Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Parasite Immunol ; 44(3): e12898, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34778983

RESUMO

AIMS: Malaria parasites exhibit daily rhythms in the intra-erythrocytic development cycle (IDC) that underpins asexual replication in the blood. The IDC schedule is aligned with the timing of host feeding-fasting rhythms. When the IDC schedule is perturbed to become mismatched to host rhythms, it readily reschedules but it is not known how. METHODS: We intensively follow four groups of infections that have different temporal alignments between host rhythms and the IDC schedule for 10 days, before and after the peak in asexual densities. We compare how the duration, synchrony and timing of the IDC differs between parasites in control infections and those forced to reschedule by 12 hours and ask whether the density of parasites affects the rescheduling process. RESULTS AND CONCLUSIONS: Our experiments reveal parasites shorten the IDC duration by 2-3 hours to become realigned to host feeding-fasting rhythms with 5-6 days, in a density-independent manner. Furthermore, parasites are able to reschedule without significant fitness costs for them or their hosts. Understanding the extent of, and limits on, plasticity in the IDC schedule may reveal targets for novel interventions, such as drugs to disrupt IDC regulation and preventing IDC dormancy conferring tolerance to existing drugs.


Assuntos
Malária , Parasitos , Plasmodium chabaudi , Animais , Ritmo Circadiano/fisiologia , Jejum , Malária/parasitologia , Malária/prevenção & controle , Plasmodium chabaudi/fisiologia
2.
PLoS Comput Biol ; 16(10): e1008211, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33031367

RESUMO

To understand why some hosts get sicker than others from the same type of infection, it is essential to explain how key processes, such as host responses to infection and parasite growth, are influenced by various biotic and abiotic factors. In many disease systems, the initial infection dose impacts host morbidity and mortality. To explore drivers of dose-dependence and individual variation in infection outcomes, we devised a mathematical model of malaria infection that allowed host and parasite traits to be linear functions (reaction norms) of the initial dose. We fitted the model, using a hierarchical Bayesian approach, to experimental time-series data of acute Plasmodium chabaudi infection across doses spanning seven orders of magnitude. We found evidence for both dose-dependent facilitation and debilitation of host responses. Most importantly, increasing dose reduced the strength of activation of indiscriminate host clearance of red blood cells while increasing the half-life of that response, leading to the maximal response at an intermediate dose. We also explored the causes of diverse infection outcomes across replicate mice receiving the same dose. Besides random noise in the injected dose, we found variation in peak parasite load was due to unobserved individual variation in host responses to clear infected cells. Individual variation in anaemia was likely driven by random variation in parasite burst size, which is linked to the rate of host cells lost to malaria infection. General host vigour in the absence of infection was also correlated with host health during malaria infection. Our work demonstrates that the reaction norm approach provides a useful quantitative framework for examining the impact of a continuous external factor on within-host infection processes.


Assuntos
Interações Hospedeiro-Parasita , Malária , Anemia/complicações , Animais , Teorema de Bayes , Biologia Computacional , Feminino , Malária/complicações , Malária/imunologia , Malária/parasitologia , Malária/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Carga Parasitária , Plasmodium chabaudi/patogenicidade , Plasmodium chabaudi/fisiologia
3.
PLoS Pathog ; 14(11): e1007371, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30427935

RESUMO

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.


Assuntos
Adaptação Fisiológica/fisiologia , Malária/parasitologia , Plasmodium/fisiologia , Adaptação Biológica/fisiologia , Animais , Evolução Biológica , Simulação por Computador , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Modelos Teóricos , Parasitos , Plasmodium chabaudi/fisiologia , Reprodução/fisiologia , Reprodução Assexuada/fisiologia
4.
Oecologia ; 190(3): 511-522, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30953168

RESUMO

Oviposition site choice affects a host of offspring phenotypes and directly impacts maternal fitness. Recent evidence suggests that oviparous reptiles often select nest sites where the landscape has been altered by anthropogenic activity, whereas natural nest sites are less often used. We leverage a long-term study of snapping turtle (Chelydra serpentina) to identify natural nest sites and anthropogenic nest sites and to compare habitat variables among nest site types. Natural and anthropogenic nest sites did not differ in average canopy closure, distance to nearest water, substrate composition, or aspect. However, anthropogenic nest sites had less ground-level vegetation and greater soil brightness, and were 3.3 °C warmer than natural nests during incubation. We used the Schoolfield model of poikilotherm development to assess differences in development rate between natural and anthropogenic nests. Because of the difference in temperature, embryos in anthropogenic nests were predicted to have undergone nearly twice as much development as embryos in natural nests during incubation. We outline why the evolution of fast embryonic development rate cannot compensate indefinitely for the low temperature incubation regimes that become increasingly prevalent at northern range margins, thereby underlining why maternal nest site choice of relatively warm anthropogenic sites may help oviparous reptiles persist in thermally constrained environments. Future research should aim to quantify both the thermal benefits of anthropogenic nest sites, as well as associated fitness costs (e.g., increased adult mortality) to elucidate whether anthropogenic disturbance of the landscape can be an ecological trap or serve a net benefit to some reptiles in northern environments.


Assuntos
Oviparidade , Répteis , Animais , Ecossistema , Feminino , Fenótipo , Temperatura
5.
PLoS Comput Biol ; 12(2): e1004718, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26890485

RESUMO

Many microparasites infect new hosts with specialized life stages, requiring a subset of the parasite population to forgo proliferation and develop into transmission forms. Transmission stage production influences infectivity, host exploitation, and the impact of medical interventions like drug treatment. Predicting how parasites will respond to public health efforts on both epidemiological and evolutionary timescales requires understanding transmission strategies. These strategies can rarely be observed directly and must typically be inferred from infection dynamics. Using malaria as a case study, we test previously described methods for inferring transmission stage investment against simulated data generated with a model of within-host infection dynamics, where the true transmission investment is known. We show that existing methods are inadequate and potentially very misleading. The key difficulty lies in separating transmission stages produced by different generations of parasites. We develop a new approach that performs much better on simulated data. Applying this approach to real data from mice infected with a single Plasmodium chabaudi strain, we estimate that transmission investment varies from zero to 20%, with evidence for variable investment over time in some hosts, but not others. These patterns suggest that, even in experimental infections where host genetics and other environmental factors are controlled, parasites may exhibit remarkably different patterns of transmission investment.


Assuntos
Malária/parasitologia , Malária/transmissão , Plasmodium chabaudi/fisiologia , Plasmodium chabaudi/patogenicidade , Animais , Biologia Computacional , Eritrócitos/parasitologia , Feminino , Estágios do Ciclo de Vida , Masculino , Camundongos , Modelos Biológicos
6.
Parasitology ; 143(7): 905-914, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26399436

RESUMO

Mathematical modelling provides an effective way to challenge conventional wisdom about parasite evolution and investigate why parasites 'do what they do' within the host. Models can reveal when intuition cannot explain observed patterns, when more complicated biology must be considered, and when experimental and statistical methods are likely to mislead. We describe how models of within-host infection dynamics can refine experimental design, and focus on the case study of malaria to highlight how integration between models and data can guide understanding of parasite fitness in three areas: (1) the adaptive significance of chronic infections; (2) the potential for tradeoffs between virulence and transmission; and (3) the implications of within-vector dynamics. We emphasize that models are often useful when they highlight unexpected patterns in parasite evolution, revealing instead why intuition yields the wrong answer and what combination of theory and data are needed to advance understanding.


Assuntos
Evolução Biológica , Interações Hospedeiro-Parasita/fisiologia , Modelos Biológicos , Animais , Humanos , Malária/parasitologia , Pesquisa/normas , Pesquisa/tendências
7.
Am Nat ; 183(2): E36-49, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24464205

RESUMO

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.


Assuntos
Malária/transmissão , Modelos Biológicos , Plasmodium chabaudi/patogenicidade , Adaptação Fisiológica , Animais , Culicidae/parasitologia , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Humanos , Malária/parasitologia , Plasmodium chabaudi/fisiologia
8.
Evolution ; 78(7): 1287-1301, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38581661

RESUMO

For parasites, robust proliferation within hosts is crucial for establishing the infection and creating opportunities for onward transmission. While faster proliferation enhances transmission rates, it is often assumed to curtail transmission duration by killing the host (virulence), a trade-off constraining parasite evolution. Yet in many diseases, including malaria, the preponderance of infections with mild or absent symptoms suggests that host mortality is not a sufficient constraint, raising the question of what restrains evolution toward faster proliferation. In malaria infections, the maximum rate of proliferation is determined by the burst size, the number of daughter parasites produced per infected red blood cell. Larger burst sizes should expand the pool of infected red blood cells that can be used to produce the specialized transmission forms needed to infect mosquitoes. We use a within-host model parameterized for rodent malaria parasites (Plasmodium chabaudi) to project the transmission consequences of burst size, focusing on initial acute infection where resource limitation and risk of host mortality are greatest. We find that resource limitation restricts evolution toward higher burst sizes below the level predicted by host mortality alone. Our results suggest resource limitation could represent a more general constraint than virulence-transmission trade-offs, preventing evolution towards faster proliferation.


Assuntos
Malária , Plasmodium chabaudi , Animais , Virulência , Plasmodium chabaudi/genética , Plasmodium chabaudi/patogenicidade , Plasmodium chabaudi/fisiologia , Malária/transmissão , Malária/parasitologia , Malária/prevenção & controle , Interações Hospedeiro-Parasita , Evolução Biológica , Eritrócitos/parasitologia , Modelos Biológicos
9.
Trends Parasitol ; 39(8): 626-637, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37336700

RESUMO

For pathogenic organisms, faster rates of multiplication promote transmission success, the potential to harm hosts, and the evolution of drug resistance. Parasite multiplication rates (PMRs) are often quantified in malaria infections, given the relative ease of sampling. Using modern and historical human infection data, we show that established methods return extraordinarily - and implausibly - large PMRs. We illustrate how inflated PMRs arise from two facets of malaria biology that are far from unique: (i) some developmental ages are easier to sample than others; (ii) the distribution of developmental ages changes over the course of infection. The difficulty of accurately quantifying PMRs demonstrates a need for robust methods and a subsequent re-evaluation of what is known even in the well-studied system of malaria.


Assuntos
Malária Falciparum , Malária , Parasitos , Animais , Humanos , Malária Falciparum/parasitologia , Plasmodium falciparum , Malária/parasitologia
10.
J R Soc Interface ; 18(177): 20210065, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33906391

RESUMO

Inferring biological processes from population dynamics is a common challenge in ecology, particularly when faced with incomplete data. This challenge extends to inferring parasite traits from within-host infection dynamics. We focus on rodent malaria infections (Plasmodium berghei), a system for which previous work inferred an immune-mediated extension in the length of the parasite development cycle within red blood cells. By developing a system of delay-differential equations to describe within-host infection dynamics and simulating data, we demonstrate the potential to obtain biased estimates of parasite (and host) traits when key biological processes are not considered. Despite generating infection dynamics using a fixed parasite developmental cycle length, we find that known sources of measurement bias in parasite stage and abundance data can affect estimates of parasite developmental duration, with stage misclassification driving inferences about extended cycle length. We discuss alternative protocols and statistical methods that can mitigate such misestimation.


Assuntos
Fenômenos Biológicos , Malária , Parasitos , Animais , Eritrócitos , Plasmodium berghei
11.
Elife ; 102021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34636723

RESUMO

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.


Assuntos
Eritrócitos/parasitologia , Malária/parasitologia , Plasmodium chabaudi/patogenicidade , Animais , Simulação por Computador , Citocinas/sangue , Modelos Animais de Doenças , Suscetibilidade a Doenças , Interações Hospedeiro-Parasita , Mediadores da Inflamação/sangue , Malária/sangue , Malária/genética , Malária/imunologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Modelos Imunológicos , Carga Parasitária , Plasmodium chabaudi/imunologia , Índice de Gravidade de Doença , Especificidade da Espécie , Fatores de Tempo
12.
Evol Med Public Health ; 2020(1): 30-34, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32099654

RESUMO

Lay Summary: Competition often occurs among diverse parasites within a single host, but control efforts could change its strength. We examined how the interplay between competition and control could shape the evolution of parasite traits like drug resistance and disease severity.

13.
Genetics ; 180(4): 1889-907, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18940790

RESUMO

The Mre11/Rad50/Nbs1 (MRN) complex is required for eukaryotic DNA double-strand break (DSB) repair and meiotic recombination. We cloned the Coprinus cinereus rad50 gene and showed that it corresponds to the complementation group previously named rad12, identified mutations in 15 rad50 alleles, and mapped two of the mutations onto molecular models of Rad50 structure. We found that C. cinereus rad50 and mre11 mutants arrest in meiosis and that this arrest is Spo11 dependent. In addition, some rad50 alleles form inducible, Spo11-dependent Rad51 foci and therefore must be forming meiotic DSBs. Thus, we think it likely that arrest in both mre11-1 and the collection of rad50 mutants is the result of unrepaired or improperly processed DSBs in the genome and that Rad50 and Mre11 are dispensable in C. cinereus for DSB formation, but required for appropriate DSB processing. We found that the ability of rad50 mutant strains to form Rad51 foci correlates with their ability to promote synaptonemal complex formation and with levels of stable meiotic pairing and that partial pairing, recombination initiation, and synapsis occur in the absence of wild-type Rad50 catalytic domains. Examination of single- and double-mutant strains showed that a spo11 mutation that prevents DSB formation enhances axial element (AE) formation for rad50-4, an allele predicted to encode a protein with intact hook region and hook-proximal coiled coils, but not for rad50-1, an allele predicted to encode a severely truncated protein, or for rad50-5, which encodes a protein whose hook-proximal coiled-coil region is disrupted. Therefore, Rad50 has an essential structural role in the formation of AEs, separate from the DSB-processing activity of the MRN complex.


Assuntos
Coprinus/genética , Proteínas Fúngicas/genética , Meiose/genética , Mutação , Recombinação Genética/genética , Complexo Sinaptonêmico/metabolismo , Alelos , Coprinus/metabolismo , Reparo do DNA , DNA Fúngico/metabolismo , Proteínas Fúngicas/metabolismo , Microscopia Eletrônica , Esporos Fúngicos/crescimento & desenvolvimento , Complexo Sinaptonêmico/genética , Complexo Sinaptonêmico/ultraestrutura
14.
Evolution ; 73(11): 2175-2188, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31495911

RESUMO

Vector-borne parasites must succeed at three scales to persist: they must proliferate within a host, establish in vectors, and transmit back to hosts. Ecology outside the host undergoes dramatic seasonal and human-induced changes, but predicting parasite evolutionary responses requires integrating their success across scales. We develop a novel, data-driven model to titrate the evolutionary impact of ecology at multiple scales on human malaria parasites. We investigate how parasites invest in transmission versus proliferation, a life-history trait that influences disease severity and spread. We find that transmission investment controls the pattern of host infectiousness over the course of infection: a trade-off emerges between early and late infectiousness, and the optimal resolution of that trade-off depends on ecology outside the host. An expanding epidemic favors rapid proliferation, and can overwhelm the evolutionary influence of host recovery rates and mosquito population dynamics. If transmission investment and recovery rate are positively correlated, then ecology outside the host imposes potent selection for aggressive parasite proliferation at the expense of transmission. Any association between transmission investment and recovery represents a key unknown, one that is likely to influence whether the evolutionary consequences of interventions are beneficial or costly for human health.


Assuntos
Evolução Molecular , Malária/transmissão , Modelos Genéticos , Plasmodium falciparum/genética , Biomassa , Interações Hospedeiro-Parasita/genética , Humanos , Malária/parasitologia , Mosquitos Vetores/parasitologia , Mosquitos Vetores/fisiologia , Plasmodium falciparum/patogenicidade , Plasmodium falciparum/fisiologia , Reprodução/genética , Seleção Genética , Virulência/genética
15.
Trends Parasitol ; 35(5): 341-355, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30952484

RESUMO

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.


Assuntos
Malária/parasitologia , Parasitologia/métodos , Plasmodium/fisiologia , Humanos , Estágios do Ciclo de Vida/fisiologia
16.
Epidemics ; 30: 100382, 2019 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-32004794

RESUMO

Identifying ecological drivers of disease transmission is central to understanding disease risks. For vector-borne diseases, temperature is a major determinant of transmission because vital parameters determining the fitness of parasites and vectors are highly temperature-sensitive, including the extrinsic incubation period required for parasites to develop within the vector. Temperature also underlies dramatic differences in the individual-level variation in the extrinsic incubation period, yet the influence of this variation in disease transmission is largely unexplored. We incorporate empirical estimates of dengue virus extrinsic incubation period and its variation across a range of temperatures into a stochastic model to examine the consequences for disease emergence. We find that such variation impacts the probability of disease emergence because exceptionally rapid, but empirically observed incubation - typically ignored by modelling only the average - increases the chance of disease emergence even at the limits of the temperature range for dengue transmission. We show that variation in the extrinsic incubation period causes the greatest proportional increase in the risk of disease emergence at cooler temperatures where the mean incubation period is long, and associated variation is large. Thus, ignoring EIP variation will likely lead to underestimation of the risk of vector-borne disease emergence in temperate climates.

17.
Evol Appl ; 11(4): 442-455, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29636798

RESUMO

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.

18.
Ecol Lett ; 10(5): 418-34, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17498141

RESUMO

The study of parasite local adaptation, whereby parasites perform better on sympatric hosts than on allopatric hosts and/or better on their own host population than do other parasites, is of great importance to both basic and applied biology. Theoretical examination of host-parasite coevolution predicts that parasite migration rate, generation time and virulence all contribute to the pattern of parasite local adaptation, such that parasites with greater dispersal ability, more frequent reproduction and/or high virulence ought to exhibit increased infectivity on local hosts. Here, we present a meta-analysis of experimental work from 57 host-parasite systems across 54 local adaptation studies to directly test theoretical predictions concerning the effect of each attribute on parasite adaptation. As expected, we find that studies of parasites with higher migration rates than their hosts report local adaptation, as measured by infection success, significantly more often than studies of parasites with relatively low migration rates. Furthermore, this synthesis serves to identify biases in the current body of work and highlight areas with the greatest need for further study. We emphasize the importance of unifying the field with regard to experimental methods, local adaptation definitions and reported statistics for cross-infection studies.


Assuntos
Adaptação Fisiológica , Parasitos/fisiologia , Animais , Comportamento Animal
19.
PLoS Negl Trop Dis ; 11(10): e0005956, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28991904

RESUMO

Blood-feeding arthropods-like mosquitoes, sand flies, and ticks-transmit many diseases that impose serious public health and economic burdens. When a blood-feeding arthropod bites a mammal, it injects saliva containing immunogenic compounds that facilitate feeding. Evidence from Leishmania, Plasmodium and arboviral infections suggests that the immune responses elicited by pre-exposure to arthropod saliva can alter disease progression if the host later becomes infected. Such pre-sensitisation of host immunity has been reported to both exacerbate and limit infection symptoms, depending on the system in question, with potential implications for recovery. To explore if and how immune pre-sensitisation alters the effects of vector control, we develop a general model of vector-borne disease. We show that the abundance of pre-sensitised infected hosts should increase when control efforts moderately increase vector mortality rates. If immune pre-sensitisation leads to more rapid clearance of infection, increasing vector mortality rates may achieve greater than expected disease control. However, when immune pre-sensitisation prolongs the duration of infection, e.g., through mildly symptomatic cases for which treatment is unlikely to be sought, vector control can actually increase the total number of infected hosts. The rising infections may go unnoticed unless active surveillance methods are used to detect such sub-clinical individuals, who could provide long-lasting reservoirs for transmission and suffer long-term health consequences of those sub-clinical infections. Sensitivity analysis suggests that these negative consequences could be mitigated through integrated vector management. While the effect of saliva pre-exposure on acute symptoms is well-studied for leishmaniasis, the immunological and clinical consequences are largely uncharted for other vector-parasite-host combinations. We find a large range of plausible epidemiological outcomes, positive and negative for public health, underscoring the need to quantify how immune pre-sensitisation modulates recovery and transmission rates in vector-borne diseases.


Assuntos
Vetores Artrópodes/imunologia , Modelos Biológicos , Saliva/imunologia , Animais , Culicidae/imunologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Tolerância Imunológica , Mordeduras e Picadas de Insetos/imunologia , Psychodidae/imunologia , Carrapatos/imunologia
20.
Evolution ; 70(7): 1542-58, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27271841

RESUMO

In vertebrate hosts, malaria parasites face a tradeoff between replicating and the production of transmission stages that can be passed onto mosquitoes. This tradeoff is analogous to growth-reproduction tradeoffs in multicellular organisms. We use a mathematical model tailored to the life cycle and dynamics of malaria parasites to identify allocation strategies that maximize cumulative transmission potential to mosquitoes. We show that plastic strategies can substantially outperform fixed allocation because parasites can achieve greater fitness by investing in proliferation early and delaying the production of transmission stages. Parasites should further benefit from restraining transmission investment later in infection, because such a strategy can help maintain parasite numbers in the face of resource depletion. Early allocation decisions are predicted to have the greatest impact on parasite fitness. If the immune response saturates as parasite numbers increase, parasites should benefit from even longer delays prior to transmission investment. The presence of a competing strain selects for consistently lower levels of transmission investment and dramatically increased exploitation of the red blood cell resource. While we provide a detailed analysis of tradeoffs pertaining to malaria life history, our approach for identifying optimal plastic allocation strategies may be broadly applicable.


Assuntos
Culicidae/parasitologia , Interações Hospedeiro-Parasita , Malária/transmissão , Plasmodium/fisiologia , Animais , Aptidão Genética , Modelos Biológicos , Plasmodium/genética , Dinâmica Populacional , Reprodução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA