. gondii is unknown. In this study, we examined the impact of host cholesterol levels on stage conversion in this parasite. Methods Growth of T. gondii tachyzoites (ME49 strain) was investigated in Chinese hamster ovary (CHO) cells using various concentrations of low-density lipoprotein (LDL), oleic acid, or glucose. Squalestatin, which is an inhibitor of squalene synthase and is, therefore, an inhibitor of sterol synthesis, was used to treat the CHO cells. Tachyzoite to bradyzoite conversion rates were analyzed by indirect fluorescent antibody tests. Results Parasite growth was significantly enhanced by addition of exogenous LDL, whereas no such enhancement occurred with oleic acids or glucose. In ME49, growth inhibition from squalestatin treatment was not obvious. Although growth of the RH strain was unaffected by squalestatin in the presence of lipoprotein, in its absence growth of this strain was suppressed. The frequency of BAG1-positive vacuoles in ME49 increased under lipoprotein-free conditions. However, addition of exogenous LDL did not increase tachyzoite to bradyzoite conversion in this strain. Furthermore, treatment with squalestatin did not enhance stage conversion. Conclusion Our results suggest that LDL-derived cholesterol levels play a crucial role in bradyzoite conversion in T. gondii.
Collapse
Affiliation(s)
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro Hokkaido 080-8555, Japan.
| |
Collapse
|
40
|
Araújo-Santos T, Rodríguez NE, Moura-Pontes S, Dixt UG, Abánades DR, Bozza PT, Wilson ME, Borges VM. Role of prostaglandin F2α production in lipid bodies from Leishmania infantum chagasi: insights on virulence. J Infect Dis 2014; 210:1951-61. [PMID: 24850789 DOI: 10.1093/infdis/jiu299] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Lipid bodies (LB; lipid droplets) are cytoplasmic organelles involved in lipid metabolism. Mammalian LBs display an important role in host-pathogen interactions, but the role of parasite LBs in biosynthesis of prostaglandin F2α (PGF2α) has not been investigated. We report herein that LBs increased in abundance during development of Leishmania infantum chagasi to a virulent metacyclic stage, as did the expression of PGF2α synthase (PGFS). The amount of parasite LBs and PGF2α were modulated by exogenous arachidonic acid. During macrophage infection, LBs were restricted to parasites inside the parasitophorous vacuoles (PV). We detected PGF2α receptor (FP) on the Leishmania PV surface. The blockage of FP with AL8810, a selective antagonist, hampered Leishmania infection, whereas the irreversible inhibition of cyclooxygenase with aspirin increased the parasite burden. These data demonstrate novel functions for parasite-derived LBs and PGF2α in the cellular metabolism of Leishmania and its evasion of the host immune response.
Collapse
Affiliation(s)
- Théo Araújo-Santos
- Gonçalo Moniz Research Center, Oswaldo Cruz Foundation (FIOCRUZ) Federal University of Bahia (UFBA), Salvador, Bahia, Brazil University of Iowa and the Iowa City VA Medical Center, Iowa
| | | | - Sara Moura-Pontes
- Gonçalo Moniz Research Center, Oswaldo Cruz Foundation (FIOCRUZ) Federal University of Bahia (UFBA), Salvador, Bahia, Brazil
| | | | - Daniel R Abánades
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | | | - Mary E Wilson
- University of Iowa and the Iowa City VA Medical Center, Iowa
| | - Valéria Matos Borges
- Gonçalo Moniz Research Center, Oswaldo Cruz Foundation (FIOCRUZ) Federal University of Bahia (UFBA), Salvador, Bahia, Brazil Institute for Investigation in Immunology, iii-INCT (National Institute of Science and Technology), São Paulo, Brazil
| |
Collapse
|
41
|
Exploitation of auxotrophies and metabolic defects in Toxoplasma as therapeutic approaches. Int J Parasitol 2014; 44:109-20. [DOI: 10.1016/j.ijpara.2013.09.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 09/22/2013] [Accepted: 09/22/2013] [Indexed: 12/30/2022]
|
42
|
Targeting lipid biosynthesis and salvage in apicomplexan parasites for improved chemotherapies. Nat Rev Microbiol 2013; 11:823-35. [DOI: 10.1038/nrmicro3139] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
43
|
Rub A, Arish M, Husain SA, Ahmed N, Akhter Y. Host-lipidome as a potential target of protozoan parasites. Microbes Infect 2013; 15:649-60. [PMID: 23811020 DOI: 10.1016/j.micinf.2013.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 04/13/2013] [Accepted: 06/18/2013] [Indexed: 12/24/2022]
Abstract
Host-lipidome caters parasite interaction by acting as first line of recognition, attachment on the cell surface, intracellular trafficking, and survival of the parasite inside the host cell. Here, we summarize how protozoan parasites exploit host-lipidome by suppressing, augmenting, engulfing, remodeling and metabolizing lipids to achieve successful parasitism inside the host.
Collapse
Affiliation(s)
- Abdur Rub
- Infection and Immunity Lab, Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India.
| | | | | | | | | |
Collapse
|
44
|
Lecoeur H, Giraud E, Prévost MC, Milon G, Lang T. Reprogramming neutral lipid metabolism in mouse dendritic leucocytes hosting live Leishmania amazonensis amastigotes. PLoS Negl Trop Dis 2013; 7:e2276. [PMID: 23785538 PMCID: PMC3681733 DOI: 10.1371/journal.pntd.0002276] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 05/06/2013] [Indexed: 11/18/2022] Open
Abstract
Background After loading with live Leishmania (L) amazonensis amastigotes, mouse myeloid dendritic leucocytes/DLs are known to undergo reprogramming of their immune functions. In the study reported here, we investigated whether the presence of live L. amazonensis amastigotes in mouse bone marrow-derived DLs is able to trigger re-programming of DL lipid, and particularly neutral lipid metabolism. Methodology/Principal Findings Affymetrix-based transcriptional profiles were determined in C57BL/6 and DBA/2 mouse bone marrow-derived DLs that had been sorted from cultures exposed or not to live L. amazonensis amastigotes. This showed that live amastigote-hosting DLs exhibited a coordinated increase in: (i) long-chain fatty acids (LCFA) and cholesterol uptake/transport, (ii) LCFA and cholesterol (re)-esterification to triacyl-sn-glycerol (TAG) and cholesteryl esters (CE), respectively. As these neutral lipids are known to make up the lipid body (LB) core, oleic acid was added to DL cultures and LB accumulation was compared in live amastigote-hosting versus amastigote-free DLs by epi-fluorescence and transmission electron microscopy. This showed that LBs were both significantly larger and more numerous in live amastigote-hosting mouse dendritic leucocytes. Moreover, many of the larger LB showed intimate contact with the membrane of the parasitophorous vacuoles hosting the live L. amazonensis amastigotes. Conclusions/Significance As leucocyte LBs are known to be more than simple neutral lipid repositories, we set about addressing two related questions. Could LBs provide lipids to live amastigotes hosted within the DL parasitophorous vacuole and also deliver? Could LBs impact either directly or indirectly on the persistence of L. amazonensis amastigotes in rodent skin? Once they have gained entry to mammals, live Leishmania (L) amazonensis amastigotes are known to subvert both macrophages and dendritic leucocytes (DLs) as host cells. These L. amazonensis amastigotes then may or may not proliferate in these two phagocytic leucocyte lineages, but in both cases the otherwise versatile differentiation program of these lineages is known to be rapidly remodeled. Here, we describe the rapid reprogramming of C57BL/6 and DBA/2 mouse bone marrow-derived DLs, with a special focus on cytosolic lipid bodies (LBs) that are known to store neutral lipids such as triacyl-sn-glycerol (TAG) and cholesteryl esters (CE). After extracting RNA from carefully sorted amastigote-free DLs and L. amazonensis amastigote-hosting DLs, an Affymetrix-based analysis clearly showed a singular and coordinated increase in DL transcripts involved in (i) long-chain fatty acid uptake, transport and esterification to TAG and (ii) cholesterol uptake and esterification to cholesteryl esters. Oleic acid was added to check that neutral lipid metabolism was both rapidly increased and reprogrammed in amastigote-hosting DLs. It should be noted that the LBs in live amastigote-hosting DLs were more numerous, and that the largest of these LBs were in contact with live amastigote- hosting parasitophorous vacuoles. We further discuss these findings in the context of live L. amazonensis amastigote-rodent host interactions.
Collapse
Affiliation(s)
- Hervé Lecoeur
- Institut Pasteur, Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Paris, France
- * E-mail: (HL); (TL)
| | - Emilie Giraud
- Institut Pasteur, Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Paris, France
| | - Marie-Christine Prévost
- Institut Pasteur, Département Biologie Cellulaire et Infection, Plateforme de Microscopie Ultrastructurale, Paris, France
| | - Geneviève Milon
- Institut Pasteur, Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Paris, France
| | - Thierry Lang
- Institut Pasteur, Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Paris, France
- Institut Pasteur, Département Infection et Epidémiologie, Laboratoire des Processus Infectieux à Trypanosomatidés, Paris, France
- * E-mail: (HL); (TL)
| |
Collapse
|
45
|
Carter CJ. Toxoplasmosis and Polygenic Disease Susceptibility Genes: Extensive Toxoplasma gondii Host/Pathogen Interactome Enrichment in Nine Psychiatric or Neurological Disorders. J Pathog 2013; 2013:965046. [PMID: 23533776 PMCID: PMC3603208 DOI: 10.1155/2013/965046] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 08/18/2012] [Accepted: 09/10/2012] [Indexed: 01/04/2023] Open
Abstract
Toxoplasma gondii is not only implicated in schizophrenia and related disorders, but also in Alzheimer's or Parkinson's disease, cancer, cardiac myopathies, and autoimmune disorders. During its life cycle, the pathogen interacts with ~3000 host genes or proteins. Susceptibility genes for multiple sclerosis, Alzheimer's disease, schizophrenia, bipolar disorder, depression, childhood obesity, Parkinson's disease, attention deficit hyperactivity disorder (P from 8.01E - 05 (ADHD) to 1.22E - 71) (multiple sclerosis), and autism (P = 0.013), but not anorexia or chronic fatigue are highly enriched in the human arm of this interactome and 18 (ADHD) to 33% (MS) of the susceptibility genes relate to it. The signalling pathways involved in the susceptibility gene/interactome overlaps are relatively specific and relevant to each disease suggesting a means whereby susceptibility genes could orient the attentions of a single pathogen towards disruption of the specific pathways that together contribute (positively or negatively) to the endophenotypes of different diseases. Conditional protein knockdown, orchestrated by T. gondii proteins or antibodies binding to those of the host (pathogen derived autoimmunity) and metabolite exchange, may contribute to this disruption. Susceptibility genes may thus be related to the causes and influencers of disease, rather than (and as well as) to the disease itself.
Collapse
Affiliation(s)
- C. J. Carter
- Polygenic Pathways, Flat 2, 40 Baldslow Road, Hastings, East Sussex TN34 2EY, UK
| |
Collapse
|
46
|
Lige B, Sampels V, Coppens I. Characterization of a second sterol-esterifying enzyme in Toxoplasma highlights the importance of cholesterol storage pathways for the parasite. Mol Microbiol 2013; 87:951-67. [PMID: 23374239 DOI: 10.1111/mmi.12142] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2012] [Indexed: 11/28/2022]
Abstract
Lipid bodies are eukaryotic structures for temporary storage of neutral lipids such as acylglycerols and steryl esters. Fatty acyl-CoA and cholesterol are two substrates for cholesteryl ester (CE) synthesis via the ACAT reaction. The intracellular parasite Toxoplasma gondii is incapable of sterol synthesis and unremittingly scavenges cholesterol from mammalian host cells. We previously demonstrated that the parasite expresses a cholesteryl ester-synthesizing enzyme, TgACAT1. In this article, we identified and characterized a second ACAT-like enzyme, TgACAT2, which shares 56% identity with TgACAT1. Both enzymes are endoplasmic reticulum-associated and contribute to CE formation for storage in lipid bodies. While TgACAT1 preferentially utilizes palmitoyl-CoA, TgACAT2 has broader fatty acid specificity and produces more CE. Genetic ablation of each individual ACAT results in parasite growth impairment whereas dual ablation of ACAT1 and ACAT2 is not tolerated by Toxoplasma. ΔACAT1 and ΔACAT2 parasites have reduced CE levels, fewer lipid bodies, and accumulate free cholesterol, which causes injurious membrane effects. Mutant parasites are particularly vulnerable to ACAT inhibitors. This study underlines the important physiological role of ACAT enzymes to store cholesterol in a sterol-auxotrophic organism such as Toxoplasma, and furthermore opens up possibilities of exploiting TgACAT as targets for the development of antitoxoplasmosis drugs.
Collapse
Affiliation(s)
- Bao Lige
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
47
|
Ehrenman K, Wanyiri JW, Bhat N, Ward HD, Coppens I. Cryptosporidium parvum scavenges LDL-derived cholesterol and micellar cholesterol internalized into enterocytes. Cell Microbiol 2013; 15:1182-97. [PMID: 23311949 DOI: 10.1111/cmi.12107] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/13/2012] [Accepted: 12/27/2012] [Indexed: 11/26/2022]
Abstract
Cryptosporidium spp. are responsible for devastating diarrhoea in immunodeficient individuals. In the intestinal tract, the developmental stages of the parasite are confined to the apical surfaces of epithelial cells. Upon invasion, Cryptosporidium incorporates the microvillous membrane of the enterocyte to form the parasitophorous vacuole (PV) and sequesters itself from the host cytoplasm by rearranging the host cytoskeleton. Cryptosporidium parvum has minimal anabolic capabilities and relies on transporters and salvage pathways to meet its basic metabolic requirements. The cholesterol salvage pathway is crucial for the development of protozoan parasites. In this study, we have examined the sources of cholesterol from C. parvum infecting enterocytes. We illustrated that the intracellular stages of Cryptosporidium as well as the oocysts shed by the host, contain cholesterol. Incubation of infected enterocytes in lipoprotein-free medium impairs parasite development and results in substantial decrease in cholesterol content associated with the PV. Among lipoproteins, LDL constitutes an important source of cholesterol for Cryptosporidium. Dietary cholesterol incorporated into micelles is internalized into enterocytes by the NPC1L1 transporter. We showed that C. parvum also obtains cholesterol from micelles in enterocytes.Pharmacological blockade of NPC1L1 function by ezetimibe or moderate downregulation of NPC1L1 expression decreases parasite infectivity. These observations indicate that, despite its dual sequestration from the intestinal lumen and the host cytoplasm, C. parvum can, in fact, obtain cholesterol both from the gut's lumen and the host cell. This study highlights the evolutionary advantages for epicellular pathogens to access to nutrients from the outside and inside of the host cell.
Collapse
Affiliation(s)
- Karen Ehrenman
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
48
|
Cell biology of chromerids: autotrophic relatives to apicomplexan parasites. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 306:333-69. [PMID: 24016529 DOI: 10.1016/b978-0-12-407694-5.00008-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chromerida are algae possessing a complex plastid surrounded by four membranes. Although isolated originally from stony corals in Australia, they seem to be globally distributed. According to their molecular phylogeny, morphology, ultrastructure, structure of organellar genomes, and noncanonical pathway for tetrapyrrole synthesis, these algae are thought to be the closest known phototrophic relatives to apicomplexan parasites. Here, we summarize the current knowledge of cell biology and evolution of this novel group of algae, which contains only two formally described species, but is apparently highly diverse and virtually ubiquitous in marine environments.
Collapse
|
49
|
Leblond JD, Dodson J, Khadka M, Holder S, Seipelt RL. Sterol Composition and Biosynthetic Genes of the Recently Discovered Photosynthetic Alveolate, Chromera velia (Chromerida), a Close Relative of Apicomplexans. J Eukaryot Microbiol 2012; 59:191-7. [DOI: 10.1111/j.1550-7408.2012.00611.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 12/05/2011] [Indexed: 11/28/2022]
Affiliation(s)
- Jeffrey D. Leblond
- Department of Biology; Middle Tennessee State University; P. O. Box 60; Murfreesboro; Tennessee; 37132
| | - Joshua Dodson
- Department of Biology; Middle Tennessee State University; P. O. Box 60; Murfreesboro; Tennessee; 37132
| | - Manoj Khadka
- Department of Biology; Middle Tennessee State University; P. O. Box 60; Murfreesboro; Tennessee; 37132
| | - Sabrina Holder
- Department of Biology; Middle Tennessee State University; P. O. Box 60; Murfreesboro; Tennessee; 37132
| | - Rebecca L. Seipelt
- Department of Biology; Middle Tennessee State University; P. O. Box 60; Murfreesboro; Tennessee; 37132
| |
Collapse
|
50
|
Deficiency of a Niemann-Pick, type C1-related protein in toxoplasma is associated with multiple lipidoses and increased pathogenicity. PLoS Pathog 2011; 7:e1002410. [PMID: 22174676 PMCID: PMC3234224 DOI: 10.1371/journal.ppat.1002410] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 10/16/2011] [Indexed: 02/07/2023] Open
Abstract
Several proteins that play key roles in cholesterol synthesis, regulation, trafficking and signaling are united by sharing the phylogenetically conserved 'sterol-sensing domain' (SSD). The intracellular parasite Toxoplasma possesses at least one gene coding for a protein containing the canonical SSD. We investigated the role of this protein to provide information on lipid regulatory mechanisms in the parasite. The protein sequence predicts an uncharacterized Niemann-Pick, type C1-related protein (NPC1) with significant identity to human NPC1, and it contains many residues implicated in human NPC disease. We named this NPC1-related protein, TgNCR1. Mammalian NPC1 localizes to endo-lysosomes and promotes the movement of sterols and sphingolipids across the membranes of these organelles. Miscoding patient mutations in NPC1 cause overloading of these lipids in endo-lysosomes. TgNCR1, however, lacks endosomal targeting signals, and localizes to flattened vesicles beneath the plasma membrane of Toxoplasma. When expressed in mammalian NPC1 mutant cells and properly addressed to endo-lysosomes, TgNCR1 restores cholesterol and GM1 clearance from these organelles. To clarify the role of TgNCR1 in the parasite, we genetically disrupted NCR1; mutant parasites were viable. Quantitative lipidomic analyses on the ΔNCR1 strain reveal normal cholesterol levels but an overaccumulation of several species of cholesteryl esters, sphingomyelins and ceramides. ΔNCR1 parasites are also characterized by abundant storage lipid bodies and long membranous tubules derived from their parasitophorous vacuoles. Interestingly, these mutants can generate multiple daughters per single mother cell at high frequencies, allowing fast replication in vitro, and they are slightly more virulent in mice than the parental strain. These data suggest that the ΔNCR1 strain has lost the ability to control the intracellular levels of several lipids, which subsequently results in the stimulation of lipid storage, membrane biosynthesis and parasite division. Based on these observations, we ascribe a role for TgNCR1 in lipid homeostasis in Toxoplasma.
Collapse
|