1
|
Nagata K, Ohashi K, Hashimoto C, Sayed AEDH, Yasuda T, Dutta B, Kajihara T, Mitani H, Suzuki M, Funayama T, Oda S, Watanabe-Asaka T. Responses of hematopoietic cells after ionizing-irradiation in anemic adult medaka ( Oryzias latipes). Int J Radiat Biol 2022; 99:663-672. [PMID: 35939385 DOI: 10.1080/09553002.2022.2110328] [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: 05/30/2022] [Revised: 07/20/2022] [Accepted: 07/28/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE Hematopoietic tissues of vertebrates are highly radiation sensitive and the effects of ionizing radiation on the hematopoiesis have been studied in mammals and teleosts for decades. In this study, radiation responses in the kidney, the main hematopoietic organ in teleosts, were investigated in Japanese medaka (Oryzias latipes), which has been a model animal and a large body of knowledge has been accumulated in radiation biology. METHODS Kidney, the main hematopoietic tissue of adult medaka fish, was locally irradiated using proton and carbon ion beams irradiation system of Takasaki Ion Accelerator for Advanced Radiation Application (TIARA), QST, and the effects on peripheral blood cells and histology of the kidney were investigated. RESULTS When only kidneys were locally irradiated with proton or carbon ion beam (15 Gy), the hematopoietic cells in the irradiated kidney and cell density in the peripheral blood decreased 7 days after the irradiation in the same manner as after the whole-body irradiation with γ-rays (15 Gy). These results demonstrate that direct irradiation of the hematopoietic cells in the kidney induced cell death and/or cell cycle arrest and stopped the supply of erythroid cells. Then, the cell density in the peripheral blood recovered to the control level within 4 days and 7 days after the γ-ray and proton beam irradiation (15 Gy), respectively, while the cell density in the peripheral blood did not recover after the carbon ion beam irradiation (15 Gy). The hematopoietic cells in the irradiated kidneys temporarily decreased and recovered to the control level within 21 days after the γ-ray or proton beam irradiation (15 Gy), while it did not recover after the carbon ion beam irradiation (15 Gy). In contrast, the recovery of the cell density in the peripheral blood delayed when anemic medaka were irradiated 1 day after the administration of phenylhydrazine. With and without γ-ray irradiation, a large number of hematopoietic cells was still proliferating in the kidney 7 days after the anemia induction. CONCLUSIONS The results obtained strongly suggest that the hematopoietic stem cells in medaka kidney prioritize to proliferate and increase peripheral blood cells to eliminate anemia, even when they are damaged by high-dose irradiation.
Collapse
Affiliation(s)
- Kento Nagata
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
- Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), National Institute of Radiological Sciences, Chiba, Japan
| | - Keita Ohashi
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Chika Hashimoto
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Alaa El-Din Hamid Sayed
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
- Zoology department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Takako Yasuda
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
- Department of Chemical and Biological Sciences, Japan Women's University, Tokyo, Japan
| | - Bibek Dutta
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Takayuki Kajihara
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Hiroshi Mitani
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Michiyo Suzuki
- Takasaki Advanced Radiation Research Institute, Quantum Beam Science Research Directorate, QST, Takasaki, Japan
| | - Tomoo Funayama
- Takasaki Advanced Radiation Research Institute, Quantum Beam Science Research Directorate, QST, Takasaki, Japan
| | - Shoji Oda
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Tomomi Watanabe-Asaka
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
- Division of Physiology, Faculty of Medicine, Tohoku Medical Pharmaceutical University, Sendai, Japan
| |
Collapse
|
2
|
Kamiya T, Davis NM, Greischar MA, Schneider D, Mideo N. Linking functional and molecular mechanisms of host resilience to malaria infection. eLife 2021; 10:e65846. [PMID: 34636723 PMCID: PMC8510579 DOI: 10.7554/elife.65846] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 08/16/2021] [Indexed: 12/30/2022] Open
Abstract
It remains challenging to understand why some hosts suffer severe illnesses, while others are unscathed by the same infection. We fitted a mathematical model to longitudinal measurements of parasite and red blood cell density in murine hosts from diverse genetic backgrounds to identify aspects of within-host interactions that explain variation in host resilience and survival during acute malaria infection. Among eight mouse strains that collectively span 90% of the common genetic diversity of laboratory mice, we found that high host mortality was associated with either weak parasite clearance, or a strong, yet imprecise response that inadvertently removes uninfected cells in excess. Subsequent cross-sectional cytokine assays revealed that the two distinct functional mechanisms of poor survival were underpinned by low expression of either pro- or anti-inflammatory cytokines, respectively. By combining mathematical modelling and molecular immunology assays, our study uncovered proximate mechanisms of diverse infection outcomes across multiple host strains and biological scales.
Collapse
Affiliation(s)
- Tsukushi Kamiya
- Department of Ecology and Evolutionary Biology, University of TorontoTorontoCanada
| | - Nicole M Davis
- Department of Microbiology and Immunology, Stanford UniversityStanfordUnited States
| | - Megan A Greischar
- Department of Ecology and Evolutionary Biology, Cornell UniversityIthacaUnited States
| | - David Schneider
- Department of Microbiology and Immunology, Stanford UniversityStanfordUnited States
| | - Nicole Mideo
- Department of Ecology and Evolutionary Biology, University of TorontoTorontoCanada
| |
Collapse
|
3
|
SONG TIANQI, WANG CHUNCHENG, TIAN BOPING. MULTIPLE PERIODIC SOLUTIONS OF A WITHIN-HOST MALARIA INFECTION MODEL WITH TIME DELAY. J BIOL SYST 2021. [DOI: 10.1142/s0218339021500108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this paper, we study a within-host malaria infection model recently proposed by Schneider et al. in 2018. The stability and Hopf bifurcation analysis at the interior equilibrium are carried out, finding that the basic reproduction number plays a key role in the dynamics of the model, and incrementing the time delay will induce Hopf bifurcation at this equilibrium. The global extension of the local Hopf branch is further tracked numerically by the MatLab package DDE-BIFTOOL. Neimark-Sacker bifurcation of Poincaré map and period-doubling bifurcation of the bifurcated periodic solution are also detected, resulting in the existence of quasi-periodic and multiple periodic solutions, respectively. These results reveal that Hopf bifurcation will indeed bring about the rich dynamics of the model.
Collapse
Affiliation(s)
- TIANQI SONG
- School of Economics and Management, Shanghai Maritime University, Shanghai, P. R. China
- School of Mathematics, Harbin Institute of Technology, Harbin, Heilongjiang, P. R. China
| | - CHUNCHENG WANG
- School of Mathematics, Harbin Institute of Technology, Harbin, Heilongjiang, P. R. China
| | - BOPING TIAN
- School of Mathematics, Harbin Institute of Technology, Harbin, Heilongjiang, P. R. China
| |
Collapse
|
4
|
Wait LF, Kamiya T, Fairlie-Clarke KJ, Metcalf CJE, Graham AL, Mideo N. Differential drivers of intraspecific and interspecific competition during malaria-helminth co-infection. Parasitology 2021; 148:1030-1039. [PMID: 33971991 PMCID: PMC11010048 DOI: 10.1017/s003118202100072x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 05/04/2021] [Indexed: 11/05/2022]
Abstract
Various host and parasite factors interact to determine the outcome of infection. We investigated the effects of two factors on the within-host dynamics of malaria in mice: initial infectious dose and co-infection with a helminth that limits the availability of red blood cells (RBCs). Using a statistical, time-series approach to model the within-host ‘epidemiology’ of malaria, we found that increasing initial dose reduced the time to peak cell-to-cell parasite propagation, but also reduced its magnitude, while helminth co-infection delayed peak cell-to-cell propagation, except at the highest malaria doses. Using a mechanistic model of within-host infection dynamics, we identified dose-dependence in parameters describing host responses to malaria infection and uncovered a plausible explanation of the observed differences in single vs co-infections. Specifically, in co-infections, our model predicted a higher background death rate of RBCs. However, at the highest dose, when intraspecific competition between malaria parasites would be highest, these effects of co-infection were not observed. Such interactions between initial dose and co-infection, although difficult to predict a priori, are key to understanding variation in the severity of disease experienced by hosts and could inform studies of malaria transmission dynamics in nature, where co-infection and low doses are the norm.
Collapse
Affiliation(s)
- L. F. Wait
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - T. Kamiya
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | | | - C. J. E. Metcalf
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - A. L. Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| | - N. Mideo
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
5
|
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
|
6
|
Illius AW, Lievaart-Peterson K, McNeilly TN, Savill NJ. Epidemiology and control of maedi-visna virus: Curing the flock. PLoS One 2020; 15:e0238781. [PMID: 32911525 PMCID: PMC7482963 DOI: 10.1371/journal.pone.0238781] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 08/17/2020] [Indexed: 11/25/2022] Open
Abstract
Maedi-visna (MV) is a complex lentiviral disease syndrome characterised by long immunological and clinical latencies and chronic progressive inflammatory pathology. Incurable at the individual level, it is widespread in most sheep-keeping countries, and is a cause of lost production and poor animal welfare. Culling seropositive animals is the main means of control, but it might be possible to manage virus transmission effectively if its epidemiology was better quantified. We derive a mathematical epidemiological model of the temporal distributions of seroconversion probabilities and estimate susceptibility, transmission rate and latencies in three serological datasets. We demonstrate the existence of epidemiological latency, which has not explicitly been recognised in the SRLV literaure. This time delay between infection and infectiousness apparently exceeds the delay between infection and seroconversion. Poor body condition was associated with more rapid seroconversion, but not with a higher probability of infection. We estimate transmission rates amongst housed sheep to be at about 1,000 times faster than when sheep were at grass, when transmission was negligible. Maternal transmission has only a small role in transmission, because lambs from infected ewes have a low probability of being infected directly by them, and only a small proportion of lambs need be retained to maintain flock size. Our results show that MV is overwhelmingly a disease of housing, where sheep are kept in close proximity. Prevalence of MV is likely to double each year from an initial low incidence in housed flocks penned in typically-sized groups of sheep (c. 50) for even a few days per year. Ewes kept entirely at grass are unlikely to experience transmission frequently enough for MV to persist, and pre-existing infection should die out as older ewes are replaced, thereby essentially curing the flock.
Collapse
Affiliation(s)
- Andrew W Illius
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland
| | | | - Tom N McNeilly
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian, Scotland
| | - Nicholas J Savill
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland
| |
Collapse
|
7
|
Foy BH, Gonçalves BP, Higgins JM. Unraveling Disease Pathophysiology with Mathematical Modeling. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2020; 15:371-394. [PMID: 31977295 DOI: 10.1146/annurev-pathmechdis-012419-032557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Modeling has enabled fundamental advances in our understanding of the mechanisms of health and disease for centuries, since at least the time of William Harvey almost 500 years ago. Recent technological advances in molecular methods, computation, and imaging generate optimism that mathematical modeling will enable the biomedical research community to accelerate its efforts in unraveling the molecular, cellular, tissue-, and organ-level processes that maintain health, predispose to disease, and determine response to treatment. In this review, we discuss some of the roles of mathematical modeling in the study of human physiology and pathophysiology and some challenges and opportunities in general and in two specific areas: in vivo modeling of pulmonary function and in vitro modeling of blood cell populations.
Collapse
Affiliation(s)
- Brody H Foy
- Center for Systems Biology and Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; .,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Bronner P Gonçalves
- Center for Systems Biology and Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; .,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - John M Higgins
- Center for Systems Biology and Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; .,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
8
|
Birget PLG, Schneider P, O’Donnell AJ, Reece SE. Adaptive phenotypic plasticity in malaria parasites is not constrained by previous responses to environmental change. EVOLUTION MEDICINE AND PUBLIC HEALTH 2019; 2019:190-198. [PMID: 31660151 PMCID: PMC6805783 DOI: 10.1093/emph/eoz028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/25/2019] [Indexed: 01/12/2023]
Abstract
Background and objectives Phenotypic plasticity enables organisms to maximize fitness by matching trait values to different environments. Such adaptive phenotypic plasticity is exhibited by parasites, which experience frequent environmental changes during their life cycle, between individual hosts and also in within-host conditions experienced during infections. Life history theory predicts that the evolution of adaptive phenotypic plasticity is limited by costs and constraints, but tests of these concepts are scarce. Methodology Here, we induce phenotypic plasticity in malaria parasites to test whether mounting a plastic response to an environmental perturbation constrains subsequent plastic responses to further environmental change. Specifically, we perturb red blood cell resource availability to induce Plasmodium chabaudi to alter the trait values of several phenotypes underpinning within-host replication and between-host transmission. We then transfer parasites to unperturbed hosts to examine whether constraints govern the parasites’ ability to alter these phenotypes in response to their new in-host environment. Results Parasites alter trait values in response to the within-host environment they are exposed to. We do not detect negative consequences, for within-host replication or between-host transmission, of previously mounting a plastic response to a perturbed within-host environment. Conclusions and implications We suggest that malaria parasites are highly plastic and adapted to adjusting their phenotypes in response to the frequent changes in the within-host conditions they experience during infections. Our findings support the growing body of evidence that medical interventions, such as anti-parasite drugs, induce plastic responses that are adaptive and can facilitate the survival and potentially, drug resistance of parasites. Lay Summary Malaria parasites have evolved flexible strategies to cope with the changing conditions they experience during infections. We show that using such flexible strategies does not impact upon the parasites’ ability to grow (resulting in disease symptoms) or transmit (spreading the disease).
Collapse
Affiliation(s)
- Philip L G Birget
- Institute of Evolutionary Biology and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh EH9 3FL, UK
| | - Petra Schneider
- Institute of Evolutionary Biology and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh EH9 3FL, UK
| | - Aidan J O’Donnell
- Institute of Evolutionary Biology and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, 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 Road, Edinburgh EH9 3FL, UK
| |
Collapse
|
9
|
Birget PLG, Prior KF, Savill NJ, Steer L, Reece SE. Plasticity and genetic variation in traits underpinning asexual replication of the rodent malaria parasite, Plasmodium chabaudi. Malar J 2019; 18:222. [PMID: 31262304 PMCID: PMC6604315 DOI: 10.1186/s12936-019-2857-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The ability of malaria (Plasmodium) parasites to adjust investment into sexual transmission stages versus asexually replicating stages is well known, but plasticity in other traits underpinning the replication rate of asexual stages in the blood has received less attention. Such traits include burst size (the number of merozoites produced per schizont), the duration of the asexual cycle, and invasion preference for different ages of red blood cell (RBC). METHODS Here, plasticity [environment (E) effects] and genetic variation [genotype (G) effects] in traits relating to asexual replication rate are examined for 4 genotypes of the rodent malaria parasite Plasmodium chabaudi. An experiment tested whether asexual dynamics differ between parasites infecting control versus anaemic hosts, and whether variation in replication rate can be explained by differences in burst size, asexual cycle, and invasion rates. RESULTS The within-host environment affected each trait to different extents but generally had similar impacts across genotypes. The dynamics of asexual densities exhibited a genotype by environment effect (G×E), in which one of the genotypes increased replication rate more than the others in anaemic hosts. Burst size and cycle duration varied between the genotypes (G), while burst size increased and cycle duration became longer in anaemic hosts (E). Variation in invasion rates of differently aged RBCs was not explained by environmental or genetic effects. Plasticity in burst size and genotype are the only traits making significant contributions to the increase in asexual densities observed in anaemic hosts, together explaining 46.4% of the variation in replication rate. CONCLUSIONS That host anaemia induces several species of malaria parasites to alter conversion rate is well documented. Here, previously unknown plasticity in other traits underpinning asexual replication is revealed. These findings contribute to mounting evidence that malaria parasites deploy a suite of sophisticated strategies to maximize fitness by coping with, or exploiting the opportunities provided by, the variable within-host conditions experienced during infections. That genetic variation and genotype by environment interactions also shape these traits highlights their evolutionary potential. Asexual replication rate is a major determinant of virulence and so, understanding the evolution of virulence requires knowledge of the ecological (within-host environment) and genetic drivers of variation among parasites.
Collapse
Affiliation(s)
- Philip L G Birget
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK.,Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Kimberley F Prior
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK. .,Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK.
| | - Nicholas J Savill
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK.,Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Lewis Steer
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK.,Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Sarah E Reece
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK.,Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| |
Collapse
|
10
|
Greischar MA, Reece SE, Savill NJ, Mideo N. The Challenge of Quantifying Synchrony in Malaria Parasites. Trends Parasitol 2019; 35:341-355. [PMID: 30952484 DOI: 10.1016/j.pt.2019.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/21/2022]
Abstract
Malaria infection is often accompanied by periodic fevers, triggered by synchronous cycles of parasite replication within the host. The degree of synchrony in parasite development influences the efficacy of drugs and immune defenses and is therefore relevant to host health and infectiousness. Synchrony is thought to vary over the course of infection and across different host-parasite genotype or species combinations, but the evolutionary significance - if any - of this diversity remains elusive. Standardized methods are lacking, but the most common metric for quantifying synchrony is the percentage of parasites in a particular developmental stage. We use a heuristic model to show that this metric is often unacceptably biased. Methodological challenges must be addressed to characterize diverse patterns of synchrony and their consequences for disease severity and spread.
Collapse
Affiliation(s)
- Megan A Greischar
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada.
| | - Sarah E Reece
- Institute of Evolutionary Biology and Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Nicholas J Savill
- Institute of Evolutionary Biology and Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Nicole Mideo
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Adaptive plasticity in the gametocyte conversion rate of malaria parasites. PLoS Pathog 2018; 14:e1007371. [PMID: 30427935 PMCID: PMC6261640 DOI: 10.1371/journal.ppat.1007371] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 11/28/2018] [Accepted: 10/02/2018] [Indexed: 11/30/2022] Open
Abstract
Sexually reproducing parasites, such as malaria parasites, experience a trade-off between the allocation of resources to asexual replication and the production of sexual forms. Allocation by malaria parasites to sexual forms (the conversion rate) is variable but the evolutionary drivers of this plasticity are poorly understood. We use evolutionary theory for life histories to combine a mathematical model and experiments to reveal that parasites adjust conversion rate according to the dynamics of asexual densities in the blood of the host. Our model predicts the direction of change in conversion rates that returns the greatest fitness after perturbation of asexual densities by different doses of antimalarial drugs. The loss of a high proportion of asexuals is predicted to elicit increased conversion (terminal investment), while smaller losses are managed by reducing conversion (reproductive restraint) to facilitate within-host survival and future transmission. This non-linear pattern of allocation is consistent with adaptive reproductive strategies observed in multicellular organisms. We then empirically estimate conversion rates of the rodent malaria parasite Plasmodium chabaudi in response to the killing of asexual stages by different doses of antimalarial drugs and forecast the short-term fitness consequences of these responses. Our data reveal the predicted non-linear pattern, and this is further supported by analyses of previous experiments that perturb asexual stage densities using drugs or within-host competition, across multiple parasite genotypes. Whilst conversion rates, across all datasets, are most strongly influenced by changes in asexual density, parasites also modulate conversion according to the availability of red blood cell resources. In summary, increasing conversion maximises short-term transmission and reducing conversion facilitates in-host survival and thus, future transmission. Understanding patterns of parasite allocation to reproduction matters because within-host replication is responsible for disease symptoms and between-host transmission determines disease spread. Malaria parasites in the host replicate asexually and, during each replication cycle, some asexuals transform into sexual stages that enable between-host transmission. It is not understood why the rate of conversion to sexual stages varies during infections despite its importance for the severity and spread of the disease. We combined a mathematical model and experiments to show that parasites adjust conversion rates depending on changes in their in-host population size. When population sizes plummet, between-host transmission is prioritised. However, smaller losses in number elicit reproductive restraint, which facilitates in-host survival and future transmission. We show that increased and decreased conversion in response to a range of in-host environments are actually part of one continuum: a sophisticated reproductive strategy similar to that of multicellular organisms.
Collapse
|
12
|
Mitochondrial DNA is critical for longevity and metabolism of transmission stage Trypanosoma brucei. PLoS Pathog 2018; 14:e1007195. [PMID: 30020996 PMCID: PMC6066258 DOI: 10.1371/journal.ppat.1007195] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/30/2018] [Accepted: 07/02/2018] [Indexed: 01/19/2023] Open
Abstract
The sleeping sickness parasite Trypanosoma brucei has a complex life cycle, alternating between a mammalian host and the tsetse fly vector. A tightly controlled developmental programme ensures parasite transmission between hosts as well as survival within them and involves strict regulation of mitochondrial activities. In the glucose-rich bloodstream, the replicative 'slender' stage is thought to produce ATP exclusively via glycolysis and uses the mitochondrial F1FO-ATP synthase as an ATP hydrolysis-driven proton pump to generate the mitochondrial membrane potential (ΔΨm). The 'procyclic' stage in the glucose-poor tsetse midgut depends on mitochondrial catabolism of amino acids for energy production, which involves oxidative phosphorylation with ATP production via the F1FO-ATP synthase. Both modes of the F1FO enzyme critically depend on FO subunit a, which is encoded in the parasite's mitochondrial DNA (kinetoplast or kDNA). Comparatively little is known about mitochondrial function and the role of kDNA in non-replicative 'stumpy' bloodstream forms, a developmental stage essential for disease transmission. Here we show that the L262P mutation in the nuclear-encoded F1 subunit γ that permits survival of 'slender' bloodstream forms lacking kDNA ('akinetoplastic' forms), via FO-independent generation of ΔΨm, also permits their differentiation into stumpy forms. However, these akinetoplastic stumpy cells lack a ΔΨm and have a reduced lifespan in vitro and in mice, which significantly alters the within-host dynamics of the parasite. We further show that generation of ΔΨm in stumpy parasites and their ability to use α-ketoglutarate to sustain viability depend on F1-ATPase activity. Surprisingly, however, loss of ΔΨm does not reduce stumpy life span. We conclude that the L262P γ subunit mutation does not enable FO-independent generation of ΔΨm in stumpy cells, most likely as a consequence of mitochondrial ATP production in these cells. In addition, kDNA-encoded genes other than FO subunit a are important for stumpy form viability.
Collapse
|
13
|
Birget PLG, Greischar MA, Reece SE, Mideo N. Altered life history strategies protect malaria parasites against drugs. Evol Appl 2018; 11:442-455. [PMID: 29636798 PMCID: PMC5891063 DOI: 10.1111/eva.12516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/30/2017] [Indexed: 11/26/2022] Open
Abstract
Drug resistance has been reported against all antimalarial drugs, and while parasites can evolve classical resistance mechanisms (e.g., efflux pumps), it is also possible that changes in life history traits could help parasites evade the effects of treatment. The life history of malaria parasites is governed by an intrinsic resource allocation problem: specialized stages are required for transmission, but producing these stages comes at the cost of producing fewer of the forms required for within-host survival. Drug treatment, by design, alters the probability of within-host survival, and so should alter the costs and benefits of investing in transmission. Here, we use a within-host model of malaria infection to predict optimal patterns of investment in transmission in the face of different drug treatment regimes and determine the extent to which alternative patterns of investment can buffer the fitness loss due to drugs. We show that over a range of drug doses, parasites are predicted to adopt "reproductive restraint" (investing more in asexual replication and less in transmission) to maximize fitness. By doing so, parasites recoup some of the fitness loss imposed by drugs, though as may be expected, increasing dose reduces the extent to which altered patterns of transmission investment can benefit parasites. We show that adaptation to drug-treated infections could result in more virulent infections in untreated hosts. This work emphasizes that in addition to classical resistance mechanisms, drug treatment generates selection for altered parasite life history. Understanding how any shifts in life history will alter the efficacy of drugs, as well as any limitations on such shifts, is important for evaluating and predicting the consequences of drug treatment.
Collapse
Affiliation(s)
- Philip L. G. Birget
- Institutes of Evolutionary Biology, Immunology and Infection ResearchUniversity of EdinburghEdinburghUK
| | - Megan A. Greischar
- Department of Ecology & Evolutionary BiologyUniversity of TorontoTorontoONCanada
| | - Sarah E. Reece
- Institutes of Evolutionary Biology, Immunology and Infection ResearchUniversity of EdinburghEdinburghUK
| | - Nicole Mideo
- Department of Ecology & Evolutionary BiologyUniversity of TorontoTorontoONCanada
| |
Collapse
|
14
|
Thakre N, Fernandes P, Mueller AK, Graw F. Examining the Reticulocyte Preference of Two Plasmodium berghei Strains during Blood-Stage Malaria Infection. Front Microbiol 2018. [PMID: 29515528 PMCID: PMC5826286 DOI: 10.3389/fmicb.2018.00166] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The blood-stage of the Plasmodium parasite is one of the key phases within its life cycle that influences disease progression during a malaria infection. The efficiency of the parasite in infecting red blood cells (RBC) determines parasite load and parasite-induced hemolysis that is responsible for the development of anemia and potentially drives severe disease progression. However, the molecular factors defining the infectivity of Plasmodium parasites have not been completely identified so far. Using the Plasmodium berghei mouse model for malaria, we characterized and compared the blood-stage infection dynamics of PbANKA WT and a mutant parasite strain lacking a novel Plasmodium antigen, PbmaLS_05, that is well conserved in both human and animal Plasmodium parasite strains. Infection of mice with parasites lacking PbmaLS_05 leads to lower parasitemia levels and less severe disease progression in contrast to mice infected with the wildtype PbANKA strain. To specifically determine the effect of deleting PbmaLS_05 on parasite infectivity we developed a mathematical model describing erythropoiesis and malarial infection of RBC. By applying our model to experimental data studying infection dynamics under normal and drug-induced altered erythropoietic conditions, we found that both PbANKA and PbmaLS_05 (-) parasite strains differed in their infectivity potential during the early intra-erythrocytic stage of infection. Parasites lacking PbmaLS_05 showed a decreased ability to infect RBC, and immature reticulocytes in particular that are usually a preferential target of the parasite. These altered infectivity characteristics limit parasite burden and affect disease progression. Our integrative analysis combining mathematical models and experimental data suggests that deletion of PbmaLS_05 affects productive infection of reticulocytes, which makes this antigen a useful target to analyze the actual processes relating RBC preferences to the development of severe disease outcomes in malaria.
Collapse
Affiliation(s)
- Neha Thakre
- Centre for Modeling and Simulation in the Biosciences, BioQuant-Center, Heidelberg University, Heidelberg, Germany
| | - Priyanka Fernandes
- Parasitology Unit, Centre for Infectious Diseases, University Hospital, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Parasitology Unit, Centre for Infectious Diseases, University Hospital, Heidelberg, Germany.,German Center for Infectious Diseases (DZIF), Heidelberg, Germany
| | - Frederik Graw
- Centre for Modeling and Simulation in the Biosciences, BioQuant-Center, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
15
|
Birget PLG, Repton C, O'Donnell AJ, Schneider P, Reece SE. Phenotypic plasticity in reproductive effort: malaria parasites respond to resource availability. Proc Biol Sci 2017; 284:20171229. [PMID: 28768894 PMCID: PMC5563815 DOI: 10.1098/rspb.2017.1229] [Citation(s) in RCA: 16] [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: 06/01/2017] [Accepted: 06/28/2017] [Indexed: 12/11/2022] Open
Abstract
The trade-off between survival and reproduction is fundamental in the life history of all sexually reproducing organisms. This includes malaria parasites, which rely on asexually replicating stages for within-host survival and on sexually reproducing stages (gametocytes) for between-host transmission. The proportion of asexual stages that form gametocytes (reproductive effort) varies during infections-i.e. is phenotypically plastic-in response to changes in a number of within-host factors, including anaemia. However, how the density and age structure of red blood cell (RBC) resources shape plasticity in reproductive effort and impacts upon parasite fitness is controversial. Here, we examine how and why the rodent malaria parasite Plasmodium chabaudi alters its reproductive effort in response to experimental perturbations of the density and age structure of RBCs. We show that all four of the genotypes studied increase reproductive effort when the proportion of RBCs that are immature is elevated during host anaemia, and that the responses of the genotypes differ. We propose that anaemia (counterintuitively) generates a resource-rich environment in which parasites can afford to allocate more energy to reproduction (i.e. transmission) and that anaemia also exposes genetic variation to selection. From an applied perspective, adaptive plasticity in parasite reproductive effort could explain the maintenance of genetic variation for virulence and why anaemia is often observed as a risk factor for transmission in human infections.
Collapse
Affiliation(s)
- Philip L G Birget
- Institutes of Evolutionary Biology, and Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Charlotte Repton
- Institutes of Evolutionary Biology, and Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Aidan J O'Donnell
- Institutes of Evolutionary Biology, and Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Petra Schneider
- Institutes of Evolutionary Biology, and Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Sarah E Reece
- Institutes of Evolutionary Biology, and Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3FL, UK
| |
Collapse
|
16
|
Watson J, Taylor WR, Menard D, Kheng S, White NJ. Modelling primaquine-induced haemolysis in G6PD deficiency. eLife 2017; 6. [PMID: 28155819 PMCID: PMC5330681 DOI: 10.7554/elife.23061] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 01/31/2017] [Indexed: 12/12/2022] Open
Abstract
Primaquine is the only drug available to prevent relapse in vivax malaria. The main adverse effect of primaquine is erythrocyte age and dose-dependent acute haemolytic anaemia in individuals with glucose-6-phosphate dehydrogenase deficiency (G6PDd). As testing for G6PDd is often unavailable, this limits the use of primaquine for radical cure. A compartmental model of the dynamics of red blood cell production and destruction was designed to characterise primaquine-induced haemolysis using a holistic Bayesian analysis of all published data and was used to predict a safer alternative to the currently recommended once weekly 0.75 mg/kg regimen for G6PDd. The model suggests that a step-wise increase in daily administered primaquine dose would be relatively safe in G6PDd. If this is confirmed, then were this regimen to be recommended for radical cure patients would not require testing for G6PDd in areas where G6PDd Viangchan or milder variants are prevalent. DOI:http://dx.doi.org/10.7554/eLife.23061.001 Malaria is the most important parasitic disease that affects humans. Over half of the malaria cases in Asia and South America are caused by a species of malaria parasite called Plasmodium vivax (known as vivax malaria). This form of malaria results in repeated illness because dormant parasites in the liver wake at intervals to infect the blood. The only available drug that can stop these relapses is a drug called primaquine, which was developed seventy years ago. Unfortunately, primaquine causes dangerous side effects in certain individuals who are deficient in an enzyme called G6PD, which helps defend red blood cells against stresses. Primaquine damages these cells so that they burst, leading to anaemia. This is a major problem because G6PD deficiency is common in regions where malaria is present: in some areas up to 30% of the population may be G6PD deficient. Since G6PD testing is not widely available, doctors often avoid prescribing primaquine to treat malaria, which results in more cases of disease relapse. Failing to prevent vivax relapses causes extensive illness and hinders efforts to eliminate malaria. Is there a way to give this drug to patients that would be safer for people with G6PD deficiency? Primaquine destroys older rather than younger red blood cells. Watson et al. used mathematical modelling to see whether it is possible to develop a primaquine treatment strategy that would allow a gradual destruction of older red blood cells in individuals with G6PD deficiency, which would be safer. The mathematical model incorporates data from previous studies in malaria patients and healthy volunteers with G6PD deficiency and combines this with knowledge of how red blood cells are produced and destroyed. Watson et al. predicted that giving primaquine over 20 days in a steadily increasing dose was safer than current recommendations. Mathematical models are simplifications of real world processes. The only way to test these findings properly will be to run a clinical trial that gives healthy volunteers who are G6PD deficient a course of primaquine treatment with a steadily increasing dose. DOI:http://dx.doi.org/10.7554/eLife.23061.002
Collapse
Affiliation(s)
- James Watson
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Walter Rj Taylor
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Didier Menard
- Unité d'Epidémiologie Moléculaire du Paludisme, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sim Kheng
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Nicholas J White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Graupner A, Eide DM, Instanes C, Andersen JM, Brede DA, Dertinger SD, Lind OC, Brandt-Kjelsen A, Bjerke H, Salbu B, Oughton D, Brunborg G, Olsen AK. Gamma radiation at a human relevant low dose rate is genotoxic in mice. Sci Rep 2016; 6:32977. [PMID: 27596356 PMCID: PMC5011728 DOI: 10.1038/srep32977] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/10/2016] [Indexed: 12/16/2022] Open
Abstract
Even today, 70 years after Hiroshima and accidents like in Chernobyl and Fukushima, we still have limited knowledge about the health effects of low dose rate (LDR) radiation. Despite their human relevance after occupational and accidental exposure, only few animal studies on the genotoxic effects of chronic LDR radiation have been performed. Selenium (Se) is involved in oxidative stress defence, protecting DNA and other biomolecules from reactive oxygen species (ROS). It is hypothesised that Se deficiency, as it occurs in several parts of the world, may aggravate harmful effects of ROS-inducing stressors such as ionising radiation. We performed a study in the newly established LDR-facility Figaro on the combined effects of Se deprivation and LDR γ exposure in DNA repair knockout mice (Ogg1−/−) and control animals (Ogg1+/−). Genotoxic effects were seen after continuous radiation (1.4 mGy/h) for 45 days. Chromosomal damage (micronucleus), phenotypic mutations (Pig-a gene mutation of RBCCD24−) and DNA lesions (single strand breaks/alkali labile sites) were significantly increased in blood cells of irradiated animals, covering three types of genotoxic activity. This study demonstrates that chronic LDR γ radiation is genotoxic in an exposure scenario realistic for humans, supporting the hypothesis that even LDR γ radiation may induce cancer.
Collapse
Affiliation(s)
- Anne Graupner
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway.,Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | - Dag M Eide
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway.,Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | - Christine Instanes
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway.,Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | - Jill M Andersen
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway.,Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | - Dag A Brede
- Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway.,Department of Environmental Sciences (IMV), Norwegian University of Life Sciences (NMBU), Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | | | - Ole C Lind
- Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway.,Department of Environmental Sciences (IMV), Norwegian University of Life Sciences (NMBU), Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | - Anicke Brandt-Kjelsen
- Department of Environmental Sciences (IMV), Norwegian University of Life Sciences (NMBU), Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | - Hans Bjerke
- Department of Monitoring and Research, Norwegian Radiation Protection Authority, Østerås 1332, Norway
| | - Brit Salbu
- Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway.,Department of Environmental Sciences (IMV), Norwegian University of Life Sciences (NMBU), Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | - Deborah Oughton
- Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway.,Department of Environmental Sciences (IMV), Norwegian University of Life Sciences (NMBU), Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | - Gunnar Brunborg
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway.,Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| | - Ann K Olsen
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway.,Centre for Environmental Radioactivity (CoE CERAD), Ås 1432, Norway
| |
Collapse
|
18
|
Ramiro RS, Pollitt LC, Mideo N, Reece SE. Facilitation through altered resource availability in a mixed-species rodent malaria infection. Ecol Lett 2016; 19:1041-50. [PMID: 27364562 PMCID: PMC5025717 DOI: 10.1111/ele.12639] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/03/2016] [Accepted: 05/13/2016] [Indexed: 12/17/2022]
Abstract
A major challenge in disease ecology is to understand how co-infecting parasite species interact. We manipulate in vivo resources and immunity to explain interactions between two rodent malaria parasites, Plasmodium chabaudi and P. yoelii. These species have analogous resource-use strategies to the human parasites Plasmodium falciparum and P. vivax: P. chabaudi and P. falciparum infect red blood cells (RBC) of all ages (RBC generalist); P. yoelii and P. vivax preferentially infect young RBCs (RBC specialist). We find that: (1) recent infection with the RBC generalist facilitates the RBC specialist (P. yoelii density is enhanced ~10 fold). This occurs because the RBC generalist increases availability of the RBC specialist's preferred resource; (2) co-infections with the RBC generalist and RBC specialist are highly virulent; (3) and the presence of an RBC generalist in a host population can increase the prevalence of an RBC specialist. Thus, we show that resources shape how parasite species interact and have epidemiological consequences.
Collapse
Affiliation(s)
- Ricardo S Ramiro
- Institutes of Evolutionary Biology, and Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JFL, UK
| | - Laura C Pollitt
- Institutes of Evolutionary Biology, and Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JFL, UK.,Centre for Immunity, Infection & Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, Edinburgh, EH9 3JFL, UK
| | - Nicole Mideo
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Canada
| | - Sarah E Reece
- Institutes of Evolutionary Biology, and Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JFL, UK.,Centre for Immunity, Infection & Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, Edinburgh, EH9 3JFL, UK
| |
Collapse
|
19
|
Greischar MA, Mideo N, Read AF, Bjørnstad ON. Predicting optimal transmission investment in malaria parasites. Evolution 2016; 70:1542-58. [DOI: 10.1111/evo.12969] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 05/07/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Megan A. Greischar
- Center For Infectious Disease Dynamics, Departments of Entomology and Biology, The Pennsylvania State University; University Park; Pennsylvania 16802
- Department of Ecology and Evolutionary Biology; University of Toronto; Toronto ON M5S 3B2 Canada
| | - Nicole Mideo
- Department of Ecology and Evolutionary Biology; University of Toronto; Toronto ON M5S 3B2 Canada
| | - Andrew F. Read
- Center For Infectious Disease Dynamics, Departments of Entomology and Biology, The Pennsylvania State University; University Park; Pennsylvania 16802
- Fogarty International Center; National Institutes of Health; Bethesda Maryland 20892
| | - Ottar N. Bjørnstad
- Center For Infectious Disease Dynamics, Departments of Entomology and Biology, The Pennsylvania State University; University Park; Pennsylvania 16802
- Fogarty International Center; National Institutes of Health; Bethesda Maryland 20892
| |
Collapse
|
20
|
Yan Y, Adam B, Galinski M, C Kissinger J, Moreno A, Gutierrez JB. Mathematical model of susceptibility, resistance, and resilience in the within-host dynamics between a Plasmodium parasite and the immune system. Math Biosci 2015; 270:213-23. [PMID: 26505135 DOI: 10.1016/j.mbs.2015.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 10/02/2015] [Accepted: 10/07/2015] [Indexed: 11/16/2022]
Abstract
We developed a coupled age-structured partial differential equation model to capture the disease dynamics during blood-stage malaria. The addition of age structure for the parasite population, with respect to previous models, allows us to better characterize the interaction between the malaria parasite and red blood cells during infection. Here we prove that the system we propose is well-posed and there exist at least two global states. We further demonstrate that the numerical simulation of the system coincides with clinically observed outcomes of primary and secondary malaria infection. The well-posedness of this system guarantees that the behavior of the model remains smooth, bounded, and continuously dependent on initial conditions; calibration with clinical data will constrain domains of parameters and variables to physiological ranges.
Collapse
Affiliation(s)
- Yi Yan
- Institute of Bioinformatics, University of Georgia, United States
| | - Brian Adam
- Department of Mathematics, University of Georgia, United States
| | - Mary Galinski
- Department of Medicine, Emory University, United States; Department of Microbiology and Immunology, Emory University, United States
| | - Jessica C Kissinger
- Institute of Bioinformatics, University of Georgia, United States; Department of Genetics, University of Georgia, United States
| | | | - Juan B Gutierrez
- Institute of Bioinformatics, University of Georgia, United States; Department of Mathematics, University of Georgia, United States.
| |
Collapse
|
21
|
Rommelaere S, Millet V, Rihet P, Atwell S, Helfer E, Chasson L, Beaumont C, Chimini G, Sambo MDR, Viallat A, Penha-Gonçalves C, Galland F, Naquet P. Serum pantetheinase/vanin levels regulate erythrocyte homeostasis and severity of malaria. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:3039-52. [PMID: 26343328 DOI: 10.1016/j.ajpath.2015.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 06/18/2015] [Accepted: 07/16/2015] [Indexed: 11/16/2022]
Abstract
Tissue pantetheinase, encoded by the VNN1 gene, regulates response to stress, and previous studies have shown that VNN genes contribute to the susceptibility to malaria. Herein, we evaluated the role of pantetheinase on erythrocyte homeostasis and on the development of malaria in patients and in a new mouse model of pantetheinase insufficiency. Patients with cerebral malaria have significantly reduced levels of serum pantetheinase activity (PA). In mouse, we show that a reduction in serum PA predisposes to severe malaria, including cerebral malaria and severe anemia. Therefore, scoring pantetheinase in serum may serve as a severity marker in malaria infection. This disease triggers an acute stress in erythrocytes, which enhances cytoadherence and hemolysis. We speculated that serum pantetheinase might contribute to erythrocyte resistance to stress under homeostatic conditions. We show that mutant mice with a reduced serum PA are anemic and prone to phenylhydrazine-induced anemia. A cytofluorometric and spectroscopic analysis documented an increased frequency of erythrocytes with an autofluorescent aging phenotype. This is associated with an enhanced oxidative stress and shear stress-induced hemolysis. Red blood cell transfer and bone marrow chimera experiments show that the aging phenotype is not cell intrinsic but conferred by the environment, leading to a shortening of red blood cell half-life. Therefore, serum pantetheinase level regulates erythrocyte life span and modulates the risk of developing complicated malaria.
Collapse
Affiliation(s)
- Samuel Rommelaere
- Immunology Center of Marseille-Luminy, Aix Marseille Université (UM2), the National Institute of Health and Medical Research INSERM U1104, the Centre National de la Recherche Scientifique CNRS UMR7280, Marseille, France
| | - Virginie Millet
- Immunology Center of Marseille-Luminy, Aix Marseille Université (UM2), the National Institute of Health and Medical Research INSERM U1104, the Centre National de la Recherche Scientifique CNRS UMR7280, Marseille, France
| | - Pascal Rihet
- Technological Advances for Genomics and Clinics (TAGC), Aix-Marseille Université, UMR_S 1090, INSERM U1090, Marseille, France
| | - Scott Atwell
- Marseilles Interdisciplinary Nanoscience Centre, Aix-Marseille Université, CNRS UMR7325, Marseille, France
| | - Emmanuèle Helfer
- Marseilles Interdisciplinary Nanoscience Centre, Aix-Marseille Université, CNRS UMR7325, Marseille, France
| | - Lionel Chasson
- Immunology Center of Marseille-Luminy, Aix Marseille Université (UM2), the National Institute of Health and Medical Research INSERM U1104, the Centre National de la Recherche Scientifique CNRS UMR7280, Marseille, France
| | - Carole Beaumont
- Biomedical Research Center Bichat-Beaujon, Université Paris Diderot, INSERM U773, Paris, France
| | - Giovanna Chimini
- Immunology Center of Marseille-Luminy, Aix Marseille Université (UM2), the National Institute of Health and Medical Research INSERM U1104, the Centre National de la Recherche Scientifique CNRS UMR7280, Marseille, France
| | | | - Annie Viallat
- Marseilles Interdisciplinary Nanoscience Centre, Aix-Marseille Université, CNRS UMR7325, Marseille, France
| | | | - Franck Galland
- Immunology Center of Marseille-Luminy, Aix Marseille Université (UM2), the National Institute of Health and Medical Research INSERM U1104, the Centre National de la Recherche Scientifique CNRS UMR7280, Marseille, France.
| | - Philippe Naquet
- Immunology Center of Marseille-Luminy, Aix Marseille Université (UM2), the National Institute of Health and Medical Research INSERM U1104, the Centre National de la Recherche Scientifique CNRS UMR7280, Marseille, France.
| |
Collapse
|
22
|
Shekh K, Khan S, Jena G, Kansara BR, Kushwaha S. 3-Aminobenzamide – a PARP inhibitor enhances the sensitivity of peripheral blood micronucleus and comet assays in mice. Toxicol Mech Methods 2014; 24:332-41. [DOI: 10.3109/15376516.2014.898355] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
23
|
Eymard N, Bessonov N, Gandrillon O, Koury MJ, Volpert V. The role of spatial organization of cells in erythropoiesis. J Math Biol 2014; 70:71-97. [PMID: 24496930 DOI: 10.1007/s00285-014-0758-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 01/16/2014] [Indexed: 10/25/2022]
Abstract
Erythropoiesis, the process of red blood cell production, occurs mainly in the bone marrow. The functional unit of mammalian erythropoiesis, the erythroblastic island, consists of a central macrophage surrounded by adherent erythroid progenitor cells (CFU-E/Pro-EBs) and their differentiating progeny, the erythroblasts. Central macrophages display on their surface or secrete various growth or inhibitory factors that influence the fate of the surrounding erythroid cells. CFU-E/Pro-EBs have three possible fates: (a) expansion of their numbers without differentiation, (b) differentiation into reticulocytes that are released into the blood, (c) death by apoptosis. CFU-E/Pro-EB fate is under the control of a complex molecular network, that is highly dependent upon environmental conditions in the erythroblastic island. In order to assess the functional role of space coupled with the complex network behavior in erythroblastic islands, we developed hybrid discrete-continuous models of erythropoiesis. A model was developed in which cells are considered as individual physical objects, intracellular regulatory networks are modeled with ordinary differential equations and extracellular concentrations by partial differential equations. We used the model to investigate the impact of an important difference between humans and mice in which mature late-stage erythroblasts produce the most Fas-ligand in humans, whereas early-stage erythroblasts produce the most Fas-ligand in mice. Although the global behaviors of the erythroblastic islands in both species were similar, differences were found, including a relatively slower response time to acute anemia in humans. Also, our modeling approach was very consistent with in vitro culture data, where the central macrophage in reconstituted erythroblastic islands has a strong impact on the dynamics of red blood cell production. The specific spatial organization of erythroblastic islands is key to the normal, stable functioning of mammalian erythropoiesis, both in vitro and in vivo. Our model of a simplified molecular network controlling cell decision provides a realistic functional unit of mammalian erythropoiesis that integrates multiple microenvironmental influences within the erythroblastic island with those of circulating regulators of erythropoiesis, such as EPO and glucocorticosteroids, that are produced at remote sites.
Collapse
Affiliation(s)
- N Eymard
- Institut Camille Jordan, UMR 5208 CNRS, University Lyon 1, Villeurbanne, France,
| | | | | | | | | |
Collapse
|
24
|
Santhanam J, Råberg L, Read AF, Savill NJ. Immune-mediated competition in rodent malaria is most likely caused by induced changes in innate immune clearance of merozoites. PLoS Comput Biol 2014; 10:e1003416. [PMID: 24465193 PMCID: PMC3900382 DOI: 10.1371/journal.pcbi.1003416] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 11/13/2013] [Indexed: 11/18/2022] Open
Abstract
Malarial infections are often genetically diverse, leading to competitive interactions between parasites. A quantitative understanding of the competition between strains is essential to understand a wide range of issues, including the evolution of virulence and drug resistance. In this study, we use dynamical-model based Bayesian inference to investigate the cause of competitive suppression of an avirulent clone of Plasmodium chabaudi (AS) by a virulent clone (AJ) in immuno-deficient and competent mice. We test whether competitive suppression is caused by clone-specific differences in one or more of the following processes: adaptive immune clearance of merozoites and parasitised red blood cells (RBCs), background loss of merozoites and parasitised RBCs, RBC age preference, RBC infection rate, burst size, and within-RBC interference. These processes were parameterised in dynamical mathematical models and fitted to experimental data. We found that just one parameter , the ratio of background loss rate of merozoites to invasion rate of mature RBCs, needed to be clone-specific to predict the data. Interestingly, was found to be the same for both clones in single-clone infections, but different between the clones in mixed infections. The size of this difference was largest in immuno-competent mice and smallest in immuno-deficient mice. This explains why competitive suppression was alleviated in immuno-deficient mice. We found that competitive suppression acts early in infection, even before the day of peak parasitaemia. These results lead us to argue that the innate immune response clearing merozoites is the most likely, but not necessarily the only, mediator of competitive interactions between virulent and avirulent clones. Moreover, in mixed infections we predict there to be an interaction between the clones and the innate immune response which induces changes in the strength of its clearance of merozoites. What this interaction is unknown, but future refinement of the model, challenged with other datasets, may lead to its discovery. Malaria infections often consist of more than one strain of the same parasitic species. Understanding the within-host competition between these various strains is essential to understanding the evolution and epidemiology of drug resistance in malarial infections. The infection process and the competition between strains involve complicated biological processes that are explained by various hypotheses. Mathematical models tested against experimental data provide quantitative measures to compare these hypotheses and enable us to discern the actual biological processes that contribute to the observed dynamics. We use a group of models against experimental data on rodent malaria to test various hypotheses. Such quantitative measures, in understanding rodent malaria, can be considered as a step towards understanding within-host parasite dynamics. Our work presented here demonstrates how confronting mathematical models with data allows the discovery of subtle and novel interactions between hosts and parasites that would be impractical to do in an experiment and allows the rejection of hypotheses that are incorrect. It is our contention that understanding the forces controlling within-host parasite dynamics in well-defined experimental model is a necessary step towards understanding these features in natural infections.
Collapse
Affiliation(s)
- Jayanthi Santhanam
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Labs, Edinburgh, Scotland
- * E-mail:
| | - Lars Råberg
- Department of Biology, Lund University, Lund, Sweden
| | - Andrew F. Read
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Nicholas Jon Savill
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Labs, Edinburgh, Scotland
| |
Collapse
|
25
|
Islam ZU, Bishop SC, Savill NJ, Rowland RRR, Lunney JK, Trible B, Doeschl-Wilson AB. Quantitative analysis of porcine reproductive and respiratory syndrome (PRRS) viremia profiles from experimental infection: a statistical modelling approach. PLoS One 2013; 8:e83567. [PMID: 24358295 PMCID: PMC3866253 DOI: 10.1371/journal.pone.0083567] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/13/2013] [Indexed: 11/18/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most economically significant viral diseases facing the global swine industry. Viremia profiles of PRRS virus challenged pigs reflect the severity and progression of infection within the host and provide crucial information for subsequent control measures. In this study we analyse the largest longitudinal PRRS viremia dataset from an in-vivo experiment. The primary objective was to provide a suitable mathematical description of all viremia profiles with biologically meaningful parameters for quantitative analysis of profile characteristics. The Wood's function, a gamma-type function, and a biphasic extended Wood's function were fit to the individual profiles using Bayesian inference with a likelihood framework. Using maximum likelihood inference and numerous fit criteria, we established that the broad spectrum of viremia trends could be adequately represented by either uni- or biphasic Wood's functions. Three viremic categories emerged: cleared (uni-modal and below detection within 42 days post infection(dpi)), persistent (transient experimental persistence over 42 dpi) and rebound (biphasic within 42 dpi). The convenient biological interpretation of the model parameters estimates, allowed us not only to quantify inter-host variation, but also to establish common viremia curve characteristics and their predictability. Statistical analysis of the profile characteristics revealed that persistent profiles were distinguishable already within the first 21 dpi, whereas it is not possible to predict the onset of viremia rebound. Analysis of the neutralizing antibody(nAb) data indicated that there was a ubiquitous strong response to the homologous PRRSV challenge, but high variability in the range of cross-protection of the nAbs. Persistent pigs were found to have a significantly higher nAb cross-protectivity than pigs that either cleared viremia or experienced rebound within 42 dpi. Our study provides novel insights into the nature and degree of variation of hosts' responses to infection as well as new informative traits for subsequent genomic and modelling studies.
Collapse
Affiliation(s)
- Zeenath U. Islam
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, Midlothian, United Kingdom
- * E-mail:
| | - Stephen C. Bishop
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, Midlothian, United Kingdom
| | - Nicholas J. Savill
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Raymond R. R. Rowland
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, United States of America
| | - Joan K. Lunney
- United State Department of Agriculture, Beltsville Agricultural Research Center, Beltsville, Maryland, United States of America
| | - Benjamin Trible
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, United States of America
| | | |
Collapse
|
26
|
Greischar MA, Read AF, Bjørnstad ON. Synchrony in malaria infections: how intensifying within-host competition can be adaptive. Am Nat 2013; 183:E36-49. [PMID: 24464205 DOI: 10.1086/674357] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Malaria parasites exhibit great diversity in the coordination of their asexual life cycle within the host, ranging from asynchronous growth to tightly synchronized cycles of invasion and emergence from red blood cells. Synchronized reproduction should come at a high cost--intensifying competition among offspring--so why would some Plasmodium species engage in such behavior and others not? We use a delayed differential equation model to show that synchronized infections can be favored when (1) there is limited interference among parasites competing for red blood cells, (2) transmission success is an accelerating function of sexual parasite abundance, (3) the target of saturating immunity is short-lived, and (4) coinfections with asynchronous parasites are rare. As a consequence, synchrony may be beneficial or costly, in line with the diverse patterns of synchronization observed in natural and lab infections. By allowing us to characterize diverse temporal dynamics, the model framework provides a basis for making predictions about disease severity and for projecting evolutionary responses to interventions.
Collapse
Affiliation(s)
- Megan A Greischar
- Center for Infectious Disease Dynamics, Departments of Entomology and Biology, Pennsylvania State University, University Park, Pennsylvania 16802
| | | | | |
Collapse
|
27
|
Strober B, Buonanno M, Clark J, Kawabata T, Tan H, Wolk R, Valdez H, Langley R, Harness J, Menter A, Papp K. Effect of tofacitinib, a Janus kinase inhibitor, on haematological parameters during 12 weeks of psoriasis treatment. Br J Dermatol 2013; 169:992-9. [DOI: 10.1111/bjd.12517] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2013] [Indexed: 01/16/2023]
Affiliation(s)
- B. Strober
- Department of Dermatology; University of Connecticut School of Medicine; Farmington CT 06032 U.S.A
- Probity Medical Research; Waterloo ON Canada
| | | | | | | | - H. Tan
- Pfizer Inc.; Groton CT U.S.A
| | - R. Wolk
- Pfizer Inc.; Groton CT U.S.A
| | | | - R.G. Langley
- Division of Dermatology; Department of Medicine; Dalhousie University; Halifax NS Canada
| | - J. Harness
- Novartis Institutes for Biomedical Research; Basel Switzerland
| | - A. Menter
- Baylor Psoriasis Research Institute; Dallas TX U.S.A
| | - K. Papp
- Probity Medical Research; Waterloo ON Canada
| |
Collapse
|
28
|
Schirm S, Engel C, Loeffler M, Scholz M. A biomathematical model of human erythropoiesis under erythropoietin and chemotherapy administration. PLoS One 2013; 8:e65630. [PMID: 23755260 PMCID: PMC3675041 DOI: 10.1371/journal.pone.0065630] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 04/25/2013] [Indexed: 11/19/2022] Open
Abstract
Anaemia is a common haematologic side effect of dose-dense multi-cycle cytotoxic polychemotherapy requiring erythrocyte transfusions or erythropoietin (EPO) administration. To simulate the effectiveness of different EPO application schedules, we performed both modelling of erythropoiesis under chemotherapy and pharmacokinetic and dynamic modelling of EPO applications in the framework of a single comprehensive biomathematical model. For this purpose, a cell kinetic model of bone marrow erythropoiesis was developed that is based on a set of differential compartment equations describing proliferation and maturation of erythropoietic cell stages. The system is regulated by several feedback loops comprising those mediated by EPO. We added a model of EPO absorption after injection at different sites and a pharmacokinetic model of EPO derivatives to account for the effects of external EPO applications. Chemotherapy is modelled by a transient depletion of bone marrow cell stages. Unknown model parameters were determined by fitting the predictions of the model to data sets of circulating erythrocytes, haemoglobin, haematocrit, percentage of reticulocytes or EPO serum concentrations derived from the literature or cooperating clinical study groups. Parameter fittings resulted in a good agreement of model and data. Depending on site of injection and derivative (Alfa, Beta, Delta, Darbepoetin), nine groups of EPO applications were distinguished differing in either absorption kinetics or pharmacokinetics. Finally, eight different chemotherapy protocols were modelled. The model was validated on the basis of scenarios not used for parameter fitting. Simulations were performed to analyze the impact of EPO applications on the risk of anaemia during chemotherapy. We conclude that we established a model of erythropoiesis under chemotherapy that explains a large set of time series data under EPO and chemotherapy applications. It allows predictions regarding yet untested EPO schedules. Prospective clinical studies are needed to validate model predictions and to explore the feasibility and effectiveness of the proposed schedules.
Collapse
Affiliation(s)
- Sibylle Schirm
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany.
| | | | | | | |
Collapse
|
29
|
Mideo N, Reece SE, Smith AL, Metcalf CJE. The Cinderella syndrome: why do malaria-infected cells burst at midnight? Trends Parasitol 2013; 29:10-6. [PMID: 23253515 PMCID: PMC3925801 DOI: 10.1016/j.pt.2012.10.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 10/31/2012] [Accepted: 10/31/2012] [Indexed: 11/20/2022]
Abstract
An interesting quirk of many malaria infections is that all parasites within a host – millions of them – progress through their cell cycle synchronously. This surprising coordination has long been recognized, yet there is little understanding of what controls it or why it has evolved. Interestingly, the conventional explanation for coordinated development in other parasite species does not seem to apply here. We argue that for malaria parasites, a critical question has yet to be answered: is the coordination due to parasites bursting at the same time or at a particular time? We explicitly delineate these fundamentally different scenarios, possible underlying mechanistic explanations and evolutionary drivers, and discuss the existing corroborating data and key evidence needed to solve this evolutionary mystery.
Collapse
|
30
|
Doeschl-Wilson AB, Bishop SC, Kyriazakis I, Villanueva B. Novel methods for quantifying individual host response to infectious pathogens for genetic analyses. Front Genet 2012; 3:266. [PMID: 23413235 PMCID: PMC3571862 DOI: 10.3389/fgene.2012.00266] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Accepted: 11/05/2012] [Indexed: 11/13/2022] Open
Abstract
We propose two novel approaches for describing and quantifying the response of individual hosts to pathogen challenge in terms of infection severity and impact on host performance. The first approach is a direct extension of the methodology for estimating group tolerance (the change in performance with respect to changes in pathogen burden in a host population) to the level of individuals. The second approach aims to capture the dynamic aspects of individual resistance and tolerance over the entire time course of infections. In contrast to the first approach, which provides a means to disentangle host resistance from tolerance, the second approach focuses on the combined effects of both characteristics. Both approaches provide new individual phenotypes for subsequent genetic analyses and come with specific data requirements. In particular, both approaches rely on the availability of repeated performance and pathogen burden measurements of individuals over the time course of one or several episodes of infection. Consideration of individual tolerance also highlights some of the assumptions hidden within the concept of group tolerance, indicating where care needs to be taken in trait definition and measurement.
Collapse
Affiliation(s)
- Andrea B Doeschl-Wilson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Edinburgh, UK
| | | | | | | |
Collapse
|
31
|
Fischer S, Kurbatova P, Bessonov N, Gandrillon O, Volpert V, Crauste F. Modeling erythroblastic islands: Using a hybrid model to assess the function of central macrophage. J Theor Biol 2012; 298:92-106. [DOI: 10.1016/j.jtbi.2012.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 11/10/2011] [Accepted: 01/03/2012] [Indexed: 11/30/2022]
|
32
|
Mideo N, Reece SE. Plasticity in parasite phenotypes: evolutionary and ecological implications for disease. Future Microbiol 2012; 7:17-24. [DOI: 10.2217/fmb.11.134] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Preventing disease is a major goal of applied bioscience and explaining variation in the harm caused by parasites, and their infectiousness, are major goals of evolutionary biology. The emerging field of evolutionary medicine integrates these two ambitions to inform the development of control strategies that retard or withstand unfavorable parasite evolution. However, as parasites live in hostile and changeable environments – the bodies of other organisms – the success of integrating evolutionary biology with medicine requires a better understanding of how natural selection has solved the problems parasites face. There is increasing appreciation that natural selection shapes parasite strategies to survive in the host and transmit between hosts through facultative (plastic) shifts in parasite traits expressed during infections and in different hosts. This article describes how integrating parasite plasticity into biomedical thinking is central to explaining disease outcomes and transmission patterns, as well as predicting the success of control measures.
Collapse
Affiliation(s)
- Nicole Mideo
- Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK
| | - Sarah E Reece
- Institutes of Evolution, Immunity & Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK
| |
Collapse
|
33
|
Mideo N, Savill NJ, Chadwick W, Schneider P, Read AF, Day T, Reece SE. Causes of variation in malaria infection dynamics: insights from theory and data. Am Nat 2011; 178:E174-E188. [PMID: 22089879 PMCID: PMC3937740 DOI: 10.1086/662670] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Parasite strategies for exploiting host resources are key determinants of disease severity (i.e., virulence) and infectiousness (i.e., transmission between hosts). By iterating the development of theory and empirical tests, we investigated whether variation in parasite traits across two genetically distinct clones of the rodent malaria parasite, Plasmodium chabaudi, explains differences in within-host infection dynamics and virulence. First, we experimentally tested key predictions of our earlier modeling work. As predicted, the more virulent genotype produced more progeny parasites per infected cell (burst size), but in contrast to predictions, invasion rates of red blood cells (RBCs) did not differ between the genotypes studied. Second, we further developed theory by confronting our earlier model with these new data, testing a new set of models that incorporate more biological realism, and developing novel theoretical tools for identifying differences between parasite genotypes. Overall, we found robust evidence that differences in burst sizes contribute to variation in dynamics and that differential interactions between parasites and host immune responses also play a role. In contrast to previous work, our model predicts that RBC age structure is not important for explaining dynamics. Integrating theory and empirical tests is a potentially powerful way of progressing understanding of disease biology.
Collapse
Affiliation(s)
- Nicole Mideo
- Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | - Nicholas J. Savill
- Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
- Institute of Immunity and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | - William Chadwick
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | - Petra Schneider
- Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | - Andrew F. Read
- Center for Infectious Disease Dynamics, Departments of Biology and Entomology, Pennsylvania State University, University Park, Pennsylvania 16802; and Fogarty International Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Troy Day
- Departments of Biology and Mathematics and Statistics, Queen’s University, Kingston, Ontario K7L 3N6, Canada
| | - Sarah E. Reece
- Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
- Institute of Immunity and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| |
Collapse
|
34
|
Yung S, Ledran M, Moreno-Gimeno I, Conesa A, Montaner D, Dopazo J, Dimmick I, Slater NJ, Marenah L, Real PJ, Paraskevopoulou I, Bisbal V, Burks D, Santibanez-Koref M, Moreno R, Mountford J, Menendez P, Armstrong L, Lako M. Large-scale transcriptional profiling and functional assays reveal important roles for Rho-GTPase signalling and SCL during haematopoietic differentiation of human embryonic stem cells. Hum Mol Genet 2011; 20:4932-46. [PMID: 21937587 DOI: 10.1093/hmg/ddr431] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Understanding the transcriptional cues that direct differentiation of human embryonic stem cells (hESCs) and human-induced pluripotent stem cells to defined and functional cell types is essential for future clinical applications. In this study, we have compared transcriptional profiles of haematopoietic progenitors derived from hESCs at various developmental stages of a feeder- and serum-free differentiation method and show that the largest transcriptional changes occur during the first 4 days of differentiation. Data mining on the basis of molecular function revealed Rho-GTPase signalling as a key regulator of differentiation. Inhibition of this pathway resulted in a significant reduction in the numbers of emerging haematopoietic progenitors throughout the differentiation window, thereby uncovering a previously unappreciated role for Rho-GTPase signalling during human haematopoietic development. Our analysis indicated that SCL was the 11th most upregulated transcript during the first 4 days of the hESC differentiation process. Overexpression of SCL in hESCs promoted differentiation to meso-endodermal lineages, the emergence of haematopoietic and erythro-megakaryocytic progenitors and accelerated erythroid differentiation. Importantly, intrasplenic transplantation of SCL-overexpressing hESC-derived haematopoietic cells enhanced recovery from induced acute anaemia without significant cell engraftment, suggesting a paracrine-mediated effect.
Collapse
Affiliation(s)
- Sun Yung
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Metcalf CJE, Graham AL, Huijben S, Barclay VC, Long GH, Grenfell BT, Read AF, Bjørnstad ON. Partitioning regulatory mechanisms of within-host malaria dynamics using the effective propagation number. Science 2011; 333:984-8. [PMID: 21852493 PMCID: PMC3891600 DOI: 10.1126/science.1204588] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Immune clearance and resource limitation (via red blood cell depletion) shape the peaks and troughs of malaria parasitemia, which in turn affect disease severity and transmission. Quantitatively partitioning the relative roles of these effects through time is challenging. Using data from rodent malaria, we estimated the effective propagation number, which reflects the relative importance of contrasting within-host control mechanisms through time and is sensitive to the inoculating parasite dose. Our analysis showed that the capacity of innate responses to restrict initial parasite growth saturates with parasite dose and that experimentally enhanced innate immunity can affect parasite density indirectly via resource depletion. Such a statistical approach offers a tool to improve targeting of drugs or vaccines for human therapy by revealing the dynamics and interactions of within-host regulatory mechanisms.
Collapse
Affiliation(s)
- C J E Metcalf
- Department of Zoology, Oxford University, Oxford OX1 3PS, UK.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Marković SD, Zižić JB, Obradović AD, Ognjanović BI, Stajn AS, Saičić ZS, Spasić MB. Energy production and redox status of rat red blood cells after reticulocytosis induced by various treatments. ACTA BIOLOGICA HUNGARICA 2011; 62:122-32. [PMID: 21555264 DOI: 10.1556/abiol.62.2011.2.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Stimulated erythropoiesis and reticulocytosis can be induced by daily bleeding, or by phenylhydrazine (PHZ) treatment. We compared the in vivo effects of PHZ and bleeding treatment on haematological, energy and redox status parameters in red blood cells (RBC) of rats. The results showed that all followed haematological parameters were significantly lower in bleeding, compared to PHZ-treated rats. PHZ induced even 2.58-fold higher reticulocytosis as compared to bleeding treatment. Although PHZ induced higher reticulocytosis, respiration intensity and energy production was lower than in bleeding-induced reticulocytes. These alterations were the consequence of increased superoxide anion and peroxynitrite concentrations in PHZ-treated rats. Bleeding treatment resulted in increased activity of an antioxidative enzyme, superoxide dismutase. In conclusion, differences in these two experimental models for reticulocytosis may be used as tools for appropriate pharmacological testing of redox-active substances considering energy and redox processes, as well as apoptosis pathways.
Collapse
Affiliation(s)
- Snežana D Marković
- Department for Biology and Ecology, University of Kragujevac Faculty of Science, Kragujevac Serbia.
| | | | | | | | | | | | | |
Collapse
|
37
|
Krishnan SM, Dixit NM. Ribavirin-induced anemia in hepatitis C virus patients undergoing combination therapy. PLoS Comput Biol 2011; 7:e1001072. [PMID: 21304937 PMCID: PMC3033369 DOI: 10.1371/journal.pcbi.1001072] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 12/29/2010] [Indexed: 01/11/2023] Open
Abstract
The current standard of care for hepatitis C virus (HCV) infection – combination therapy with pegylated interferon and ribavirin – elicits sustained responses in only ∼50% of the patients treated. No alternatives exist for patients who do not respond to combination therapy. Addition of ribavirin substantially improves response rates to interferon and lowers relapse rates following the cessation of therapy, suggesting that increasing ribavirin exposure may further improve treatment response. A key limitation, however, is the toxic side-effect of ribavirin, hemolytic anemia, which often necessitates a reduction of ribavirin dosage and compromises treatment response. Maximizing treatment response thus requires striking a balance between the antiviral and hemolytic activities of ribavirin. Current models of viral kinetics describe the enhancement of treatment response due to ribavirin. Ribavirin-induced anemia, however, remains poorly understood and precludes rational optimization of combination therapy. Here, we develop a new mathematical model of the population dynamics of erythrocytes that quantitatively describes ribavirin-induced anemia in HCV patients. Based on the assumption that ribavirin accumulation decreases erythrocyte lifespan in a dose-dependent manner, model predictions capture several independent experimental observations of the accumulation of ribavirin in erythrocytes and the resulting decline of hemoglobin in HCV patients undergoing combination therapy, estimate the reduced erythrocyte lifespan during therapy, and describe inter-patient variations in the severity of ribavirin-induced anemia. Further, model predictions estimate the threshold ribavirin exposure beyond which anemia becomes intolerable and suggest guidelines for the usage of growth hormones, such as erythropoietin, that stimulate erythrocyte production and avert the reduction of ribavirin dosage, thereby improving treatment response. Our model thus facilitates, in conjunction with models of viral kinetics, the rational identification of treatment protocols that maximize treatment response while curtailing side effects. The treatment of HCV infection poses a major global health-care challenge today. The current standard of care, combination therapy with interferon and ribavirin, works in only about half of the patients treated. Because no alternatives are available yet for patients in whom combination therapy fails, identifying ways to improve response to combination therapy is critical. Increasing exposure to ribavirin does improve response but is associated with the severe side-effect, anemia. One way to maximize treatment response therefore is to increase ribavirin exposure to levels just below where anemia becomes intolerable. A second way is to supplement combination therapy with growth hormones, such as erythropoietin, that increase the production of red blood cells (erythrocytes) and compensate for ribavirin-induced anemia. Rational optimization of combination therapy thus relies on a quantitative description of ribavirin-induced anemia, which is currently lacking. Here, we develop a model of the population dynamics of erythrocytes in individuals exposed to ribavirin that quantitatively describes ribavirin-induced anemia. Model predictions capture several independent observations of ribavirin-induced anemia in HCV patients undergoing combination therapy, estimate the threshold ribavirin exposure beyond which anemia becomes intolerable, suggest guidelines for the usage of growth hormones, and facilitate rational optimization of therapy.
Collapse
Affiliation(s)
- Sheeja M. Krishnan
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
| | - Narendra M. Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
- Bioinformatics Centre, Indian Institute of Science, Bangalore, India
- * E-mail:
| |
Collapse
|
38
|
Abstract
Red blood cells (RBCs) often have a short circulating half-life in hemodialysis patients, which increases the difficulty of achieving a stable hemoglobin level. Fluctuations in erythropoietin (EPO) levels contribute to this increased RBC turnover because a decline in the level of EPO triggers the preferential destruction of newly-formed RBC, a process termed neocytolysis. The RBCs that are released during the treatment of renal anemia are often hypochromic, with a low content of iron; these RBCs are vulnerable to rapid turnover because iron-deficiency affects RBCs in several ways, such as, increased exposure of the phagocytic signaling molecule phosphatidylserine, loss of deformability, and increased oxidative stress. Both EPO fluctuation and the release of iron-deficient RBCs are characteristic events occurring during the management of renal anemia, and the shorter RBC lifetime is a component of the large fluctuations in hemoglobin level seen in patients on hemodialysis.
Collapse
Affiliation(s)
- Garry J Handelman
- Clinical Lab and Nutrition Sciences, University of Massachusetts, Lowell, MA 01854, USA.
| | | |
Collapse
|
39
|
O'Donnell AJ, Schneider P, McWatters HG, Reece SE. Fitness costs of disrupting circadian rhythms in malaria parasites. Proc Biol Sci 2011; 278:2429-36. [PMID: 21208950 PMCID: PMC3125626 DOI: 10.1098/rspb.2010.2457] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Circadian biology assumes that biological rhythms maximize fitness by enabling organisms to coordinate with their environment. Despite circadian clocks being such a widespread phenomenon, demonstrating the fitness benefits of temporal coordination is challenging and such studies are rare. Here, we tested the consequences--for parasites--of being temporally mismatched to host circadian rhythms using the rodent malaria parasite, Plasmodium chabaudi. The cyclical nature of malaria infections is well known, as the cell cycles across parasite species last a multiple of approximately 24 h, but the evolutionary explanations for periodicity are poorly understood. We demonstrate that perturbation of parasite rhythms results in a twofold cost to the production of replicating and transmission stages. Thus, synchronization with host rhythms influences in-host survival and between-host transmission potential, revealing a role for circadian rhythms in the evolution of host-parasite interactions. More generally, our results provide a demonstration of the adaptive value of circadian rhythms and the utility of using an evolutionary framework to understand parasite traits.
Collapse
Affiliation(s)
- Aidan J O'Donnell
- Institute of Evolution, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3JT, UK
| | | | | | | |
Collapse
|
40
|
Miller MR, Råberg L, Read AF, Savill NJ. Quantitative analysis of immune response and erythropoiesis during rodent malarial infection. PLoS Comput Biol 2010; 6:e1000946. [PMID: 20941388 PMCID: PMC2947982 DOI: 10.1371/journal.pcbi.1000946] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Accepted: 08/31/2010] [Indexed: 12/20/2022] Open
Abstract
Malarial infection is associated with complex immune and erythropoietic responses in the host. A quantitative understanding of these processes is essential to help inform malaria therapy and for the design of effective vaccines. In this study, we use a statistical model-fitting approach to investigate the immune and erythropoietic responses in Plasmodium chabaudi infections of mice. Three mouse phenotypes (wildtype, T-cell-deficient nude mice, and nude mice reconstituted with T-cells taken from wildtype mice) were infected with one of two parasite clones (AS or AJ). Under a Bayesian framework, we use an adaptive population-based Markov chain Monte Carlo method and fit a set of dynamical models to observed data on parasite and red blood cell (RBC) densities. Model fits are compared using Bayes' factors and parameter estimates obtained. We consider three independent immune mechanisms: clearance of parasitised RBCs (pRBC), clearance of unparasitised RBCs (uRBC), and clearance of parasites that burst from RBCs (merozoites). Our results suggest that the immune response of wildtype mice is associated with less destruction of uRBCs, compared to the immune response of nude mice. There is a greater degree of synchronisation between pRBC and uRBC clearance than between either mechanism and merozoite clearance. In all three mouse phenotypes, control of the peak of parasite density is associated with pRBC clearance. In wildtype mice and AS-infected nude mice, control of the peak is also associated with uRBC clearance. Our results suggest that uRBC clearance, rather than RBC infection, is the major determinant of RBC dynamics from approximately day 12 post-innoculation. During the first 2-3 weeks of blood-stage infection, immune-mediated clearance of pRBCs and uRBCs appears to have a much stronger effect than immune-mediated merozoite clearance. Upregulation of erythropoiesis is dependent on mouse phenotype and is greater in wildtype and reconstitited mice. Our study highlights the informative power of statistically rigorous model-fitting techniques in elucidating biological systems.
Collapse
Affiliation(s)
- Martin R. Miller
- Centre for Infectious Diseases, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Lars Råberg
- Department of Animal Ecology, Lund University, Lund, Sweden
| | - Andrew F. Read
- Center for Infectious Disease Dynamics and Departments of Biology and Entomology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Nicholas J. Savill
- Centre for Infectious Diseases, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
41
|
Pasini EM, Kirkegaard M, Mortensen P, Mann M, Thomas AW. Deep-coverage rhesus red blood cell proteome: a first comparison with the human and mouse red blood cell. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2010; 8 Suppl 3:s126-39. [PMID: 20606743 PMCID: PMC2897199 DOI: 10.2450/2010.020s] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
BACKGROUND Macaques are the closest evolutionary relatives of humans routinely used in basic and applied biomedical research. Their genetic, physiological, immunological and metabolic similarity to humans, second only to that of the great apes, makes them invaluable models of human disease. These similarities also mean that macaques are often the only experimental models available for evaluating increasingly specific drugs in development, and as a proof-of-concept bridge can help reduce the numbers of compounds that fail in clinical pharmaceutical research. In vertebrates, red blood cells (RBCs) diseases are frequently severe as their role as sole gas transporter makes them indispensable to survival; much research has therefore focused on an in-depth understanding of the functioning of the RBC. RBCs also host malaria, babesia and other parasites. Recently, we presented an in-depth proteome for the human RBC and a comparative human/mouse RBC proteome. MATERIAL AND METHODS Here, we present directly comparable data for the human, mouse and rhesus RBC proteomes. All proteins were identified, validated and categorized in terms of sub-cellular localization, protein family and function and, in comparison with the human and mouse RBC, were classified as orthologues, family-related or unique. Splice isoforms were identified and polypeptides migrating with anomalous apparent molecular weights were grouped into putatively ubiquitinylated or partially degraded complexes. RESULTS AND DISCUSSION Overall there was close concordance between mouse, human and rhesus proteomes, confirming the unexpected RBC complexity. Several novel findings in the human and mouse proteomes have been confirmed here. This comparison sheds light on several open issues in RBC biology and provides a departure point for more comprehensive understanding of RBC function.
Collapse
Affiliation(s)
- Erica M. Pasini
- Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Morten Kirkegaard
- Center for Experimental Bioinformatics, University of Southern Denmark, Odense, Denmark
| | - Peter Mortensen
- Center for Experimental Bioinformatics, University of Southern Denmark, Odense, Denmark
| | - Matthias Mann
- Center for Experimental Bioinformatics, University of Southern Denmark, Odense, Denmark
- Dept. of Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, Martinsried, Germany
| | - Alan W. Thomas
- Biomedical Primate Research Centre, Rijswijk, The Netherlands
- Correspondence: Alan Thomas, Dept. Parasitology, Biomedical Primate Research Centre, Lange Kleiweg 139, 2288 GJ Rijswijk, The Netherlands, E-mail:
| |
Collapse
|