1
|
Prenen F, Pollenus E, Meers H, Knoops S, Sadler R, Deckers M, Mills EL, Van den Steen PE. Itaconate Has Limited Protective Effects in Experimental Malaria Models. Eur J Immunol 2025; 55:e202451595. [PMID: 40346757 DOI: 10.1002/eji.202451595] [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: 10/22/2024] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/12/2025]
Abstract
In severe malaria, dysregulated metabolism and excessive inflammatory responses contribute to fatal outcomes. Therapeutic strategies that address both metabolic and inflammatory balances are thus required. Itaconate, a metabolite produced by aconitate decarboxylase 1 (ACOD1), is a potent inhibitor of both inflammation and glycolysis with protective effects in various inflammatory diseases. Although elevated itaconate levels have been observed in Plasmodium-infected individuals, its role in malaria is still poorly understood, making further investigation essential for assessing its therapeutic potential. We investigated the role of itaconate in both severe and mild malaria using Plasmodium berghei NK65 (PbNK65) and Plasmodium chabaudi AS (PcAS) models, respectively. Using 13C-tracer metabolomics, we detected increased itaconate levels in various organs during infection and identified inflammatory monocytes as the source of this production. Nevertheless, ACOD1 knockout mice displayed no significant changes in phenotype after PbNK65 infection, and treatment of PbNK65-infected mice with 4-octyl itaconate did not affect disease severity either. However, in the PcAS model, ACOD1 deficiency worsened the disease, as indicated by increased weight loss, higher clinical scores, and elevated parasitemia. Therefore, in contrast to the findings in recent literature, our study shows that itaconate does not contribute to susceptibility, but rather provides limited protection to malaria.
Collapse
Affiliation(s)
- Fran Prenen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Hanne Meers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Rebecca Sadler
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Margot Deckers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Laboratory of Host-Pathogen Interactions, Department of Pathology, University of Utah, Salt Lake City, USA
| | - Evanna L Mills
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Philippe E Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Das MK, Savidge B, Pearl JE, Yates T, Miles G, Pareek M, Haldar P, Cooper AM. Altered hepatic metabolic landscape and insulin sensitivity in response to pulmonary tuberculosis. PLoS Pathog 2024; 20:e1012565. [PMID: 39331683 PMCID: PMC11463835 DOI: 10.1371/journal.ppat.1012565] [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: 12/12/2023] [Revised: 10/09/2024] [Accepted: 08/26/2024] [Indexed: 09/29/2024] Open
Abstract
Chronic inflammation triggers development of metabolic disease, and pulmonary tuberculosis (TB) generates chronic systemic inflammation. Whether TB induced-inflammation impacts metabolic organs and leads to metabolic disorder is ill defined. The liver is the master regulator of metabolism and to determine the impact of pulmonary TB on this organ we undertook an unbiased mRNA and protein analyses of the liver in mice with TB and reanalysed published data on human disease. Pulmonary TB led to upregulation of genes in the liver related to immune signalling and downregulation of genes encoding metabolic processes. In liver, IFN signalling pathway genes were upregulated and this was reflected in increased biochemical evidence of IFN signalling, including nuclear location of phosphorylated Stat-1 in hepatocytes. The liver also exhibited reduced expression of genes encoding the gluconeogenesis rate-limiting enzymes Pck1 and G6pc. Phosphorylation of CREB, a transcription factor controlling gluconeogenesis was drastically reduced in the livers of mice with pulmonary TB as was phosphorylation of other glucose metabolism-related kinases, including GSK3a, AMPK, and p42. In support of the upregulated IFN signalling being linked to the downregulated metabolic functions in the liver, we found suppression of gluconeogenic gene expression and reduced CREB phosphorylation in hepatocyte cell lines treated with interferons. The impact of reduced gluconeogenic gene expression in the liver was seen when infected mice were less able to convert pyruvate, a gluconeogenesis substrate, to the same extent as uninfected mice. Infected mice also showed evidence of reduced systemic and hepatic insulin sensitivity. Similarly, in humans with TB, we found that changes in a metabolite-based signature of insulin resistance correlates temporally with successful treatment of active TB and with progression to active TB following exposure. These data support the hypothesis that TB drives interferon-mediated alteration of hepatic metabolism resulting in reduced gluconeogenesis and drives systemic reduction of insulin sensitivity.
Collapse
Affiliation(s)
- Mrinal K. Das
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
| | - Ben Savidge
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
| | - John E. Pearl
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
| | - Thomas Yates
- Diabetes Research Centre, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Gareth Miles
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, United Kingdom
| | - Manish Pareek
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Pranabashis Haldar
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
- NIHR Respiratory Biomedical Research Centre, Leicester, Glenfield Hospital, Groby Road, Leicester, United Kingdom
| | - Andrea M. Cooper
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| |
Collapse
|
3
|
Kotepui KU, Masangkay FR, Wangdi K, Mahittikorn A, Majima HJ, Kotepui M. A systematic review and meta-analysis of cortisol levels in Plasmodium infections. Sci Rep 2024; 14:18162. [PMID: 39107355 PMCID: PMC11303744 DOI: 10.1038/s41598-024-68596-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
Malaria has complex interactions with host physiology, including alterations in cortisol levels. Cortisol, a key hormone in the stress response, is known to be dysregulated in various infectious diseases. This systematic review and meta-analysis aimed to elucidate the relationship between Plasmodium infection and cortisol levels, shedding light on the intricate interplay between the parasite and the host's endocrine system. The methodological protocol for assessing cortisol levels in malaria patients was registered in PROSPERO (CRD42024496578), a widely recognized international prospective register of systematic reviews. This registration ensures transparency and minimizes the risk of bias in our research. A comprehensive search strategy was employed across major databases, including Embase, PubMed, Scopus, and Medline, to include studies that reported cortisol levels in infected patients. The qualitative synthesis was undertaken to synthesize the difference in cortisol levels between malaria-infected and uninfected individuals. The meta-analysis employed the random effects model in the quantitative synthesis to calculate the effect estimate. The review included a total of 20 studies, with a substantial number conducted in Africa, followed by Asia and South America. Most included studies (13/20, 65%) reported higher cortisol levels in infected patients than in uninfected patients. The meta-analysis confirmed significantly higher cortisol levels in infected patients compared to uninfected individuals (P < 0.0001, standardized mean difference (SMD): 1.354, 95% confidence interval: 0.913 to 1.795, I2: 88.3%, across 15 studies). Notably, the method for cortisol measurement and the type of blood sample used (serum or plasma) were significant moderators in the analysis, indicating that these factors may influence the observed relationship between Plasmodium infection and cortisol levels. The systematic review and meta-analysis confirmed that Plasmodium infection is associated with increased cortisol levels, highlighting the intricate relationship between the disease and the host stress response. These findings underscore the potential of cortisol as a supplementary biomarker for understanding the pathophysiological impact of malaria. By providing insights into the stress-related mechanisms of malaria, this comprehensive understanding can inform future research and potentially enhance disease management and treatment strategies, particularly in regions heavily burdened by malaria.
Collapse
Affiliation(s)
- Kwuntida Uthaisar Kotepui
- Medical Technology Program, Faculty of Science, Nakhon Phanom University, Nakhon Phanom, 48000, Thailand
| | | | - Kinley Wangdi
- HEAL Global Research Centre, Health Research Institute, Faculty of Health, University of Canberra, Bruce, ACT, 2617, Australia
- National Centre for Epidemiology and Population Health, College of Health and Medicine, ANU, Acton, ACT, 2601, Australia
| | - Aongart Mahittikorn
- Department of Protozoology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| | - Hideyuki J Majima
- Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, 80160, Thailand
| | - Manas Kotepui
- Medical Technology Program, Faculty of Science, Nakhon Phanom University, Nakhon Phanom, 48000, Thailand.
| |
Collapse
|
4
|
Bauer M, Ermolaeva M, Singer M, Wetzker R, Soares MP. Hormesis as an adaptive response to infection. Trends Mol Med 2024; 30:633-641. [PMID: 38744580 DOI: 10.1016/j.molmed.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 05/16/2024]
Abstract
Hormesis is a phenomenon whereby low-level stress can improve cellular, organ, or organismal fitness in response to a subsequent similar or other stress insult. Whereas hormesis is thought to contribute to the fitness benefits arising from symbiotic host-microbe interactions, the putative benefits of hormesis in host-pathogen interactions have yet to be explored. Hormetic responses have nonetheless been reported in experimental models of infection, a common feature of which is regulation of host mitochondrial function. We propose that these mitohormetic responses could be harnessed therapeutically to limit the severity of infectious diseases.
Collapse
Affiliation(s)
- Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| | - Maria Ermolaeva
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany; Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, UK
| | - Reinhard Wetzker
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Miguel P Soares
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany; Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|
5
|
Kreimendahl S, Pernas L. Metabolic immunity against microbes. Trends Cell Biol 2024; 34:496-508. [PMID: 38030541 DOI: 10.1016/j.tcb.2023.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/11/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
Pathogens, including viruses, bacteria, fungi, and parasites, remodel the metabolism of their host to acquire the nutrients they need to proliferate. Thus, host cells are often perceived as mere exploitable nutrient pools during infection. Mounting reports challenge this perception and instead suggest that host cells can actively reprogram their metabolism to the detriment of the microbial invader. In this review, we present metabolic mechanisms that host cells use to defend against pathogens. We highlight the contribution of domesticated microbes to host defenses and discuss examples of host-pathogen arms races that are derived from metabolic conflict.
Collapse
Affiliation(s)
| | - Lena Pernas
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
6
|
Carvalho Cabral P, Weinerman J, Olivier M, Cermakian N. Time of day and circadian disruption influence host response and parasite growth in a mouse model of cerebral malaria. iScience 2024; 27:109684. [PMID: 38680656 PMCID: PMC11053314 DOI: 10.1016/j.isci.2024.109684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/20/2023] [Accepted: 04/04/2024] [Indexed: 05/01/2024] Open
Abstract
Malaria is a disease caused by infection with parasite Plasmodium spp. We studied the circadian regulation of host responses to the parasite, in a mouse model of cerebral malaria. The course of the disease was markedly affected by time of infection, with decreased parasitemia and increased inflammation upon infection in the middle of the night. At this time, there were fewer reticulocytes, which are target cells of the parasites. We next investigated the effects of desynchronization of host clocks on the infection: after 10 weeks of recurrent jet lags, mice showed decreased parasite growth and lack of parasite load rhythmicity, paralleled by a loss of glucose rhythm. Accordingly, disrupting host metabolic rhythms impacted parasite load rhythmicity. In summary, our findings of a circadian modulation of malaria parasite growth and infection shed light on aspects of the disease relevant to human malaria and could contribute to new therapeutic or prophylactic measures.
Collapse
Affiliation(s)
- Priscilla Carvalho Cabral
- Douglas Research Centre, McGill University, Montréal, QC H4H 1R3, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
| | - Joelle Weinerman
- Douglas Research Centre, McGill University, Montréal, QC H4H 1R3, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
- Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Nicolas Cermakian
- Douglas Research Centre, McGill University, Montréal, QC H4H 1R3, Canada
| |
Collapse
|
7
|
Kong W, Ding G, Cheng G, Yang P, Xu Z. Mucosal immune responses to Ichthyophthirius multifiliis in the ocular mucosa of rainbow trout ( Oncorhynchus mykiss, Walbaum), an ancient teleost fish. MARINE LIFE SCIENCE & TECHNOLOGY 2024; 6:266-279. [PMID: 38827132 PMCID: PMC11136906 DOI: 10.1007/s42995-023-00199-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/16/2023] [Indexed: 06/04/2024]
Abstract
The eye, as a specialized visual organ, is directly exposed to the external environment, and, therefore, it faces constant challenges from external pathogenic organisms and toxins. In the ocular mucosa (OM) of mammals, mucosal-associated lymphoid tissues (MALTs) constitute the primary line of defense. However, the immune defense role of the OM remains unknown in aquatic vertebrates. To gain insights into the immune processes within the OM of teleost fish, we developed an infection model of rainbow trout (Oncorhynchus mykiss) OM using a parasite, Ichthyophthirius multifiliis (Ich). Immunofluorescence, qPCR, and H&E staining revealed that Ich successfully infiltrates the OM of rainbow trout, leading to pathological structural changes, as evidenced by A&B staining. Importantly, the qPCR results indicate an up-regulation of immune-related genes following Ich infection in the OM. Moreover, transcriptome analyses were conducted to detect immune responses and impairments in eye function within the OM of rainbow trout with Ich infection. The results of the transcriptome analysis that Ich infection can cause an extensive immune response in the OM, ultimately affecting ocular function. To the best of our knowledge, our findings represent for the first time that the teleost OM could act as an invasion site for parasites and trigger a strong mucosal immune response to parasitic infection. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-023-00199-6.
Collapse
Affiliation(s)
- Weiguang Kong
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 China
| | - Guangyi Ding
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 China
| | - Gaofeng Cheng
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070 China
| | - Peng Yang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 China
| | - Zhen Xu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 China
| |
Collapse
|
8
|
Ramos S, Jeney V, Figueiredo A, Paixão T, Sambo MR, Quinhentos V, Martins R, Gouveia Z, Carlos AR, Ferreira A, Pais TF, Lainé H, Faísca P, Rebelo S, Cardoso S, Tolosano E, Penha-Gonçalves C, Soares MP. Targeting circulating labile heme as a defense strategy against malaria. Life Sci Alliance 2024; 7:e202302276. [PMID: 38307624 PMCID: PMC10837040 DOI: 10.26508/lsa.202302276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/04/2024] Open
Abstract
Severe presentations of malaria emerge as Plasmodium (P.) spp. parasites invade and lyse red blood cells (RBC), producing extracellular hemoglobin (HB), from which labile heme is released. Here, we tested whether scavenging of extracellular HB and/or labile heme, by haptoglobin (HP) and/or hemopexin (HPX), respectively, counter the pathogenesis of severe presentations of malaria. We found that circulating labile heme is an independent risk factor for cerebral and non-cerebral presentations of severe P. falciparum malaria in children. Labile heme was negatively correlated with circulating HP and HPX, which were, however, not risk factors for severe P. falciparum malaria. Genetic Hp and/or Hpx deletion in mice led to labile heme accumulation in plasma and kidneys, upon Plasmodium infection This was associated with higher incidence of mortality and acute kidney injury (AKI) in ageing but not adult Plasmodium-infected mice, and was corroborated by an inverse correlation between heme and HPX with serological markers of AKI in P. falciparum malaria. In conclusion, HP and HPX act in an age-dependent manner to prevent the pathogenesis of severe presentation of malaria in mice and presumably in humans.
Collapse
Affiliation(s)
- Susana Ramos
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | - Tiago Paixão
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Maria Rosário Sambo
- Hospital Pediátrico David Bernardino, Luanda, Angola
- Faculdade de Medicina, Universidade Agostinho Neto, Luanda, Angola
| | - Vatúsia Quinhentos
- Hospital Pediátrico David Bernardino, Luanda, Angola
- Faculdade de Medicina, Universidade Agostinho Neto, Luanda, Angola
| | - Rui Martins
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | - Ana Ferreira
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | - Hugo Lainé
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Pedro Faísca
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Sofia Rebelo
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | - Emanuela Tolosano
- Department Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | | | | |
Collapse
|
9
|
Wu Q, Carlos AR, Braza F, Bergman ML, Kitoko JZ, Bastos-Amador P, Cuadrado E, Martins R, Oliveira BS, Martins VC, Scicluna BP, Landry JJ, Jung FE, Ademolue TW, Peitzsch M, Almeida-Santos J, Thompson J, Cardoso S, Ventura P, Slot M, Rontogianni S, Ribeiro V, Domingues VDS, Cabral IA, Weis S, Groth M, Ameneiro C, Fidalgo M, Wang F, Demengeot J, Amsen D, Soares MP. Ferritin heavy chain supports stability and function of the regulatory T cell lineage. EMBO J 2024; 43:1445-1483. [PMID: 38499786 PMCID: PMC11021483 DOI: 10.1038/s44318-024-00064-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/20/2024] Open
Abstract
Regulatory T (TREG) cells develop via a program orchestrated by the transcription factor forkhead box protein P3 (FOXP3). Maintenance of the TREG cell lineage relies on sustained FOXP3 transcription via a mechanism involving demethylation of cytosine-phosphate-guanine (CpG)-rich elements at conserved non-coding sequences (CNS) in the FOXP3 locus. This cytosine demethylation is catalyzed by the ten-eleven translocation (TET) family of dioxygenases, and it involves a redox reaction that uses iron (Fe) as an essential cofactor. Here, we establish that human and mouse TREG cells express Fe-regulatory genes, including that encoding ferritin heavy chain (FTH), at relatively high levels compared to conventional T helper cells. We show that FTH expression in TREG cells is essential for immune homeostasis. Mechanistically, FTH supports TET-catalyzed demethylation of CpG-rich sequences CNS1 and 2 in the FOXP3 locus, thereby promoting FOXP3 transcription and TREG cell stability. This process, which is essential for TREG lineage stability and function, limits the severity of autoimmune neuroinflammation and infectious diseases, and favors tumor progression. These findings suggest that the regulation of intracellular iron by FTH is a stable property of TREG cells that supports immune homeostasis and limits the pathological outcomes of immune-mediated inflammation.
Collapse
Affiliation(s)
- Qian Wu
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University, School of Medicine, Yiwu, Zhejiang, China
| | - Ana Rita Carlos
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Departamento de Biologia Animal, Centro de Ecologia, Evolução e Alterações Ambientais, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Faouzi Braza
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | | | - Eloy Cuadrado
- Department of Hematopoiesis and Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Rui Martins
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, and Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Jonathan Jm Landry
- Genomic Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Ferris E Jung
- Genomic Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
| | | | | | | | | | - Manon Slot
- Department of Hematopoiesis and Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Stamatia Rontogianni
- Department of Hematopoiesis and Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Vanessa Ribeiro
- Departamento de Biologia Animal, Centro de Ecologia, Evolução e Alterações Ambientais, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | | | | | - Sebastian Weis
- Department for Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll Institute-HKI, Jena, Germany
| | - Marco Groth
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Cristina Ameneiro
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela-Health Research Institute (IDIS), Santiago de Compostela, Spain
| | - Miguel Fidalgo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela-Health Research Institute (IDIS), Santiago de Compostela, Spain
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | | | - Derk Amsen
- Department of Hematopoiesis and Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
10
|
Jentho E, Sousa AGG, Ramos S, Ademolue TW, Sobral J, Costa J, Brito D, Manteiro M, Leite RB, Lilue J, Soares MP. Single-cell RNA sequencing and analysis of rodent blood stage Plasmodium. STAR Protoc 2023; 4:102491. [PMID: 37581982 PMCID: PMC10436236 DOI: 10.1016/j.xpro.2023.102491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/30/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
Bulk RNA sequencing of Plasmodium spp., the causative parasite of malaria, fails to discriminate developmental-stage-specific gene regulation. Here, we provide a protocol that uses single-cell RNA sequencing of FACS-sorted Plasmodium-chabaudi-chabaudi-AS-infected red blood cells (iRBCs) to characterize developmental-stage-specific modulation of gene expression during malaria blood stage. We describe steps for infecting mice, monitoring disease progression, preparing iRBCs, and single-cell sequencing iRBCs. We then detail procedures for analyzing scRNA-seq data. For complete details on the use and execution of this protocol, please refer to Ramos et al.1.
Collapse
Affiliation(s)
- Elisa Jentho
- Instituto Gulbenkian de Ciência, Oeiras, Portugal; Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University, Jena, Germany.
| | | | - Susana Ramos
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | - João Sobral
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - João Costa
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Denise Brito
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | | | | |
Collapse
|
11
|
Shafi AM, Végvári Á, Zubarev RA, Penha-Gonçalves C. Brain endothelial cells exposure to malaria parasites links type I interferon signalling to antigen presentation, immunoproteasome activation, endothelium disruption, and cellular metabolism. Front Immunol 2023; 14:1149107. [PMID: 36993973 PMCID: PMC10042232 DOI: 10.3389/fimmu.2023.1149107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 02/27/2023] [Indexed: 03/14/2023] Open
Abstract
IntroductionCerebral malaria (CM) lethality is attributable to induction of brain edema induction but the cellular mechanisms involving brain microvascular endothelium in CM pathogenesis are unexplored.ResultsActivation of the STING-INFb-CXCL10 axis in brain endothelial cells (BECs) is a prominent component of the innate immune response in CM development in mouse models. Using a T cell-reporter system, we show that Type 1 IFN signaling in BECs exposed to Plasmodium berghei-infected erythrocytes (PbA-IE), functionally enhances MHC Class-I antigen presentation through gamma-interferon independent immunoproteasome activation and impacted the proteome functionally related to vesicle trafficking, protein processing/folding and antigen presentation. In vitro assays showed that Type 1 IFN signaling and immunoproteasome activation are also involved in the dysfunction of the endothelial barrier through disturbing gene expression in the Wnt/ß-catenin signaling pathway. We demonstrate that IE exposure induces a substantial increase in BECs glucose uptake while glycolysis blockade abrogates INFb secretion impairing immunoproteasome activation, antigen presentation and Wnt/ß-catenin signaling.DiscussionMetabolome analysis show that energy demand and production are markedly increased in BECs exposed to IE as revealed by enriched content in glucose and amino acid catabolites. In accordance, glycolysis blockade in vivo delayed the clinical onset of CM in mice. Together the results show that increase in glucose uptake upon IE exposure licenses Type 1 IFN signaling and subsequent immunoproteasome activation contributing to enhanced antigen presentation and impairment of endothelial barrier function. This work raises the hypothesis that Type 1 IFN signaling-immunoproteasome induction in BECs contributes to CM pathology and fatality (1) by increasing antigen presentation to cytotoxic CD8+ T cells and (2) by promoting endothelial barrier dysfunction, that likely favor brain vasogenic edema.
Collapse
Affiliation(s)
| | - Ákos Végvári
- Proteomics Biomedicum, Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Roman A. Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Carlos Penha-Gonçalves
- Disease Genetics, Instituto Gulbenkian de Ciência, Oeiras, Portugal
- *Correspondence: Carlos Penha-Gonçalves,
| |
Collapse
|
12
|
Wu Q, Sacomboio E, Valente de Souza L, Martins R, Kitoko J, Cardoso S, Ademolue TW, Paixão T, Lehtimäki J, Figueiredo A, Norden C, Tharaux PL, Weiss G, Wang F, Ramos S, Soares MP. Renal control of life-threatening malarial anemia. Cell Rep 2023; 42:112057. [PMID: 36735532 DOI: 10.1016/j.celrep.2023.112057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/30/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Iron recycling prevents the development of anemia under homeostatic conditions. Whether iron recycling was co-opted as a defense strategy to prevent the development of anemia in response to infection is unclear. We find that in severe Plasmodium falciparum malaria, the onset of life-threatening anemia is associated with acute kidney injury (AKI), irrespective of parasite load. Using a well-established experimental rodent model of malaria anemia, we identify a transcriptional response that endows renal proximal tubule epithelial cells (RPTECs) with the capacity to store and recycle iron during P. chabaudi chabaudi (Pcc) infection. This response encompasses the induction of ferroportin 1/SLC40A1, which exports iron from RPTECs and counteracts AKI while supporting compensatory erythropoiesis and preventing the onset of life-threatening malarial anemia. Iron recycling by myeloid cells is dispensable to this protective response, suggesting that RPTECs provide an iron-recycling salvage pathway that prevents the pathogenesis of life-threatening malarial anemia.
Collapse
Affiliation(s)
- Qian Wu
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | - Lara Valente de Souza
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Rui Martins
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Jamil Kitoko
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | - Tiago Paixão
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | - Caren Norden
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Center (PARCC), Institut National de la Santé et de la Recherche Médicale (Inserm), Université Paris Cité, Paris, France
| | - Guenter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Fudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Susana Ramos
- Instituto Gulbenkian de Ciência, Oeiras, Portugal.
| | | |
Collapse
|
13
|
Gluconeogenesis: Adding a new piece to the puzzle of malaria parasitism. Cell Metab 2022; 34:1083-1085. [PMID: 35921815 DOI: 10.1016/j.cmet.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Disorders of carbohydrate metabolism, including hypoglycemia and lactic acidosis, are common features of malaria. In this issue of Cell Metabolism, Ramos et al. report that regulation of gluconeogenesis and glycemia by the host glucose-6-phosphatase catalytic subunit 1 (G6Pc1) is a key metabolic step that affects both Plasmodium replication and clinical outcome of disease.
Collapse
|