1
|
Ananthasubramaniam B, Venkataramanan R. Rhythm profiling using COFE reveals multi-omic circadian rhythms in human cancers in vivo. PLoS Biol 2025; 23:e3003196. [PMID: 40424435 DOI: 10.1371/journal.pbio.3003196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
The study of ubiquitous circadian rhythms in human physiology requires regular measurements across time. Repeated sampling of the different internal tissues that house circadian clocks is both practically and ethically infeasible. Here, we present a novel unsupervised machine learning approach (COFE) that can use single high-throughput omics samples (without time labels) from individuals to reconstruct circadian rhythms across cohorts. COFE can simultaneously assign time labels to samples and identify rhythmic data features used for temporal reconstruction, while also detecting invalid orderings. With COFE, we discovered widespread de novo circadian gene expression rhythms in 11 different human adenocarcinomas using data from The Cancer Genome Atlas (TCGA) database. The arrangement of peak times of core clock gene expression was conserved across cancers and resembled a healthy functional clock except for the mistiming of a few key genes. Moreover, rhythms in the transcriptome were strongly associated with the cancer-relevant proteome. The rhythmic genes and proteins common to all cancers were involved in metabolism and the cell cycle. Although these rhythms were synchronized with the cell cycle in many cancers, they were uncoupled with clocks in healthy matched tissue. The targets of most of FDA-approved and potential anti-cancer drugs were rhythmic in tumor tissue with different amplitudes and peak times. These findings emphasize the utility of considering "time" in cancer therapy, and suggest a focus on clocks in healthy tissue rather than free-running clocks in cancer tissue. Our approach thus creates new opportunities to repurpose data without time labels to study circadian rhythms.
Collapse
|
2
|
Wada A, Asahi H. Identification of a Class of Iron-Grabbing Compounds with Antiplasmodial Activity: Impact of Coordination Structures and Electronic Regularity on the Intraerythrocytic Growth Cycle of Plasmodium falciparum. ACS Infect Dis 2025; 11:799-804. [PMID: 40162597 DOI: 10.1021/acsinfecdis.4c00846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Innovative antimalarials are required to combat malaria, a global infectious disease caused by Plasmodium falciparum. To explore the untapped antiplasmodial compounds that can target the iron source vital at the blood stages of P. falciparum, we investigated the antiplasmodial activities of natural siderophores and synthetic compounds with metal-binding affinity. The assessment of their IC50 values and spectroscopic analytical data revealed that terpyridyl compounds specifically bound to target Fe(II) ions and strongly induced the growth inhibition of intraerythrocytic parasites. Furthermore, the IC50 values of the 4,4',4''-substituted terpyridines were linearly correlated with the sum of the para Hammett constants of their substitutions, suggesting that their growth inhibitory effects depended on the electronic states of the coordinating nitrogen atoms. Considering the specific developmental blockage at the trophozoite stage and selective antiplasmodial activities of the iron-grabbing compounds, these findings provide insights into the development of antimalarials that can disrupt iron homeostasis.
Collapse
Affiliation(s)
- Akira Wada
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hiroko Asahi
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
3
|
Circadian rhythms mediate malaria transmission. Nat Microbiol 2025; 10:821-822. [PMID: 40175720 DOI: 10.1038/s41564-025-01950-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
|
4
|
Motta FC, McGoff K, Cummins B, Haase SB. Generalized measures of population synchrony. Math Biosci 2025; 380:109344. [PMID: 39732161 DOI: 10.1016/j.mbs.2024.109344] [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: 06/25/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/30/2024]
Abstract
Synchronized behavior among individuals, broadly defined, is a ubiquitous feature of populations. Understanding mechanisms of (de)synchronization demands meaningful, interpretable, computable quantifications of synchrony, relevant to measurements that can be made of complex, dynamic populations. Despite the importance to analyzing and modeling populations, existing notions of synchrony often lack rigorous definitions, may be specialized to a particular experimental system and/or measurement, or may have undesirable properties that limit their utility. Here we introduce a notion of synchrony for populations of individuals occupying a compact metric space that depends on the Fréchet variance of the distribution of individuals across the space. We establish several fundamental and desirable mathematical properties of our proposed measure of synchrony, including continuity and invariance to metric scaling. We establish a general approximation result that controls the disparity between synchrony in the true space and the synchrony observed through a discretization of state space, as may occur when observable states are limited by measurement constraints. We develop efficient algorithms to compute synchrony for distributions in a variety of state spaces, including all finite state spaces and empirical distributions on the circle, and provide accessible implementations in an open-source Python module. To demonstrate the usefulness of the synchrony measure in biological applications, we investigate several biologically relevant models of mechanisms that can alter the dynamics of population synchrony over time, and reanalyze published experimental and model data concerning the dynamics of the intraerythrocytic developmental cycles of Plasmodium parasites. We anticipate that the rigorous definition of population synchrony and the mathematical and biological results presented here will be broadly useful in analyzing and modeling populations in a variety of contexts.
Collapse
Affiliation(s)
- Francis C Motta
- Department of Mathematics and Statistics, Florida Atlantic University, Boca Raton, FL, USA.
| | - Kevin McGoff
- Department of Mathematics and Statistics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Breschine Cummins
- Department of Mathematical Sciences, Montana State University, Bozeman, MT, USA
| | | |
Collapse
|
5
|
Larrondo LF. Circadian rhythms: pervasive, and often times evasive. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230477. [PMID: 39842475 DOI: 10.1098/rstb.2023.0477] [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: 04/23/2024] [Revised: 08/23/2024] [Accepted: 08/30/2024] [Indexed: 01/24/2025] Open
Abstract
Most circadian texts begin by stating that clocks are pervasive throughout the tree of life. Indeed, clock mechanisms have been described from cyanobacteria to humans, representing a notable example of convergent evolution: yet, there are several phyla in animals, protists or within fungi and bacteria, in which homologs of some-or all-known clock components seem to be absent, posing inevitable questions about the evolution of circadian systems. Moreover, as we move away from model organisms, there are several taxa in which core clock elements can be identified at the genomic levels. However, the functional description of those putative clocks has been hard to achieve, as rhythmicity is not observed unless defined abiotic or nutritional cues are provided. The mechanisms 'conditioning' the functionality of clocks remain uncertain, emphasizing the need to delve further into non-model circadian systems. As the absence of evidence is not evidence of absence, the lack of known core-clock homologs or of observable rhythms in a given organism, cannot be an a priori criterion to discard the presence of a functional clock, as rhythmicity may be limited to yet untested experimental conditions or phenotypes. This article seeks to reflect on these topics, highlighting some of the pressing questions awaiting to be addressed.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Luis F Larrondo
- ANID-Millennium Science Initiative Program-Millennium Institute for Integrative Biology (iBio), Santiago 8331150, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| |
Collapse
|
6
|
Hirako IC, Ramalho T, Gazzinelli RT. Immune regulation of host energy metabolism and periodicity of malaria parasites. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230511. [PMID: 39842477 PMCID: PMC11753876 DOI: 10.1098/rstb.2023.0511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 08/29/2024] [Accepted: 11/06/2024] [Indexed: 01/24/2025] Open
Abstract
The synchronization of Plasmodium parasites as they replicate within red blood cells of their vertebrate host remains largely unexplored. Understanding this synchronization could reveal how parasites optimize their lifecycle to maximize transmission, evade the immune response and maximize energy acquisition. Rhythmic replication fulfils some criteria of an endogenous oscillator with time of day cues potentially provided by temperature, oxygen levels, hormones and/or nutrient availability. Recent research on a rodent malaria model has highlighted that rhythms associated with the host's feeding/fasting cycle are a crucial factor influencing the synchronization of the erythrocytic stages of Plasmodium to the host's circadian cycle. Innate immune responses are also rhythmic and can regulate host metabolism, suggesting that the innate immune response triggered by Plasmodium contributes to its rhythmic replication. Here, we outline how the interplay between immune responses and metabolism could influence the timing and synchronization of Plasmodium's replication rhythm, focusing on the roles of the cytokine tumour necrosis factor, mitochondrial function and metabolites generated by the tricarboxylic acid cycle in highly activated monocytes. These processes are pivotal in controlling parasitemia and determining disease outcome, suggesting that a better understanding of energy metabolism on rhythmic host-parasite interactions may provide new insights for therapeutic interventions against malaria.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Isabella Cristina Hirako
- Laboratory of Immunopathology - Instituto René Rachou, Fundação Oswaldo Cruz - Minas, Belo Horizonte30190-002, Brazil
| | - Theresa Ramalho
- Department of Molecular Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA01605, USA
| | - Ricardo Tostes Gazzinelli
- Laboratory of Immunopathology - Instituto René Rachou, Fundação Oswaldo Cruz - Minas, Belo Horizonte30190-002, Brazil
- Department of Molecular Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA01605, USA
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
7
|
Singh MK, Bonnell VA, Tojal Da Silva I, Santiago VF, Moraes MS, Adderley J, Doerig C, Palmisano G, Llinas M, Garcia CRS. A Plasmodium falciparum MORC protein complex modulates epigenetic control of gene expression through interaction with heterochromatin. eLife 2024; 12:RP92201. [PMID: 39412522 PMCID: PMC11483127 DOI: 10.7554/elife.92201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Dynamic control of gene expression is critical for blood stage development of malaria parasites. Here, we used multi-omic analyses to investigate transcriptional regulation by the chromatin-associated microrchidia protein, MORC, during asexual blood stage development of the human malaria parasite Plasmodium falciparum. We show that PfMORC (PF3D7_1468100) interacts with a suite of nuclear proteins, including APETALA2 (ApiAP2) transcription factors (PfAP2-G5, PfAP2-O5, PfAP2-I, PF3D7_0420300, PF3D7_0613800, PF3D7_1107800, and PF3D7_1239200), a DNA helicase DS60 (PF3D7_1227100), and other chromatin remodelers (PfCHD1 and PfEELM2). Transcriptomic analysis of PfMORCHA-glmS knockdown parasites revealed 163 differentially expressed genes belonging to hypervariable multigene families, along with upregulation of genes mostly involved in host cell invasion. In vivo genome-wide chromatin occupancy analysis during both trophozoite and schizont stages of development demonstrates that PfMORC is recruited to repressed, multigene families, including the var genes in subtelomeric chromosomal regions. Collectively, we find that PfMORC is found in chromatin complexes that play a role in the epigenetic control of asexual blood stage transcriptional regulation and chromatin organization.
Collapse
Affiliation(s)
- Maneesh Kumar Singh
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São PauloSão PauloBrazil
| | - Victoria Ann Bonnell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University ParkHarrisburgUnited States
- Huck Institutes Center for Eukaryotic Gene Regulation, Pennsylvania State University, University ParkHarrisburgUnited States
- Huck Institutes Center for Malaria Research, Pennsylvania State University, University ParkHarrisburgUnited States
| | | | | | - Miriam Santos Moraes
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São PauloSão PauloBrazil
| | - Jack Adderley
- School of Health and Biomedical Sciences, RMIT UniversityBundooraAustralia
| | - Christian Doerig
- School of Health and Biomedical Sciences, RMIT UniversityBundooraAustralia
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Science, University of São PauloSão PauloBrazil
| | - Manuel Llinas
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University ParkHarrisburgUnited States
- Huck Institutes Center for Eukaryotic Gene Regulation, Pennsylvania State University, University ParkHarrisburgUnited States
- Huck Institutes Center for Malaria Research, Pennsylvania State University, University ParkHarrisburgUnited States
- Department of Chemistry, Pennsylvania State University, University ParkHarrisburgUnited States
| | - Celia RS Garcia
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São PauloSão PauloBrazil
| |
Collapse
|
8
|
MacAulay S, Cable J. Gyrodactylus in the spotlight: how exposure to light impacts disease and the feeding behavior of the freshwater tropical guppy (Poecilia reticulata). JOURNAL OF FISH BIOLOGY 2024; 105:682-690. [PMID: 38828698 DOI: 10.1111/jfb.15816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/05/2024]
Abstract
Artificial light at night (ALAN) negatively impacts organisms in many ways, from their feeding behaviors to their response and ability to deal with disease. Our knowledge of ALAN is focused on hosts, but we must also consider their parasites, which constitute half of all described animal species. Here, we assessed the impact of light exposure on a model host-parasite system (Poecilia reticulata and the ectoparasitic monogenean Gyrodactylus turnbulli). First, parasite-free fish were exposed to 12:12 h light:dark (control) or 24:0 h light:dark (ALAN) for 21 days followed by experimental infection. Second, naturally acquired G. turnbulli infections were monitored for 28 days during exposure of their hosts to a specified light regime (6:18 h, 12:12 h, or 24:0 h light:dark). Experimentally infected fish exposed to constant light had, on average, a greater maximum parasite burden than controls, but no other measured parasite metrics were impacted. Host feeding behavior was also significantly affected: fish under ALAN fed faster and took more bites than controls, whilst fish exposed to reduced light fed slower. Thus, ALAN can impact parasite burdens, even in the short term, and altering light conditions will impact fish feeding behavior. Such responses could initiate disease outbreaks or perturb food-webs with wider ecological impacts.
Collapse
Affiliation(s)
| | - Jo Cable
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
9
|
Dias BKM, Mohanty A, Garcia CRS. Melatonin as a Circadian Marker for Plasmodium Rhythms. Int J Mol Sci 2024; 25:7815. [PMID: 39063057 PMCID: PMC11277106 DOI: 10.3390/ijms25147815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Plasmodium, a digenetic parasite, requires a host and a vector for its life cycle completion. Most Plasmodium species display circadian rhythmicity during their intraerythrocytic cycle within the host, aiding in immune evasion. This rhythmicity, however, diminishes in in vitro cultures, highlighting the importance of host-derived signals for synchronizing the parasite's asexual cycle. Studies indicate a species-specific internal clock in Plasmodium, dependent on these host signals. Melatonin, a hormone the pineal gland produces under circadian regulation, impacts various physiological functions and is extensively reviewed as the primary circadian marker affecting parasite rhythms. Research suggests that melatonin facilitates synchronization through the PLC-IP3 signaling pathway, activating phospholipase C, which triggers intracellular calcium release and gene expression modulation. This evidence strongly supports the role of melatonin as a key circadian marker for parasite synchronization, presenting new possibilities for targeting the melatonin pathway when developing novel therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Célia R. S. Garcia
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, SP, Brazil; (B.K.M.D.); (A.M.)
| |
Collapse
|
10
|
Holland JG, Prior KF, O'Donnell AJ, Reece SE. Testing the evolutionary drivers of malaria parasite rhythms and their consequences for host-parasite interactions. Evol Appl 2024; 17:e13752. [PMID: 39006006 PMCID: PMC11246599 DOI: 10.1111/eva.13752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Undertaking certain activities at the time of day that maximises fitness is assumed to explain the evolution of circadian clocks. Organisms often use daily environmental cues such as light and food availability to set the timing of their clocks. These cues may be the environmental rhythms that ultimately determine fitness, act as proxies for the timing of less tractable ultimate drivers, or are used simply to maintain internal synchrony. While many pathogens/parasites undertake rhythmic activities, both the proximate and ultimate drivers of their rhythms are poorly understood. Explaining the roles of rhythms in infections offers avenues for novel interventions to interfere with parasite fitness and reduce the severity and spread of disease. Here, we perturb several rhythms in the hosts of malaria parasites to investigate why parasites align their rhythmic replication to the host's feeding-fasting rhythm. We manipulated host rhythms governed by light, food or both, and assessed the fitness implications for parasites, and the consequences for hosts, to test which host rhythms represent ultimate drivers of the parasite's rhythm. We found that alignment with the host's light-driven rhythms did not affect parasite fitness metrics. In contrast, aligning with the timing of feeding-fasting rhythms may be beneficial for the parasite, but only when the host possess a functional canonical circadian clock. Because parasites in clock-disrupted hosts align with the host's feeding-fasting rhythms and yet derive no apparent benefit, our results suggest cue(s) from host food act as a proxy rather than being a key selective driver of the parasite's rhythm. Alternatively, parasite rhythmicity may only be beneficial because it promotes synchrony between parasite cells and/or allows parasites to align to the biting rhythms of vectors. Our results also suggest that interventions can disrupt parasite rhythms by targeting the proxies or the selective factors driving them without impacting host health.
Collapse
Affiliation(s)
- Jacob G. Holland
- Institute of Ecology and EvolutionUniversity of EdinburghEdinburghUK
| | | | | | - Sarah E. Reece
- Institute of Ecology and EvolutionUniversity of EdinburghEdinburghUK
- Institute of Immunology and Infection ResearchUniversity of EdinburghEdinburghUK
| |
Collapse
|
11
|
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
|
12
|
Owolabi ATY, Schneider P, Reece SE. Virulence is associated with daily rhythms in the within-host replication of the malaria parasite Plasmodium chabaudi. Evol Appl 2024; 17:e13696. [PMID: 38721594 PMCID: PMC11078297 DOI: 10.1111/eva.13696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 04/10/2024] [Indexed: 01/06/2025] Open
Abstract
Most malaria (Plasmodium spp.) parasite species undergo asexual replication synchronously within the red blood cells of their vertebrate host. Rhythmicity in this intraerythrocytic developmental cycle (IDC) enables parasites to maximise exploitation of the host and align transmission activities with the time of day that mosquito vectors blood feed. The IDC is also responsible for the major pathologies associated with malaria, and plasticity in the parasite's rhythm can confer tolerance to antimalarial drugs. Both the severity of infection (virulence) and synchrony of the IDC vary across species and between genotypes of Plasmodium; however, this variation is poorly understood. The theory predicts that virulence and IDC synchrony are negatively correlated, and we tested this hypothesis using two closely related genotypes of the rodent malaria model Plasmodium chabaudi that differ markedly in virulence. We also test the predictions that, in response to perturbations to the timing (phase) of the IDC schedule relative to the phase of host rhythms (misalignment), the virulent parasite genotype recovers the correct phase relationship faster, incurs less fitness losses and so hosts benefit less from misalignment when infected with a virulent genotype. Our predictions are partially supported by results suggesting that the virulent parasite genotype is less synchronous in some circumstances and recovers faster from misalignment. While hosts were less anaemic when infected by misaligned parasites, the extent of this benefit did not depend on parasite virulence. Overall, our results suggest that interventions to perturb the alignment between the IDC schedule, and host rhythms and increase synchrony between parasites within each IDC, could alleviate disease symptoms. However, virulent parasites, which are better at withstanding conventional antimalarial treatment, would also be intrinsically better able to tolerate such interventions.
Collapse
Affiliation(s)
- Alíz T. Y. Owolabi
- School of Biological Sciences, Institute of Ecology and EvolutionUniversity of EdinburghEdinburghUK
| | - Petra Schneider
- School of Biological Sciences, Institute of Ecology and EvolutionUniversity of EdinburghEdinburghUK
| | - Sarah E. Reece
- School of Biological Sciences, Institute of Ecology and EvolutionUniversity of EdinburghEdinburghUK
| |
Collapse
|
13
|
Dwivedi SL, Quiroz LF, Spillane C, Wu R, Mattoo AK, Ortiz R. Unlocking allelic variation in circadian clock genes to develop environmentally robust and productive crops. PLANTA 2024; 259:72. [PMID: 38386103 PMCID: PMC10884192 DOI: 10.1007/s00425-023-04324-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/24/2023] [Indexed: 02/23/2024]
Abstract
MAIN CONCLUSION Molecular mechanisms of biological rhythms provide opportunities to harness functional allelic diversity in core (and trait- or stress-responsive) oscillator networks to develop more climate-resilient and productive germplasm. The circadian clock senses light and temperature in day-night cycles to drive biological rhythms. The clock integrates endogenous signals and exogenous stimuli to coordinate diverse physiological processes. Advances in high-throughput non-invasive assays, use of forward- and inverse-genetic approaches, and powerful algorithms are allowing quantitation of variation and detection of genes associated with circadian dynamics. Circadian rhythms and phytohormone pathways in response to endogenous and exogenous cues have been well documented the model plant Arabidopsis. Novel allelic variation associated with circadian rhythms facilitates adaptation and range expansion, and may provide additional opportunity to tailor climate-resilient crops. The circadian phase and period can determine adaptation to environments, while the robustness in the circadian amplitude can enhance resilience to environmental changes. Circadian rhythms in plants are tightly controlled by multiple and interlocked transcriptional-translational feedback loops involving morning (CCA1, LHY), mid-day (PRR9, PRR7, PRR5), and evening (TOC1, ELF3, ELF4, LUX) genes that maintain the plant circadian clock ticking. Significant progress has been made to unravel the functions of circadian rhythms and clock genes that regulate traits, via interaction with phytohormones and trait-responsive genes, in diverse crops. Altered circadian rhythms and clock genes may contribute to hybrid vigor as shown in Arabidopsis, maize, and rice. Modifying circadian rhythms via transgenesis or genome-editing may provide additional opportunities to develop crops with better buffering capacity to environmental stresses. Models that involve clock gene‒phytohormone‒trait interactions can provide novel insights to orchestrate circadian rhythms and modulate clock genes to facilitate breeding of all season crops.
Collapse
Affiliation(s)
| | - Luis Felipe Quiroz
- Agriculture and Bioeconomy Research Centre, Ryan Institute, University of Galway, University Road, Galway, H91 REW4, Ireland
| | - Charles Spillane
- Agriculture and Bioeconomy Research Centre, Ryan Institute, University of Galway, University Road, Galway, H91 REW4, Ireland.
| | - Rongling Wu
- Beijing Yanqi Lake Institute of Mathematical Sciences and Applications, Beijing, 101408, China
| | - Autar K Mattoo
- USDA-ARS, Sustainable Agricultural Systems Laboratory, Beltsville, MD, 20705-2350, USA
| | - Rodomiro Ortiz
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Sundsvagen, 10, Box 190, SE 23422, Lomma, Sweden.
| |
Collapse
|
14
|
Borenstein JT, Cummins G, Dutta A, Hamad E, Hughes MP, Jiang X, Lee HH, Lei KF, Tang XS, Zheng Y, Chen J. Bionanotechnology and bioMEMS (BNM): state-of-the-art applications, opportunities, and challenges. LAB ON A CHIP 2023; 23:4928-4949. [PMID: 37916434 DOI: 10.1039/d3lc00296a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
The development of micro- and nanotechnology for biomedical applications has defined the cutting edge of medical technology for over three decades, as advancements in fabrication technology developed originally in the semiconductor industry have been applied to solving ever-more complex problems in medicine and biology. These technologies are ideally suited to interfacing with life sciences, since they are on the scale lengths as cells (microns) and biomacromolecules (nanometers). In this paper, we review the state of the art in bionanotechnology and bioMEMS (collectively BNM), including developments and challenges in the areas of BNM, such as microfluidic organ-on-chip devices, oral drug delivery, emerging technologies for managing infectious diseases, 3D printed microfluidic devices, AC electrokinetics, flexible MEMS devices, implantable microdevices, paper-based microfluidic platforms for cellular analysis, and wearable sensors for point-of-care testing.
Collapse
Affiliation(s)
| | - Gerard Cummins
- School of Engineering, University of Birmingham, Edgbaston, B15 2TT, UK.
| | - Abhishek Dutta
- Department of Electrical & Computer Engineering, University of Connecticut, USA.
| | - Eyad Hamad
- Biomedical Engineering Department, School of Applied Medical Sciences, German Jordanian University, Amman, Jordan.
| | - Michael Pycraft Hughes
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates.
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, China.
| | - Hyowon Hugh Lee
- Weldon School of Biomedical Engineering, Center for Implantable Devices, Purdue University, West Lafayette, IN, USA.
| | | | | | | | - Jie Chen
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
15
|
Kioko M, Pance A, Mwangi S, Goulding D, Kemp A, Rono M, Ochola-Oyier LI, Bull PC, Bejon P, Rayner JC, Abdi AI. Extracellular vesicles could be a putative posttranscriptional regulatory mechanism that shapes intracellular RNA levels in Plasmodium falciparum. Nat Commun 2023; 14:6447. [PMID: 37833314 PMCID: PMC10575976 DOI: 10.1038/s41467-023-42103-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Plasmodium falciparum secretes extracellular vesicles (PfEVs) that contain parasite-derived RNA. However, the significance of the secreted RNA remains unexplored. Here, we compare secreted and intracellular RNA from asexual cultures of six P. falciparum lines. We find that secretion of RNA via extracellular vesicles is not only periodic throughout the asexual intraerythrocytic developmental cycle but is also highly conserved across P. falciparum isolates. We further demonstrate that the phases of RNA secreted via extracellular vesicles are discernibly shifted compared to those of the intracellular RNA within the secreting whole parasite. Finally, transcripts of genes with no known function during the asexual intraerythrocytic developmental cycle are enriched in PfEVs compared to the whole parasite. We conclude that the secretion of extracellular vesicles could be a putative posttranscriptional RNA regulation mechanism that is part of or synergise the classic RNA decay processes to maintain intracellular RNA levels in P. falciparum.
Collapse
Affiliation(s)
- Mwikali Kioko
- Bioscience Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Open University, Milton Keynes, UK
| | - Alena Pance
- Pathogens and Microbes Programme, Wellcome Sanger Institute, Cambridge, UK
- School of Life and Medical Science, University of Hertfordshire, Hatfield, UK
| | - Shaban Mwangi
- Bioscience Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - David Goulding
- Pathogens and Microbes Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Alison Kemp
- Cambridge Institute of Medical Research, University of Cambridge, Cambridge, UK
| | - Martin Rono
- Bioscience Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Pwani University Biosciences Research Centre, Pwani University, Kilifi, Kenya
| | | | - Pete C Bull
- Bioscience Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Philip Bejon
- Bioscience Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Julian C Rayner
- Cambridge Institute of Medical Research, University of Cambridge, Cambridge, UK
| | - Abdirahman I Abdi
- Bioscience Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.
- Pwani University Biosciences Research Centre, Pwani University, Kilifi, Kenya.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
16
|
Xiong LI, Garfinkel A. Are physiological oscillations physiological? J Physiol 2023. [PMID: 37622389 DOI: 10.1113/jp285015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023] Open
Abstract
Despite widespread and striking examples of physiological oscillations, their functional role is often unclear. Even glycolysis, the paradigm example of oscillatory biochemistry, has seen questions about its oscillatory function. Here, we take a systems approach to argue that oscillations play critical physiological roles, such as enabling systems to avoid desensitization, to avoid chronically high and therefore toxic levels of chemicals, and to become more resistant to noise. Oscillation also enables complex physiological systems to reconcile incompatible conditions such as oxidation and reduction, by cycling between them, and to synchronize the oscillations of many small units into one large effect. In pancreatic β-cells, glycolytic oscillations synchronize with calcium and mitochondrial oscillations to drive pulsatile insulin release, critical for liver regulation of glucose. In addition, oscillation can keep biological time, essential for embryonic development in promoting cell diversity and pattern formation. The functional importance of oscillatory processes requires a re-thinking of the traditional doctrine of homeostasis, holding that physiological quantities are maintained at constant equilibrium values, a view that has largely failed in the clinic. A more dynamic approach will initiate a paradigm shift in our view of health and disease. A deeper look into the mechanisms that create, sustain and abolish oscillatory processes requires the language of nonlinear dynamics, well beyond the linearization techniques of equilibrium control theory. Nonlinear dynamics enables us to identify oscillatory ('pacemaking') mechanisms at the cellular, tissue and system levels.
Collapse
Affiliation(s)
- Lingyun Ivy Xiong
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Alan Garfinkel
- Departments of Medicine (Cardiology) and Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Rankawat S, Kundal K, Chakraborty S, Kumar R, Ray S. A comprehensive rhythmicity analysis of host proteins and immune factors involved in malaria pathogenesis to decipher the importance of host circadian clock in malaria. Front Immunol 2023; 14:1210299. [PMID: 37638001 PMCID: PMC10449258 DOI: 10.3389/fimmu.2023.1210299] [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: 04/24/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Background Circadian rhythms broadly impact human health by regulating our daily physiological and metabolic processes. The circadian clocks substantially regulate our immune responses and susceptibility to infections. Malaria parasites have intrinsic molecular oscillations and coordinate their infection cycle with host rhythms. Considering the cyclical nature of malaria, a clear understanding of the circadian regulations in malaria pathogenesis and host responses is of immense importance. Methods We have thoroughly investigated the transcript level rhythmic patterns in blood proteins altered in falciparum and vivax malaria and malaria-related immune factors in mice, baboons, and humans by analyzing datasets from published literature and comprehensive databases. Using the Metascape and DAVID platforms, we analyzed Gene Ontology terms and physiological pathways associated with the rhythmic malaria-associated host immune factors. Results We observed that almost 50% of the malaria-associated host immune factors are rhythmic in mice and humans. Overlapping rhythmic genes identified in mice, baboons, and humans, exhibited enrichment (Q < 0.05, fold-enrichment > 5) of multiple physiological pathways essential for host immune and defense response, including cytokine production, leukocyte activation, cellular defense, and response, regulation of kinase activity, B-cell receptor signaling pathway, and cellular response to cytokine stimulus. Conclusions Our analysis indicates a robust circadian regulation on multiple interconnected host response pathways and immunological networks in malaria, evident from numerous rhythmic genes involved in those pathways. Host immune rhythms play a vital role in the temporal regulation of host-parasite interactions and defense machinery in malaria.
Collapse
Affiliation(s)
| | | | | | | | - Sandipan Ray
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| |
Collapse
|
18
|
Kobayashi Y, Komatsuya K, Imamura S, Nozaki T, Watanabe YI, Sato S, Dodd AN, Kita K, Tanaka K. Coordination of apicoplast transcription in a malaria parasite by internal and host cues. Proc Natl Acad Sci U S A 2023; 120:e2214765120. [PMID: 37406097 PMCID: PMC10334805 DOI: 10.1073/pnas.2214765120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 05/25/2023] [Indexed: 07/07/2023] Open
Abstract
The malaria parasite Plasmodium falciparum has a nonphotosynthetic plastid called the apicoplast, which contains its own genome. Regulatory mechanisms for apicoplast gene expression remain poorly understood, despite this organelle being crucial for the parasite life cycle. Here, we identify a nuclear-encoded apicoplast RNA polymerase σ subunit (sigma factor) which, along with the α subunit, appears to mediate apicoplast transcript accumulation. This has a periodicity reminiscent of parasite circadian or developmental control. Expression of the apicoplast subunit gene, apSig, together with apicoplast transcripts, increased in the presence of the blood circadian signaling hormone melatonin. Our data suggest that the host circadian rhythm is integrated with intrinsic parasite cues to coordinate apicoplast genome transcription. This evolutionarily conserved regulatory system might be a future target for malaria treatment.
Collapse
Affiliation(s)
- Yuki Kobayashi
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama226-8503, Japan
| | - Keisuke Komatsuya
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
- Laboratory of Biomembrane, Tokyo Metropolitan Institute of Medical Science, Tokyo156-8506, Japan
| | - Sousuke Imamura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama226-8503, Japan
- Space Environment and Energy Laboratories, Nippon Telegraph and Telephone Corporation, Tokyo180-8585, Japan
| | - Tomoyoshi Nozaki
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| | - Yoh-ichi Watanabe
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| | - Shigeharu Sato
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama226-8503, Japan
- Department of Pathology and Microbiology, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah88400, Malaysia
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah88400, Malaysia
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki852-8523, Japan
| | - Antony N. Dodd
- Department of Cell and Developmental Biology, John Innes Centre, NorwichNR4 7RU, United Kingdom
| | - Kiyoshi Kita
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki852-8523, Japan
- Department of Host-Defense Biochemistry, Institute of Tropical Medicine, Nagasaki University, Nagasaki852-8523, Japan
| | - Kan Tanaka
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama226-8503, Japan
| |
Collapse
|
19
|
Basco LK. Cultivation of Asexual Intraerythrocytic Stages of Plasmodium falciparum. Pathogens 2023; 12:900. [PMID: 37513747 PMCID: PMC10384318 DOI: 10.3390/pathogens12070900] [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: 05/17/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Successfully developed in 1976, the continuous in vitro culture of Plasmodium falciparum has many applications in the field of malaria research. It has become an important experimental model that directly uses a human pathogen responsible for a high prevalence of morbidity and mortality in many parts of the world and is a major source of biological material for immunological, biochemical, molecular, and pharmacological studies. Until present, the basic techniques described by Trager and Jensen and Haynes et al. remain unchanged in many malaria research laboratories. Nonetheless, different factors, including culture media, buffers, serum substitutes and supplements, sources of erythrocytes, and conditions of incubation (especially oxygen concentration), have been modified by different investigators to adapt the original technique in their laboratories or enhance the in vitro growth of the parasites. The possible effects and benefits of these modifications for the continuous cultivation of asexual intraerythrocytic stages of P. falciparum, as well as future challenges in developing a serum-free cultivation system and axenic cultures, are discussed.
Collapse
Affiliation(s)
- Leonardo K Basco
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Service de Santé des Armées (SSA), Unité Mixte de Recherche (UMR) Vecteurs-Infections Tropicales et Méditerranéennes (VITROME), 13005 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
| |
Collapse
|
20
|
Kuehnel RM, Ganga E, Balestra AC, Suarez C, Wyss M, Klages N, Brusini L, Maco B, Brancucci N, Voss TS, Soldati D, Brochet M. A Plasmodium membrane receptor platform integrates cues for egress and invasion in blood forms and activation of transmission stages. SCIENCE ADVANCES 2023; 9:eadf2161. [PMID: 37327340 PMCID: PMC10275601 DOI: 10.1126/sciadv.adf2161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 05/11/2023] [Indexed: 06/18/2023]
Abstract
Critical events in the life cycle of malaria-causing parasites depend on cyclic guanosine monophosphate homeostasis by guanylyl cyclases (GCs) and phosphodiesterases, including merozoite egress or invasion of erythrocytes and gametocyte activation. These processes rely on a single GCα, but in the absence of known signaling receptors, how this pathway integrates distinct triggers is unknown. We show that temperature-dependent epistatic interactions between phosphodiesterases counterbalance GCα basal activity preventing gametocyte activation before mosquito blood feed. GCα interacts with two multipass membrane cofactors in schizonts and gametocytes: UGO (unique GC organizer) and SLF (signaling linking factor). While SLF regulates GCα basal activity, UGO is essential for GCα up-regulation in response to natural signals inducing merozoite egress and gametocyte activation. This work identifies a GC membrane receptor platform that senses signals triggering processes specific to an intracellular parasitic lifestyle, including host cell egress and invasion to ensure intraerythrocytic amplification and transmission to mosquitoes.
Collapse
Affiliation(s)
- Ronja Marie Kuehnel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 12111 Geneva, Switzerland
| | - Emma Ganga
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 12111 Geneva, Switzerland
| | - Aurélia C. Balestra
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 12111 Geneva, Switzerland
| | - Catherine Suarez
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 12111 Geneva, Switzerland
| | - Matthias Wyss
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Natacha Klages
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 12111 Geneva, Switzerland
| | - Lorenzo Brusini
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 12111 Geneva, Switzerland
| | - Bohumil Maco
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 12111 Geneva, Switzerland
| | - Nicolas Brancucci
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Till S. Voss
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Dominique Soldati
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 12111 Geneva, Switzerland
| | - Mathieu Brochet
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 12111 Geneva, Switzerland
| |
Collapse
|
21
|
Motta FC, McGoff K, Moseley RC, Cho CY, Kelliher CM, Smith LM, Ortiz MS, Leman AR, Campione SA, Devos N, Chaorattanakawee S, Uthaimongkol N, Kuntawunginn W, Thongpiam C, Thamnurak C, Arsanok M, Wojnarski M, Vanchayangkul P, Boonyalai N, Smith PL, Spring MD, Jongsakul K, Chuang I, Harer J, Haase SB. The parasite intraerythrocytic cycle and human circadian cycle are coupled during malaria infection. Proc Natl Acad Sci U S A 2023; 120:e2216522120. [PMID: 37279274 PMCID: PMC10268210 DOI: 10.1073/pnas.2216522120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/09/2023] [Indexed: 06/08/2023] Open
Abstract
During infections with the malaria parasites Plasmodium vivax, patients exhibit rhythmic fevers every 48 h. These fever cycles correspond with the time the parasites take to traverse the intraerythrocytic cycle (IEC). In other Plasmodium species that infect either humans or mice, the IEC is likely guided by a parasite-intrinsic clock [Rijo-Ferreiraet al., Science 368, 746-753 (2020); Smith et al., Science 368, 754-759 (2020)], suggesting that intrinsic clock mechanisms may be a fundamental feature of malaria parasites. Moreover, because Plasmodium cycle times are multiples of 24 h, the IECs may be coordinated with the host circadian clock(s). Such coordination could explain the synchronization of the parasite population in the host and enable alignment of IEC and circadian cycle phases. We utilized an ex vivo culture of whole blood from patients infected with P. vivax to examine the dynamics of the host circadian transcriptome and the parasite IEC transcriptome. Transcriptome dynamics revealed that the phases of the host circadian cycle and the parasite IEC are correlated across multiple patients, showing that the cycles are phase coupled. In mouse model systems, host-parasite cycle coupling appears to provide a selective advantage for the parasite. Thus, understanding how host and parasite cycles are coupled in humans could enable antimalarial therapies that disrupt this coupling.
Collapse
Affiliation(s)
- Francis C. Motta
- Department of Mathematical Sciences, Florida Atlantic University, Boca Raton, FL33431
| | - Kevin McGoff
- Department of Mathematics and Statistics, University of North Carolina, Charlotte, NC28223
| | | | - Chun-Yi Cho
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA94143
| | - Christina M. Kelliher
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH03755
| | | | | | | | | | | | - Suwanna Chaorattanakawee
- Department of Parasitology and Entomology, Faculty of Public Health, Mahidol University, Bangkok10400, Thailand
| | | | | | - Chadin Thongpiam
- US-Armed Forces Research Institute of Medical Sciences, Bangkok10400, Thailand
| | | | - Montri Arsanok
- US-Armed Forces Research Institute of Medical Sciences, Bangkok10400, Thailand
| | | | | | - Nonlawat Boonyalai
- US-Armed Forces Research Institute of Medical Sciences, Bangkok10400, Thailand
| | - Philip L. Smith
- U.S. Military HIV Research Program Walter Reed Army Institute of Research, Bethesda, MD20817
| | - Michele D. Spring
- US-Armed Forces Research Institute of Medical Sciences, Bangkok10400, Thailand
| | - Krisada Jongsakul
- US-Armed Forces Research Institute of Medical Sciences, Bangkok10400, Thailand
| | - Ilin Chuang
- US Naval Medical Research Center-Asia in Singapore, Assigned to Armed Forces Research Institute of Medical Sciences, Bangkok10400, Thailand
| | - John Harer
- Geometric Data Analytics, Durham, NC27701
| | - Steven B. Haase
- Department of Biology, Duke University, Durham, NC27708
- Department of Medicine Duke University, Durham, NC27710
| |
Collapse
|
22
|
Platon L, Baker DA, Ménard D. Modified Plasmodium falciparum Ring-Stage Survival Assay with ML10 Kinase Inhibitor. Antimicrob Agents Chemother 2023; 67:e0001723. [PMID: 37098950 PMCID: PMC10190288 DOI: 10.1128/aac.00017-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/10/2023] [Indexed: 04/27/2023] Open
Abstract
The ring-stage survival assay is the reference assay to measure in vitro Plasmodium falciparum artemisinin partial resistance. The main challenge of the standard protocol is to generate 0-to-3-h postinvasion ring stages (the stage least susceptible to artemisinin) from schizonts obtained by sorbitol treatment and Percoll gradient. We report here a modified protocol facilitating the production of synchronized schizonts when multiple strains are tested simultaneously, by using ML10, a protein kinase inhibitor, that reversibly blocks merozoite egress.
Collapse
Affiliation(s)
- Lucien Platon
- Institut Pasteur, Université Paris Cité, Malaria Genetics and Resistance Unit, INSERM U1201, Paris, France
- Sorbonne Université, Paris, France
- Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, Paris, France
| | - David A. Baker
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Didier Ménard
- Institut Pasteur, Université Paris Cité, Malaria Genetics and Resistance Unit, INSERM U1201, Paris, France
- Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, Paris, France
- Université de Strasbourg, Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, Strasbourg, France
- CHU Strasbourg, Laboratory of Parasitology and Medical Mycology, Strasbourg, France
| |
Collapse
|
23
|
Asher G, Zhu B. Beyond circadian rhythms: emerging roles of ultradian rhythms in control of liver functions. Hepatology 2023; 77:1022-1035. [PMID: 35591797 PMCID: PMC9674798 DOI: 10.1002/hep.32580] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/08/2022]
Abstract
The mammalian liver must cope with various metabolic and physiological changes that normally recur every day and primarily stem from daily cycles of rest-activity and fasting-feeding. Although a large body of evidence supports the reciprocal regulation of circadian rhythms and liver function, the research on the hepatic ultradian rhythms have largely been lagging behind. However, with the advent of more cost-effective high-throughput omics technologies, high-resolution time-lapse imaging, and more robust and powerful mathematical tools, several recent studies have shed new light on the presence and functions of hepatic ultradian rhythms. In this review, we will first very briefly discuss the basic principles of circadian rhythms, and then cover in greater details the recent literature related to ultradian rhythms. Specifically, we will highlight the prevalence and mechanisms of hepatic 12-h rhythms, and 8-h rhythms, which cycle at the second and third harmonics of circadian frequency. Finally, we also refer to ultradian rhythms with other frequencies and examine the limitations of the current approaches as well as the challenges related to identifying ultradian rhythm and addressing their molecular underpinnings.
Collapse
Affiliation(s)
- Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Bokai Zhu
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pennsylvania, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
24
|
Ramesh A, Hall SR. Niche theory for within-host parasite dynamics: Analogies to food web modules via feedback loops. Ecol Lett 2023; 26:351-368. [PMID: 36632705 DOI: 10.1111/ele.14142] [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: 08/18/2021] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 01/13/2023]
Abstract
Why do parasites exhibit a wide dynamical range within their hosts? For instance, why does infecting dose either lead to infection or immune clearance? Why do some parasites exhibit boom-bust, oscillatory dynamics? What maintains parasite diversity, that is coinfection v single infection due to exclusion or priority effects? For insights on parasite dose, dynamics and diversity governing within-host infection, we turn to niche models. An omnivory food web model (IGP) blueprints one parasite competing with immune cells for host energy (PIE). Similarly, a competition model (keystone predation, KP) mirrors a new coinfection model (2PIE). We then drew analogies between models using feedback loops. The following three points arise: first, like in IGP, parasites oscillate when longer loops through parasites, immune cells and resource regulate parasite growth. Shorter, self-limitation loops (involving resources and enemies) stabilise those oscillations. Second, IGP can produce priority effects that resemble immune clearance. But, despite comparable loop structure, PIE cannot due to constraints imposed by production of immune cells. Third, despite somewhat different loop structure, KP and 2PIE share apparent and resource competition mechanisms that produce coexistence (coinfection) or priority effects of prey or parasites. Together, this mechanistic niche framework for within-host dynamics offers new perspective to improve individual health.
Collapse
|
25
|
Huang H, Mehta A, Kalmanovich J, Anand A, Bejarano MC, Garg T, Khan N, Tonpouwo GK, Shkodina AD, Bardhan M. Immunological and inflammatory effects of infectious diseases in circadian rhythm disruption and future therapeutic directions. Mol Biol Rep 2023; 50:3739-3753. [PMID: 36656437 PMCID: PMC9851103 DOI: 10.1007/s11033-023-08276-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/11/2023] [Indexed: 01/20/2023]
Abstract
BACKGROUND Circadian rhythm is characterised by daily variations in biological activity to align with the light and dark cycle. These diurnal variations, in turn, influence physiological functions such as blood pressure, temperature, and sleep-wake cycle. Though it is well established that the circadian pathway is linked to pro-inflammatory responses and circulating immune cells, its association with infectious diseases is widely unknown. OBJECTIVE This comprehensive review aims to describe the association between circadian rhythm and host immune response to various kinds of infection. METHODS We conducted a literature search in databases Pubmed/Medline and Science direct. Our paper includes a comprehensive analysis of findings from articles in English which was related to our hypothesis. FINDINGS Molecular clocks determine circadian rhythm disruption in response to infection, influencing the host's response toward infection. Moreover, there is a complex interplay with intrinsic oscillators of pathogens and the influence of specific infectious processes on the CLOCK: BMAL1 pathway. Such mechanisms vary for bacterial and viral infections, both well studied in the literature. However, less is known about the association of parasitic infections and fungal pathogens with circadian rhythm modulation. CONCLUSION It is shown that bidirectional relationships exist between circadian rhythm disruption and infectious process, which contains interplay between the host's and pathogens' circadian oscillator, immune response, and the influence of specific infectious. Further studies exploring the modulations of circadian rhythm and immunity can offer novel explanations of different susceptibilities to infection and can lead to therapeutic avenues in circadian immune modulation of infectious diseases.
Collapse
Affiliation(s)
- Helen Huang
- Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Aashna Mehta
- Faculty of Medicine, University of Debrecen, Debrecen, 4032 Hungary
| | | | - Ayush Anand
- B. P. Koirala Institute of Health Sciences, Dharan, Nepal
| | - Maria Chilo Bejarano
- Facultad de Ciencias de la Salud Humana, Universidad Autónoma Gabriel René Moreno, Santa Cruz de la Sierra, Bolivia
| | - Tulika Garg
- Government Medical College and Hospital, Chandigarh, India
| | - Nida Khan
- Jinnah Sindh Medical University, Karachi, Pakistan
| | - Gauvain Kankeu Tonpouwo
- Faculté de Médecine, Université de Lubumbashi, Plaine Tshombé, Lubumbashi, Democratic Republic of the Congo
| | | | - Mainak Bardhan
- ICMR-National Institute of Cholera and Enteric Diseases (NICED), Kolkata, India
| |
Collapse
|
26
|
Stewart LB, Freville A, Voss TS, Baker DA, Awandare GA, Conway DJ. Plasmodium falciparum Sexual Commitment Rate Variation among Clinical Isolates and Diverse Laboratory-Adapted Lines. Microbiol Spectr 2022; 10:e0223422. [PMID: 36409095 PMCID: PMC9769538 DOI: 10.1128/spectrum.02234-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/20/2022] [Indexed: 11/23/2022] Open
Abstract
Asexual blood-stage malaria parasites must produce sexual progeny to infect mosquitoes. It is important to understand the scope and causes of intraspecific variation in sexual commitment rates, particularly for the major human parasite P. falciparum. First, two alternative assay methods of measuring sexual commitment were compared to test a genetically modified P. falciparum line with elevated commitment rates inducible by overexpression of GDV1. The methods yielded correlated measurements with higher sensitivity and precision being achieved by one employing detection of the early gametocyte differentiation marker Pfs16. Thus, this was used to survey a diverse range of parasite lines and test each in multiple biological replicate assays in a serum-free medium supplemented with Albumax. There were differences among six recent clinical isolates from Ghana in their mean rates of sexual commitment per cycle, ranging from 3.3% to 12.2%. Among 13 diverse long-term laboratory-adapted lines, mean sexual commitment rates for most ranged from 4.7% to 13.4%, a few had lower rates with means from 0.3 to 1.6%, and one with a nonfunctional ap2-g gene always showed zero commitment. Among a subset of lines tested for the effects of exogenous choline to suppress commitment, there were significant differences. As expected, there was no effect in a line that had lost the gdv1 gene and that had generally low commitment, whereas the others showed quantitatively variable but significant responses to choline, suggesting potential trait variation. The results indicated the value of performing multiple replicate assays for understanding the variation of this key reproductive trait that likely affects transmission. IMPORTANCE Only sexual-stage malaria parasites are transmitted from human blood to mosquitoes. Thus, it is vital to understand variations in sexual commitment rates because these may be modifiable or susceptible to blocking. Two different methods of commitment rate measurement were first compared, demonstrating higher sensitivity and precision by the detection of an early differentiation marker, which was subsequently used to survey diverse lines. Clinical isolates from Ghana showed significant variation in mean per-cycle commitment rates and variation among biological replicates. Laboratory-adapted lines of diverse origins had a wider range with most being within the range observed for the clinical isolates, while a minority consistently had lower or zero rates. There was quantitative variation in the effects when adding choline to suppress commitment, indicating differing responsiveness of parasites to this environmental modification. Performing multiple assay replicates and comparisons of diverse isolates was important to understand this trait and its potential effects on transmission.
Collapse
Affiliation(s)
- Lindsay B. Stewart
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Aline Freville
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Till S. Voss
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basal, Switzerland
| | - David A. Baker
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Gordon A. Awandare
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - David J. Conway
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
27
|
Habtewold T, Tapanelli S, Masters EKG, Windbichler N, Christophides GK. The circadian clock modulates Anopheles gambiae infection with Plasmodium falciparum. PLoS One 2022; 17:e0278484. [PMID: 36454885 PMCID: PMC9714873 DOI: 10.1371/journal.pone.0278484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
Key behaviours, physiologies and gene expressions in Anopheles mosquitoes impact the transmission of Plasmodium. Such mosquito factors are rhythmic to closely follow diel rhythms. Here, we set to explore the impact of the mosquito circadian rhythm on the tripartite interaction between the vector, the parasite and the midgut microbiota, and investigate how this may affect the parasite infection outcomes. We assess Plasmodium falciparum infection prevalence and intensity, as a proxy for gametocyte infectivity, in Anopheles gambiae mosquitoes that received a gametocyte-containing bloodfeed and measure the abundance of the midgut microbiota at different times of the mosquito rearing light-dark cycle. Gametocyte infectivity is also compared in mosquitoes reared and maintained under a reversed light-dark regime. The effect of the circadian clock on the infection outcome is also investigated through silencing of the CLOCK gene that is central in the regulation of animal circadian rhythms. The results reveal that the A. gambiae circadian cycle plays a key role in the intensity of infection of P. falciparum gametocytes. We show that parasite gametocytes are more infectious during the night-time, where standard membrane feeding assays (SMFAs) at different time points in the mosquito natural circadian rhythm demonstrate that gametocytes are more infectious when ingested at midnight than midday. When mosquitoes were cultured under a reversed light/dark regime, disrupting their natural physiological homeostasis, and infected with P. falciparum at evening hours, the infection intensity and prevalence were significantly decreased. Similar results were obtained in mosquitoes reared under the standard light/dark regime upon silencing of CLOCK, a key regulator of the circadian rhythm, highlighting the importance of the circadian rhythm for the mosquito vectorial capacity. At that time, the mosquito midgut microbiota load is significantly reduced, while the expression of lysozyme C-1 (LYSC-1) is elevated, which is involved in both the immune response and microbiota digestion. We conclude that the tripartite interactions between the mosquito vector, the malaria parasite and the mosquito gut microbiota are finely tuned to support and maintain malaria transmission. Our data add to the knowledge framework required for designing appropriate and biologically relevant SMFA protocols.
Collapse
Affiliation(s)
- Tibebu Habtewold
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Sofia Tapanelli
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Ellen K. G. Masters
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Nikolai Windbichler
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | |
Collapse
|
28
|
Rijo-Ferreira F, Takahashi JS. Circadian rhythms in infectious diseases and symbiosis. Semin Cell Dev Biol 2022; 126:37-44. [PMID: 34625370 PMCID: PMC9183220 DOI: 10.1016/j.semcdb.2021.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
Timing is everything. Many organisms across the tree of life have evolved timekeeping mechanisms that regulate numerous of their cellular functions to optimize timing by anticipating changes in the environment. The specific environmental changes that are sensed depends on the organism. For animals, plants, and free-living microbes, environmental cues include light/dark cycles, daily temperature fluctuations, among others. In contrast, for a microbe that is never free-living, its rhythmic environment is its host's rhythmic biology. Here, we describe recent research on the interactions between hosts and microbes, from the perspective both of symbiosis as well as infections. In addition to describing the biology of the microbes, we focus specifically on how circadian clocks modulate these host-microbe interactions.
Collapse
Affiliation(s)
- Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
29
|
Hunt R, Cable J, Ellison A. Daily patterns in parasite processes: diel variation in fish louse transcriptomes. Int J Parasitol 2022; 52:509-518. [PMID: 35533730 DOI: 10.1016/j.ijpara.2022.04.001] [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: 12/15/2021] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 11/05/2022]
Abstract
Parasites, similar to all other organisms, time themselves to environmental cues using a molecular clock to generate and maintain rhythms. Chronotherapeutic (timed treatment) techniques based on such rhythms offer great potential for improving control of chronic, problematic parasites. Fish lice are a key disease threat in aquaculture, with current control insufficient. Assessing the rhythmicity of fish lice transcriptomes offers not only insight into the viability of chronotherapy, but the opportunity to identify new drug targets. Here, for the first known time in any crustacean parasite, diel changes in gene transcription are examined, revealing that approximately half of the Argulus foliaceus annotated transcriptome displays significant daily rhythmicity. We identified rhythmically transcribed putative clock genes including core clock/cycle and period/timeless pairs, alongside rhythms in feeding-associated genes and processes involving immune response, as well as fish louse drug targets. A substantial number of gene pathways showed peak transcription in hours immediately preceding onset of light, potentially in anticipation of peak host anti-parasite responses or in preparation for increased feeding activity. Genes related to immune haemocyte activity and chitin development were more highly transcribed 4 h post light onset, although inflammatory gene transcription was highest during dark periods. Our study provides an important resource for application of chronotherapy in fish lice; timed application could increase efficacy and/or reduce dose requirement, improving the current landscape of drug resistance and fish health while reducing the economic cost of infection.
Collapse
Affiliation(s)
- R Hunt
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom
| | - J Cable
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom
| | - A Ellison
- School of Natural Sciences, Bangor University, Deiniol Road, Bangor, Gwynedd, LL57 2UW, United Kingdom.
| |
Collapse
|
30
|
Foster GJ, Sievert MAC, Button-Simons K, Vendrely KM, Romero-Severson J, Ferdig MT. Cyclical regression covariates remove the major confounding effect of cyclical developmental gene expression with strain-specific drug response in the malaria parasite Plasmodium falciparum. BMC Genomics 2022; 23:180. [PMID: 35247977 PMCID: PMC8897900 DOI: 10.1186/s12864-021-08281-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 12/24/2021] [Indexed: 12/21/2022] Open
Abstract
Background The cyclical nature of gene expression in the intraerythrocytic development cycle (IDC) of the malaria parasite, Plasmodium falciparum, confounds the accurate detection of specific transcriptional differences, e.g. as provoked by the development of drug resistance. In lab-based studies, P. falciparum cultures are synchronized to remove this confounding factor, but the rapid detection of emerging resistance to artemisinin therapies requires rapid analysis of transcriptomes extracted directly from clinical samples. Here we propose the use of cyclical regression covariates (CRC) to eliminate the major confounding effect of developmentally driven transcriptional changes in clinical samples. We show that elimination of this confounding factor reduces both Type I and Type II errors and demonstrate the effectiveness of this approach using a published dataset of 1043 transcriptomes extracted directly from patient blood samples with different patient clearance times after treatment with artemisinin. Results We apply this method to two publicly available datasets and demonstrate its ability to reduce the confounding of differences in transcript levels due to misaligned intraerythrocytic development time. Adjusting the clinical 1043 transcriptomes dataset with CRC results in detection of fewer functional categories than previously reported from the same data set adjusted using other methods. We also detect mostly the same functional categories, but observe fewer genes within these categories. Finally, the CRC method identifies genes in a functional category that was absent from the results when the dataset was adjusted using other methods. Analysis of differential gene expression in the clinical data samples that vary broadly for developmental stage resulted in the detection of far fewer transcripts in fewer functional categories while, at the same time, identifying genes in two functional categories not present in the unadjusted data analysis. These differences are consistent with the expectation that CRC reduces both false positives and false negatives with the largest effect on datasets from samples with greater variance in developmental stage. Conclusions Cyclical regression covariates have immediate application to parasite transcriptome sequencing directly from clinical blood samples and to cost-constrained in vitro experiments. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08281-y.
Collapse
|
31
|
Else KJ, Gibbs JE. Parasites-The importance of time. Parasite Immunol 2022; 44:e12906. [PMID: 35092020 PMCID: PMC9286470 DOI: 10.1111/pim.12906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 11/30/2022]
Abstract
The special edition of Parasite Immunology 'Parasites-The importance of time' embraces the intersection between three established research disciplines-parasitology, immunology, and circadian biology. Each of these research areas has a longstanding history littered with landmark discoveries with the intersect between the three bringing exciting findings and new questions and perhaps even a greater sense of awe in terms of how parasites have evolved to interact and live with their hosts.
Collapse
Affiliation(s)
- Kathryn J. Else
- Lydia Becker Institute for Immunology and Inflammation & Centre for Biological TimingFaculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| | - Julie E. Gibbs
- Lydia Becker Institute for Immunology and Inflammation & Centre for Biological TimingFaculty of Biology Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
32
|
Hunter FK, Butler TD, Gibbs JE. Circadian rhythms in immunity and host-parasite interactions. Parasite Immunol 2022; 44:e12904. [PMID: 34971451 PMCID: PMC9285061 DOI: 10.1111/pim.12904] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 11/30/2022]
Abstract
The mammalian immune system adheres to a 24 h circadian schedule, exhibiting daily rhythmic patterns in homeostatic immune processes, such as immune cell trafficking, as well as the inflammatory response to infection. These diurnal rhythms are driven by endogenous molecular clocks within immune cells which are hierarchically coordinated by a light-entrained central clock in the suprachiasmatic nucleus of the hypothalamus and responsive to local rhythmic cues including temperature, hormones and feeding time. Circadian control of immunity may enable animals to anticipate daily pathogenic threat from parasites and gate the magnitude of the immune response, potentially enhancing fitness. However, parasites also strive for optimum fitness and some may have co-evolved to benefit from host circadian timing mechanisms, possibly via the parasites' own intrinsic molecular clocks. In this review, we summarize the current knowledge surrounding the influence of the circadian clock on the mammalian immune system and the host-parasitic interaction. We also discuss the potential for chronotherapeutic strategies in the treatment of parasitic diseases.
Collapse
Affiliation(s)
- Felicity K Hunter
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Thomas D Butler
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Julie E Gibbs
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
33
|
Carvalho Cabral P, Tekade K, Stegeman SK, Olivier M, Cermakian N. The involvement of host circadian clocks in the regulation of the immune response to parasitic infections in mammals. Parasite Immunol 2021; 44:e12903. [PMID: 34964129 DOI: 10.1111/pim.12903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 11/29/2022]
Abstract
Circadian rhythms are recurring variations of physiology with a period of ~24 hours, generated by circadian clocks located throughout the body. Studies have shown a circadian regulation of many aspects of immunity. Immune cells have intrinsic clock mechanisms, and innate and adaptive immune responses - such as leukocyte migration, magnitude of inflammation, cytokine production and cell differentiation - are under circadian control. This circadian regulation has consequences for infections including parasitic infections. In the context of Leishmania infection, the circadian clock within host immune cells modulates the magnitude of the infection and the inflammatory response triggered by the parasite. As for malaria, rhythms within the immune system were shown to impact the developmental cycles of Plasmodium parasites within red blood cells. Further, host circadian rhythms impact infections by multicellular parasites; for example, infection with helminth Trichuris muris shows different kinetics of worm expulsion depending on time of day of infection, a variation that depends on the dendritic cell clock. Although the research on the circadian control of immunity in the context of parasitic infections is in its infancy, the research reviewed here suggests a crucial involvement of host circadian rhythms in immunity on the development and progression of parasitic infections.
Collapse
Affiliation(s)
| | - Kimaya Tekade
- Douglas Research Centre, McGill University, Montreal, QC, H4H 1R3, Canada
| | - Sophia K Stegeman
- Douglas Research Centre, McGill University, Montreal, QC, H4H 1R3, Canada
| | - Martin Olivier
- Research Institute of the McGill University Health Center, McGill University, Montreal, QC, H4A 3J1, Canada
| | - Nicolas Cermakian
- Douglas Research Centre, McGill University, Montreal, QC, H4H 1R3, Canada
| |
Collapse
|
34
|
Rawlinson KA, Reid AJ, Lu Z, Driguez P, Wawer A, Coghlan A, Sankaranarayanan G, Buddenborg SK, Soria CD, McCarthy C, Holroyd N, Sanders M, Hoffmann KF, Wilcockson D, Rinaldi G, Berriman M. Daily rhythms in gene expression of the human parasite Schistosoma mansoni. BMC Biol 2021; 19:255. [PMID: 34852797 PMCID: PMC8638415 DOI: 10.1186/s12915-021-01189-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 11/10/2021] [Indexed: 12/13/2022] Open
Abstract
Background The consequences of the earth’s daily rotation have led to 24-h biological rhythms in most organisms. Even some parasites are known to have daily rhythms, which, when in synchrony with host rhythms, can optimise their fitness. Understanding these rhythms may enable the development of control strategies that take advantage of rhythmic vulnerabilities. Recent work on protozoan parasites has revealed 24-h rhythms in gene expression, drug sensitivity and the presence of an intrinsic circadian clock; however, similar studies on metazoan parasites are lacking. To address this, we investigated if a metazoan parasite has daily molecular oscillations, whether they reveal how these longer-lived organisms can survive host daily cycles over a lifespan of many years and if animal circadian clock genes are present and rhythmic. We addressed these questions using the human blood fluke Schistosoma mansoni that lives in the vasculature for decades and causes the tropical disease schistosomiasis. Results Using round-the-clock transcriptomics of male and female adult worms collected from experimentally infected mice, we discovered that ~ 2% of its genes followed a daily pattern of expression. Rhythmic processes included a stress response during the host’s active phase and a ‘peak in metabolic activity’ during the host’s resting phase. Transcriptional profiles in the female reproductive system were mirrored by daily patterns in egg laying (eggs are the main drivers of the host pathology). Genes cycling with the highest amplitudes include predicted drug targets and a vaccine candidate. These 24-h rhythms may be driven by host rhythms and/or generated by a circadian clock; however, orthologs of core clock genes are missing and secondary clock genes show no 24-h rhythmicity. Conclusions There are daily rhythms in the transcriptomes of adult S. mansoni, but they appear less pronounced than in other organisms. The rhythms reveal temporally compartmentalised internal processes and host interactions relevant to within-host survival and between-host transmission. Our findings suggest that if these daily rhythms are generated by an intrinsic circadian clock then the oscillatory mechanism must be distinct from that in other animals. We have shown which transcripts oscillate at this temporal scale and this will benefit the development and delivery of treatments against schistosomiasis. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01189-9.
Collapse
Affiliation(s)
| | - Adam J Reid
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Zhigang Lu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Patrick Driguez
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,King Abdullah University of Science and Technology, Thuwal, Makkah, Saudi Arabia
| | - Anna Wawer
- Institute of Biological, Environmental, and Rural Sciences, Aberystwyth University, Aberystwyth, UK
| | - Avril Coghlan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | | | | | | | - Nancy Holroyd
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Mandy Sanders
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Karl F Hoffmann
- Institute of Biological, Environmental, and Rural Sciences, Aberystwyth University, Aberystwyth, UK
| | - David Wilcockson
- Institute of Biological, Environmental, and Rural Sciences, Aberystwyth University, Aberystwyth, UK
| | - Gabriel Rinaldi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | |
Collapse
|
35
|
Borges-Pereira L, Dias BKM, Singh MK, Garcia CRS. Malaria parasites and circadian rhythm: New insights into an old puzzle. CURRENT RESEARCH IN MICROBIAL SCIENCES 2021; 2:100017. [PMID: 34841309 PMCID: PMC8610328 DOI: 10.1016/j.crmicr.2020.100017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/20/2020] [Accepted: 11/25/2020] [Indexed: 12/04/2022] Open
Abstract
Discuss molecular components for the coordination of circadian rhythm of malaria parasites inside the vertebrate host. Synthetic indole compounds show antimalarial activity in vitro against P.falciparum 3D7. Plasmodium falciparum synchronizes in cell culture upon melatonin treatment.
Collapse
Affiliation(s)
- Lucas Borges-Pereira
- Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| | - Bárbara K M Dias
- Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil.,Department of Parasitology, University of São Paulo, São Paulo, Brazil
| | - Maneesh Kumar Singh
- Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| | - Celia R S Garcia
- Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
O'Donnell AJ, Greischar MA, Reece SE. Mistimed malaria parasites re-synchronize with host feeding-fasting rhythms by shortening the duration of intra-erythrocytic development. Parasite Immunol 2021; 44:e12898. [PMID: 34778983 PMCID: PMC9285586 DOI: 10.1111/pim.12898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/24/2022]
Abstract
AIMS Malaria parasites exhibit daily rhythms in the intra-erythrocytic development cycle (IDC) that underpins asexual replication in the blood. The IDC schedule is aligned with the timing of host feeding-fasting rhythms. When the IDC schedule is perturbed to become mismatched to host rhythms, it readily reschedules but it is not known how. METHODS We intensively follow four groups of infections that have different temporal alignments between host rhythms and the IDC schedule for 10 days, before and after the peak in asexual densities. We compare how the duration, synchrony and timing of the IDC differs between parasites in control infections and those forced to reschedule by 12 hours and ask whether the density of parasites affects the rescheduling process. RESULTS AND CONCLUSIONS Our experiments reveal parasites shorten the IDC duration by 2-3 hours to become realigned to host feeding-fasting rhythms with 5-6 days, in a density-independent manner. Furthermore, parasites are able to reschedule without significant fitness costs for them or their hosts. Understanding the extent of, and limits on, plasticity in the IDC schedule may reveal targets for novel interventions, such as drugs to disrupt IDC regulation and preventing IDC dormancy conferring tolerance to existing drugs.
Collapse
Affiliation(s)
- Aidan J O'Donnell
- Institute of Evolutionary Biology, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Megan A Greischar
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| | - Sarah E Reece
- Institute of Evolutionary Biology, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
37
|
Impact of Sickle Cell Trait Hemoglobin on the Intraerythrocytic Transcriptional Program of Plasmodium falciparum. mSphere 2021; 6:e0075521. [PMID: 34668757 PMCID: PMC8527989 DOI: 10.1128/msphere.00755-21] [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] [Indexed: 11/20/2022] Open
Abstract
Sickle-trait hemoglobin (HbAS) confers nearly complete protection from severe, life-threatening falciparum malaria in African children. Despite this clear protection, the molecular mechanisms by which HbAS confers these protective phenotypes remain incompletely understood. As a forward genetic screen for aberrant parasite transcriptional responses associated with parasite neutralization in HbAS red blood cells (RBCs), we performed comparative transcriptomic analyses of Plasmodium falciparum in normal (HbAA) and HbAS erythrocytes during both in vitro cultivation of reference parasite strains and naturally occurring P. falciparum infections in Malian children with HbAA or HbAS. During in vitro cultivation, parasites matured normally in HbAS RBCs, and the temporal expression was largely unperturbed of the highly ordered transcriptional program that underlies the parasite’s maturation throughout the intraerythrocytic development cycle (IDC). However, differential expression analysis identified hundreds of transcripts aberrantly expressed in HbAS, largely occurring late in the IDC. Surprisingly, transcripts encoding members of the Maurer’s clefts were overexpressed in HbAS despite impaired parasite protein export in these RBCs, while parasites in HbAS RBCs underexpressed transcripts associated with the endoplasmic reticulum and those encoding serine repeat antigen proteases that promote parasite egress. Analyses of P. falciparum transcriptomes from 32 children with uncomplicated malaria identified stage-specific differential expression: among infections composed of ring-stage parasites, only cyclophilin 19B was underexpressed in children with HbAS, while trophozoite-stage infections identified a range of differentially expressed transcripts, including downregulation in HbAS of several transcripts associated with severe malaria in collateral studies. Collectively, our comparative transcriptomic screen in vitro and in vivo indicates that P. falciparum adapts to HbAS by altering its protein chaperone and folding machinery, oxidative stress response, and protein export machinery. Because HbAS consistently protects from severe P. falciparum, modulation of these responses may offer avenues by which to neutralize P. falciparum parasites. IMPORTANCE Sickle-trait hemoglobin (HbAS) confers nearly complete protection from severe, life-threatening malaria, yet the molecular mechanisms that underlie HbAS protection from severe malaria remain incompletely understood. Here, we used transcriptome sequencing (RNA-seq) to measure the impact of HbAS on the blood-stage transcriptome of Plasmodium falciparum in in vitro time series experiments and in vivo samples from natural infections. Our in vitro time series data reveal that, during its blood stage, P. falciparum’s gene expression in HbAS is impacted primarily through alterations in the abundance of gene products as opposed to variations in the timing of gene expression. Collectively, our in vitro and in vivo data indicate that P. falciparum adapts to HbAS by altering its protein chaperone and folding machinery, oxidative stress response, and protein export machinery. Due to the persistent association of HbAS and protection from severe disease, these processes that are modified in HbAS may offer strategies to neutralize P. falciparum.
Collapse
|
38
|
Prior KF, Middleton B, Owolabi AT, Westwood ML, Holland J, O'Donnell AJ, Blackman MJ, Skene DJ, Reece SE. Synchrony between daily rhythms of malaria parasites and hosts is driven by an essential amino acid. Wellcome Open Res 2021; 6:186. [PMID: 34805551 PMCID: PMC8577053.2 DOI: 10.12688/wellcomeopenres.16894.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Rapid asexual replication of blood stage malaria parasites is responsible for the severity of disease symptoms and fuels the production of transmission forms. Here, we demonstrate that a Plasmodium chabaudi's schedule for asexual replication can be orchestrated by isoleucine, a metabolite provided to the parasite in a periodic manner due to the host's rhythmic intake of food. Methods: We infect female C57BL/6 and Per1/2-null mice which have a disrupted canonical (transcription translation feedback loop, TTFL) clock with 1×10 5 red blood cells containing P. chabaudi (DK genotype). We perturb the timing of rhythms in asexual replication and host feeding-fasting cycles to identify nutrients with rhythms that match all combinations of host and parasite rhythms. We then test whether perturbing the availability of the best candidate nutrient in vitro changes the schedule for asexual development. Results: Our large-scale metabolomics experiment and follow up experiments reveal that only one metabolite - the amino acid isoleucine - fits criteria for a time-of-day cue used by parasites to set the schedule for replication. The response to isoleucine is a parasite strategy rather than solely the consequences of a constraint imposed by host rhythms, because unlike when parasites are deprived of other essential nutrients, they suffer no apparent costs from isoleucine withdrawal. Conclusions: Overall, our data suggest parasites can use the daily rhythmicity of blood-isoleucine concentration to synchronise asexual development with the availability of isoleucine, and potentially other resources, that arrive in the blood in a periodic manner due to the host's daily feeding-fasting cycle. Identifying both how and why parasites keep time opens avenues for interventions; interfering with the parasite's time-keeping mechanism may stall replication, increasing the efficacy of drugs and immune responses, and could also prevent parasites from entering dormancy to tolerate drugs.
Collapse
Affiliation(s)
- Kimberley F. Prior
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK,
| | - Benita Middleton
- School of Biosciences and Medicine, University of Surrey, Surrey, UK
| | - Alíz T.Y. Owolabi
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| | - Mary L. Westwood
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| | - Jacob Holland
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| | - Aidan J. O'Donnell
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, Francis Crick Institute, London, UK,Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Debra J. Skene
- School of Biosciences and Medicine, University of Surrey, Surrey, UK
| | - Sarah E. Reece
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
39
|
Prior KF, Middleton B, Owolabi AT, Westwood ML, Holland J, O'Donnell AJ, Blackman MJ, Skene DJ, Reece SE. Synchrony between daily rhythms of malaria parasites and hosts is driven by an essential amino acid. Wellcome Open Res 2021; 6:186. [PMID: 34805551 PMCID: PMC8577053 DOI: 10.12688/wellcomeopenres.16894.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2021] [Indexed: 11/20/2022] Open
Abstract
Background: Rapid asexual replication of blood stage malaria parasites is responsible for the severity of disease symptoms and fuels the production of transmission forms. Here, we demonstrate that the Plasmodium chabaudi's schedule for asexual replication can be orchestrated by isoleucine, a metabolite provided to the parasite in periodic manner due to the host's rhythmic intake of food. Methods: We infect female C57BL/6 and Per1/2-null TTFL clock-disrupted mice with 1×10 5 red blood cells containing P. chabaudi (DK genotype). We perturb the timing of rhythms in asexual replication and host feeding-fasting cycles to identify nutrients with rhythms that match all combinations of host and parasite rhythms. We then test whether perturbing the availability of the best candidate nutrient in vitro elicits changes their schedule for asexual development. Results: Our large-scale metabolomics experiment and follow up experiments reveal that only one metabolite - the amino acid isoleucine - fits criteria for a time-of-day cue used by parasites to set the schedule for replication. The response to isoleucine is a parasite strategy rather than solely the consequences of a constraint imposed by host rhythms, because unlike when parasites are deprived of other essential nutrients, they suffer no apparent costs from isoleucine withdrawal. Conclusions: Overall, our data suggest parasites can use the daily rhythmicity of blood-isoleucine concentration to synchronise asexual development with the availability of isoleucine, and potentially other resources, that arrive in the blood in a periodic manner due to the host's daily feeding-fasting cycle. Identifying both how and why parasites keep time opens avenues for interventions; interfering with the parasite's time-keeping mechanism may stall replication, increasing the efficacy of drugs and immune responses, and could also prevent parasites from entering dormancy to tolerate drugs.
Collapse
Affiliation(s)
- Kimberley F. Prior
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK,
| | - Benita Middleton
- School of Biosciences and Medicine, University of Surrey, Surrey, UK
| | - Alíz T.Y. Owolabi
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| | - Mary L. Westwood
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| | - Jacob Holland
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| | - Aidan J. O'Donnell
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, Francis Crick Institute, London, UK,Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Debra J. Skene
- School of Biosciences and Medicine, University of Surrey, Surrey, UK
| | - Sarah E. Reece
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK,Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
40
|
Mofatteh M, Echegaray-Iturra F, Alamban A, Dalla Ricca F, Bakshi A, Aydogan MG. Autonomous clocks that regulate organelle biogenesis, cytoskeletal organization, and intracellular dynamics. eLife 2021; 10:e72104. [PMID: 34586070 PMCID: PMC8480978 DOI: 10.7554/elife.72104] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/14/2021] [Indexed: 12/27/2022] Open
Abstract
How do cells perceive time? Do cells use temporal information to regulate the production/degradation of their enzymes, membranes, and organelles? Does controlling biological time influence cytoskeletal organization and cellular architecture in ways that confer evolutionary and physiological advantages? Potential answers to these fundamental questions of cell biology have historically revolved around the discussion of 'master' temporal programs, such as the principal cyclin-dependent kinase/cyclin cell division oscillator and the circadian clock. In this review, we provide an overview of the recent evidence supporting an emerging concept of 'autonomous clocks,' which under normal conditions can be entrained by the cell cycle and/or the circadian clock to run at their pace, but can also run independently to serve their functions if/when these major temporal programs are halted/abrupted. We begin the discussion by introducing recent developments in the study of such clocks and their roles at different scales and complexities. We then use current advances to elucidate the logic and molecular architecture of temporal networks that comprise autonomous clocks, providing important clues as to how these clocks may have evolved to run independently and, sometimes at the cost of redundancy, have strongly coupled to run under the full command of the cell cycle and/or the circadian clock. Next, we review a list of important recent findings that have shed new light onto potential hallmarks of autonomous clocks, suggestive of prospective theoretical and experimental approaches to further accelerate their discovery. Finally, we discuss their roles in health and disease, as well as possible therapeutic opportunities that targeting the autonomous clocks may offer.
Collapse
Affiliation(s)
- Mohammad Mofatteh
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Fabio Echegaray-Iturra
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Andrew Alamban
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Francesco Dalla Ricca
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Anand Bakshi
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Mustafa G Aydogan
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
41
|
Zanghi G, Vaughan AM. Plasmodium vivax pre-erythrocytic stages and the latent hypnozoite. Parasitol Int 2021; 85:102447. [PMID: 34474178 DOI: 10.1016/j.parint.2021.102447] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/02/2023]
Abstract
Plasmodium vivax is the most geographically widespread malaria parasite on the planet. This is largely because after mosquito transmission, P. vivax sporozoites can invade hepatocytes and form latent liver stages known as hypnozoites. These persistent liver stages can activate weeks, months or even years after an infected individual suffers a primary clinical infection. Activation then leads to replication and liver stage schizont maturation that ultimately cause relapse of blood stage infection, disease, and onward transmission. Thus, the latent hypnozoite can lie in wait during times when onward transmission is unlikely due to conditions that do not favor the mosquito. For example, in temperate climates where mosquito prevalence is only seasonal. Furthermore, the elimination of hypnozoites is challenging since the hypnozoite reservoir is currently undetectable and not killed by most antimalarial drugs. Here, we review our current knowledge of the pre-erythrocytic stages of the malaria parasite - the sporozoite and liver stages, including the elusive and enigmatic hypnozoite. We focus on our understanding of sporozoite biology, the novel animal models that are available to study the hypnozoite and hypnozoite activation and the ongoing efforts to understand the biological makeup of the hypnozoite that allow for its persistence in the human host.
Collapse
Affiliation(s)
| | - Ashley M Vaughan
- Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
42
|
Dos Santos BM, Pereira PH, Garcia CR. Molecular basis of synchronous replication of malaria parasites in the blood stage. Curr Opin Microbiol 2021; 63:210-215. [PMID: 34428626 DOI: 10.1016/j.mib.2021.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
The search for host factors that leads to malaria parasite synchronization has been the focus of several laboratories. The host hormone melatonin synchronizes Plasmodium falciparum in culture by increasing the number of mature parasite stages through a PLC-IP3 activation. Melatonin signaling is linked to crosstalk between Ca2+-cAMP that results in PKA activation. Two other kinases, PfPK7 and PfeIK1, and the nuclear protein PfMORC that lacks melatonin sensitivity in the inducible knock-down parasites are also identified as part of the hormone-signal transduction pathways. Melatonin also modulates P. falciparum mitochondrial fission genes FIS1, DYN1, and DYN2 in a stage-specific manner. How these multiple molecular mechanisms are orchestrated to lead to parasite synchronization is a fascinating and opened biological question.
Collapse
Affiliation(s)
- Benedito M Dos Santos
- Laboratory of Functional Genomics and Antimalarial Discovery, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, 05508-000, São Paulo, Brazil
| | - Pedro Hs Pereira
- Laboratory of Functional Genomics and Antimalarial Discovery, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, 05508-000, São Paulo, Brazil
| | - Célia Rs Garcia
- Laboratory of Functional Genomics and Antimalarial Discovery, Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, 05508-000, São Paulo, Brazil.
| |
Collapse
|
43
|
Schäfer C, Zanghi G, Vaughan AM, Kappe SHI. Plasmodium vivax Latent Liver Stage Infection and Relapse: Biological Insights and New Experimental Tools. Annu Rev Microbiol 2021; 75:87-106. [PMID: 34196569 DOI: 10.1146/annurev-micro-032421-061155] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Plasmodium vivax is the most widespread human malaria parasite, in part because it can form latent liver stages known as hypnozoites after transmission by female anopheline mosquitoes to human hosts. These persistent stages can activate weeks, months, or even years after the primary clinical infection; replicate; and initiate relapses of blood stage infection, which causes disease and recurring transmission. Eliminating hypnozoites is a substantial obstacle for malaria treatment and eradication since the hypnozoite reservoir is undetectable and unaffected by most antimalarial drugs. Importantly, in some parts of the globe where P. vivax malaria is endemic, as many as 90% of P. vivax blood stage infections are thought to be relapses rather than primary infections, rendering the hypnozoite a major driver of P. vivax epidemiology. Here, we review the biology of the hypnozoite and recent discoveries concerning this enigmatic parasite stage. We discuss treatment and prevention challenges, novel animal models to study hypnozoites and relapse, and hypotheses related to hypnozoite formation and activation. Expected final online publication date for the Annual Review of Microbiology, Volume 75 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Carola Schäfer
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , ,
| | - Gigliola Zanghi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , ,
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , , .,Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , , .,Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA.,Deparment of Global Health, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
44
|
Simon CS, Stürmer VS, Guizetti J. How Many Is Enough? - Challenges of Multinucleated Cell Division in Malaria Parasites. Front Cell Infect Microbiol 2021; 11:658616. [PMID: 34026661 PMCID: PMC8137892 DOI: 10.3389/fcimb.2021.658616] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Regulating the number of progeny generated by replicative cell cycles is critical for any organism to best adapt to its environment. Classically, the decision whether to divide further is made after cell division is completed by cytokinesis and can be triggered by intrinsic or extrinsic factors. Contrarily, cell cycles of some species, such as the malaria-causing parasites, go through multinucleated cell stages. Hence, their number of progeny is determined prior to the completion of cell division. This should fundamentally affect how the process is regulated and raises questions about advantages and challenges of multinucleation in eukaryotes. Throughout their life cycle Plasmodium spp. parasites undergo four phases of extensive proliferation, which differ over three orders of magnitude in the amount of daughter cells that are produced by a single progenitor. Even during the asexual blood stage proliferation parasites can produce very variable numbers of progeny within one replicative cycle. Here, we review the few factors that have been shown to affect those numbers. We further provide a comparative quantification of merozoite numbers in several P. knowlesi and P. falciparum parasite strains, and we discuss the general processes that may regulate progeny number in the context of host-parasite interactions. Finally, we provide a perspective of the critical knowledge gaps hindering our understanding of the molecular mechanisms underlying this exciting and atypical mode of parasite multiplication.
Collapse
Affiliation(s)
| | | | - Julien Guizetti
- Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
45
|
O'Donnell AJ, Reece SE. Ecology of asynchronous asexual replication: the intraerythrocytic development cycle of Plasmodium berghei is resistant to host rhythms. Malar J 2021; 20:105. [PMID: 33608011 PMCID: PMC7893937 DOI: 10.1186/s12936-021-03643-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/10/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Daily periodicity in the diverse activities of parasites occurs across a broad taxonomic range. The rhythms exhibited by parasites are thought to be adaptations that allow parasites to cope with, or exploit, the consequences of host activities that follow daily rhythms. Malaria parasites (Plasmodium) are well-known for their synchronized cycles of replication within host red blood cells. Whilst most species of Plasmodium appear sensitive to the timing of the daily rhythms of hosts, and even vectors, some species present no detectable rhythms in blood-stage replication. Why the intraerythrocytic development cycle (IDC) of, for example Plasmodium chabaudi, is governed by host rhythms, yet seems completely independent of host rhythms in Plasmodium berghei, another rodent malaria species, is mysterious. METHODS This study reports a series of five experiments probing the relationships between the asynchronous IDC schedule of P. berghei and the rhythms of hosts and vectors by manipulating host time-of-day, photoperiod and feeding rhythms. RESULTS The results reveal that: (i) a lack coordination between host and parasite rhythms does not impose appreciable fitness costs on P. berghei; (ii) the IDC schedule of P. berghei is impervious to host rhythms, including altered photoperiod and host-feeding-related rhythms; (iii) there is weak evidence for daily rhythms in the density and activities of transmission stages; but (iv), these rhythms have little consequence for successful transmission to mosquitoes. CONCLUSIONS Overall, host rhythms do not affect the performance of P. berghei and its asynchronous IDC is resistant to the scheduling forces that underpin synchronous replication in closely related parasites. This suggests that natural variation in the IDC schedule across species represents different parasite strategies that maximize fitness. Thus, subtle differences in the ecological interactions between parasites and their hosts/vectors may select for the evolution of very different IDC schedules.
Collapse
Affiliation(s)
- Aidan J O'Donnell
- Institute of Evolutionary Biology, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Rd, Edinburgh, EH9 3FL, UK.
| | - Sarah E Reece
- Institute of Evolutionary Biology, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Rd, Edinburgh, EH9 3FL, UK
| |
Collapse
|
46
|
A nuclear protein, PfMORC confers melatonin dependent synchrony of the human malaria parasite P. falciparum in the asexual stage. Sci Rep 2021; 11:2057. [PMID: 33479315 PMCID: PMC7820235 DOI: 10.1038/s41598-021-81235-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/24/2020] [Indexed: 01/05/2023] Open
Abstract
The host hormone melatonin is known to modulate the asexual cell-cycle of the human malaria parasite Plasmodium falciparum and the kinase PfPK7 is fundamental in the downstream signaling pathways. The nuclear protein PfMORC displays a histidine kinase domain and is involved in parasite cell cycle control. By using a real-time assay, we show a 24 h (h) rhythmic expression of PfMORC at the parasite asexual cycle and the expression is dramatically changed when parasites were treated with 100 nM melatonin for 17 h. Moreover, PfMORC expression was severely affected in PfPK7 knockout (PfPK7−) parasites following melatonin treatment. Parasites expressing 3D7morc-GFP shows nuclear localization of the protein during the asexual stage of parasite development. Although the PfMORC knockdown had no significant impact on the parasite proliferation in vitro it significantly changed the ratio of the different asexual intraerythrocytic stages of the parasites upon the addition of melatonin. Our data reveal that in addition to the upstream melatonin signaling pathways such as IP3 generation, calcium, and cAMP rise, a nuclear protein, PfMORC is essential for the hormone response in parasite synchronization.
Collapse
|
47
|
Adams KL, Sun EF, Alaidrous W, de Roode JC. Constant Light and Frequent Schedule Changes Do Not Impact Resistance to Parasites in Monarch Butterflies. J Biol Rhythms 2021; 36:286-296. [PMID: 33445989 DOI: 10.1177/0748730420985312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Organisms have evolved internal biological clocks to regulate their activities based on external environmental cues, such as light, temperature, and food. Environmental disruption of these rhythms, such as caused by constant light or frequent light schedule changes, has been shown to impair development, reduce survival, and increase infection susceptibility and disease progression in numerous organisms. However, the precise role of the biological clock in host-parasite interactions is understudied and has focused on unnatural host-parasite combinations in lab-adapted inbred models. Here, we use the natural interaction between monarch butterflies (Danaus plexippus) and their virulent protozoan parasite, Ophryocystis elektroscirrha, to investigate the effects of constant light and frequent light schedule changes on development, survival, and parasite susceptibility. We show that constant light exposure slows the monarchs' rate of development but does not increase susceptibility to parasitic infection. Furthermore, frequent schedule changes decrease parasite growth, but have no effect on egg-to-adult survival of infected monarchs. Interestingly, these conditions are usually disruptive to the biological clock, but do not significantly impact the clock of monarch larvae. These unexpected findings show that constant light and frequent schedule changes can uncouple host and parasite performance and highlight how natural relationships are needed to expand our understanding of clocks in host-parasite interactions.
Collapse
Affiliation(s)
- Kandis L Adams
- Department of Biology, Emory University, Atlanta, GA, USA
| | | | - Wajd Alaidrous
- Department of Biology, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
48
|
Pereira-Silva JW, Martins-Campos KM, Sabrina Dos Reis Martins E, de Souza Menezes A, Guimarães Lacerda MV, Costa Pessoa FA, Ríos-Velásquez CM. Long-lasting infectivity of Plasmodium vivax present in malarial patient blood to Anopheles aquasalis. Exp Parasitol 2021; 222:108064. [PMID: 33421382 DOI: 10.1016/j.exppara.2021.108064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 11/18/2020] [Accepted: 01/04/2021] [Indexed: 11/15/2022]
Abstract
Experimental studies for understanding the relationship between Plasmodium vivax and its vector hosts are difficult, because of to the lack of a long-term, in vitro continuous culture system unavailability of infected blood samples, seasonality of the disease, and the concentration of most cases in remote areas. This study evaluates the duration of the infectivity of P. vivax to Anopheles aquasalis after collecting blood from malaria-infected patients. Blood was collected from patients and stored at 4 °C and 37 °C. Every day, for 4 days, the blood was fed to An. aquasalis adult females, and a Giemsa-stained thick blood smear was mounted to account for sexual (gametocytes) and asexual (trophozoites and schizonts) stages and calculate parasitemia. Oocysts in the midgut of the mosquitoes were counted on the seventh day after feeding. Kruskal-Wallis test was used to compare the mean number of oocysts (MO) and the parasite density (PD) in each storage condition and post-infection time-points. The Mann-Whitney test was used to compare the number of oocysts for each day between temperatures. The results show that P. vivax stored at 4 °C and at 37 °C has its infectivity to An. aquasalis preserved for 2 days and 3 days, respectively. Infection rate (IR), PD and MO were higher on the day of blood collection and decreased gradually over time. The parasite density (number of parasites/μL) diminished faster at 4 °C than at 37 °C. In this study, a preservation protocol is shown for long-lasting infectivity of P. vivax in a blood sample taken from malaria-infected patients. These results show that infectivity of P. vivax stored at 4 °C and at 37 °C to An. aquasalis persist until 3 days after blood collection, but parasite density, infection rate, and mean of oocysts decreased 24h after blood collection. Since the malaria cases are increasingly far from the urban areas these results indicate that is possible, losing some infectivity, to realize experimental infections several dozen hours after the blood collection. However, it is necessary to improve the procedures for preserving P. vivax gametocytes for mosquito infection in the laboratory.
Collapse
Affiliation(s)
- Jordam William Pereira-Silva
- Lab. Ecologia de Doenças Transmissíveis Na Amazônia, Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Brazil; Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil; PPG Medicina Tropical, Escola Superior de Ciências da Saúde, Universidade Do Estado Do Amazonas, Manaus, Brazil
| | | | | | - Alexandre de Souza Menezes
- Lab. Ecologia de Doenças Transmissíveis Na Amazônia, Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Brazil; PPG Biologia da Interação Patógeno-Hospedeiro, Instituto Leônidas e Maria Deane, Fiocruz, Manaus, AM, Brazil
| | - Marcus Vinicius Guimarães Lacerda
- Lab. Diagnóstico e Controle de Doenças Infecciosas Na Amazônia, Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Brazil; Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil; PPG Medicina Tropical, Escola Superior de Ciências da Saúde, Universidade Do Estado Do Amazonas, Manaus, Brazil; PPG Biologia da Interação Patógeno-Hospedeiro, Instituto Leônidas e Maria Deane, Fiocruz, Manaus, AM, Brazil
| | - Felipe Arley Costa Pessoa
- Lab. Ecologia de Doenças Transmissíveis Na Amazônia, Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Brazil; PPG Biologia da Interação Patógeno-Hospedeiro, Instituto Leônidas e Maria Deane, Fiocruz, Manaus, AM, Brazil
| | - Claudia Maria Ríos-Velásquez
- Lab. Ecologia de Doenças Transmissíveis Na Amazônia, Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Brazil; PPG Medicina Tropical, Escola Superior de Ciências da Saúde, Universidade Do Estado Do Amazonas, Manaus, Brazil; PPG Biologia da Interação Patógeno-Hospedeiro, Instituto Leônidas e Maria Deane, Fiocruz, Manaus, AM, Brazil.
| |
Collapse
|
49
|
Pereira PHS, Garcia CRS. Melatonin action in Plasmodium infection: Searching for molecules that modulate the asexual cycle as a strategy to impair the parasite cycle. J Pineal Res 2021; 70:e12700. [PMID: 33025644 PMCID: PMC7757246 DOI: 10.1111/jpi.12700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/26/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Half of the world's population lives in countries at risk of malaria infection, which results in approximately 450,000 deaths annually. Malaria parasites infect erythrocytes in a coordinated manner, with cycle durations in multiples of 24 hours, which reflects a behavior consistent with the host's circadian cycle. Interference in cycle coordination can help the immune system to naturally fight infection. Consequently, there is a search for new drugs that interfere with the cycle duration for combined treatment with conventional antimalarials. Melatonin appears to be a key host hormone responsible for regulating circadian behavior in the parasite cycle. In addition to host factors, there are still unknown factors intrinsic to the parasite that control the cycle duration. In this review, we present a series of reports of indole compounds and melatonin derivatives with antimalarial activity that were tested on several species of Plasmodium to evaluate the cytotoxicity to parasites and human cells, in addition to the ability to interfere with the development of the erythrocytic cycle. Most of the reported compounds had an IC50 value in the low micromolar range, without any toxicity to human cells. Triptosil, an indole derivative of melatonin, was able to inhibit the effect of melatonin in vitro without causing changes to the parasitemia. The wide variety of tested compounds indicates that it is possible to develop a compound capable of safely eliminating parasites from the host and interfering with the life cycle, which is promising for the development of new combined therapies against malaria.
Collapse
Affiliation(s)
- Pedro H. S. Pereira
- Department of Clinical and Toxicological AnalysesSchool of Pharmaceutical SciencesUniversity of São PauloSão PauloBrazil
| | - Celia R. S. Garcia
- Department of Clinical and Toxicological AnalysesSchool of Pharmaceutical SciencesUniversity of São PauloSão PauloBrazil
| |
Collapse
|
50
|
Intrinsic multiplication rate variation and plasticity of human blood stage malaria parasites. Commun Biol 2020; 3:624. [PMID: 33116247 PMCID: PMC7595149 DOI: 10.1038/s42003-020-01349-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022] Open
Abstract
Pathogen multiplication rate is theoretically an important determinant of virulence, although often poorly understood and difficult to measure accurately. We show intrinsic asexual blood stage multiplication rate variation of the major human malaria parasite Plasmodium falciparum to be associated with blood-stage infection intensity in patients. A panel of clinical isolates from a highly endemic West African population was analysed repeatedly during five months of continuous laboratory culture, showing a range of exponential multiplication rates at all timepoints tested, mean rates increasing over time. All isolates had different genome sequences, many containing within-isolate diversity that decreased over time in culture, but increases in multiplication rates were not primarily attributable to genomic selection. New mutants, including premature stop codons emerging in a few isolates, did not attain sufficiently high frequencies to substantially affect overall multiplication rates. Significantly, multiplication rate variation among the isolates at each of the assayed culture timepoints robustly correlated with parasite levels seen in patients at clinical presentation, indicating innate parasite control of multiplication rate that contributes to virulence. Lindsay Stewart et al. analyze clinical isolates of the human malaria parasite Plasmodium falciparum from a highly endemic West African population and show that intrinsic multiplication rate variation is associated with blood-stage infection intensity. Their results indicate that parasite control of multiplication contributes to virulence.
Collapse
|