Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Am Nat ; 204(2): 133-146, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39008835

RESUMO

AbstractInfectious disease dynamics operate across biological scales: pathogens replicate within hosts but transmit among populations. Functional changes in the pathogen-host interaction thus generate cascading effects across organizational scales. We investigated within-host dynamics and among-host transmission of three strains (SAT-1, -2, -3) of foot-and-mouth disease viruses (FMDVs) in their wildlife host, African buffalo. We combined data on viral dynamics and host immune responses with mathematical models to ask the following questions: How do viral and immune dynamics vary among strains? Which viral and immune parameters determine viral fitness within hosts? And how do within-host dynamics relate to virus transmission? Our data reveal contrasting within-host dynamics among viral strains, with SAT-2 eliciting more rapid and effective immune responses than SAT-1 and SAT-3. Within-host viral fitness was overwhelmingly determined by variation among hosts in immune response activation rates but not by variation among individual hosts in viral growth rate. Our analyses investigating across-scale linkages indicate that viral replication rate in the host correlates with transmission rates among buffalo and that adaptive immune activation rate determines the infectious period. These parameters define the virus's relative basic reproductive number (ℛ0), suggesting that viral invasion potential may be predictable from within-host dynamics.


Assuntos
Búfalos , Vírus da Febre Aftosa , Febre Aftosa , Animais , Búfalos/virologia , Vírus da Febre Aftosa/imunologia , Vírus da Febre Aftosa/crescimento & desenvolvimento , Febre Aftosa/transmissão , Febre Aftosa/virologia , Febre Aftosa/imunologia , Interações Hospedeiro-Patógeno/imunologia , Replicação Viral , Modelos Biológicos
2.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33431676

RESUMO

Pathogen interactions arising during coinfection can exacerbate disease severity, for example when the immune response mounted against one pathogen negatively affects defense of another. It is also possible that host immune responses to a pathogen, shaped by historical evolutionary interactions between host and pathogen, may modify host immune defenses in ways that have repercussions for other pathogens. In this case, negative interactions between two pathogens could emerge even in the absence of concurrent infection. Parasitic worms and tuberculosis (TB) are involved in one of the most geographically extensive of pathogen interactions, and during coinfection worms can exacerbate TB disease outcomes. Here, we show that in a wild mammal natural resistance to worms affects bovine tuberculosis (BTB) severity independently of active worm infection. We found that worm-resistant individuals were more likely to die of BTB than were nonresistant individuals, and their disease progressed more quickly. Anthelmintic treatment moderated, but did not eliminate, the resistance effect, and the effects of resistance and treatment were opposite and additive, with untreated, resistant individuals experiencing the highest mortality. Furthermore, resistance and anthelmintic treatment had nonoverlapping effects on BTB pathology. The effects of resistance manifested in the lungs (the primary site of BTB infection), while the effects of treatment manifested almost entirely in the lymph nodes (the site of disseminated disease), suggesting that resistance and active worm infection affect BTB progression via distinct mechanisms. Our findings reveal that interactions between pathogens can occur as a consequence of processes arising on very different timescales.


Assuntos
Búfalos/imunologia , Resistência à Doença , Hemoncose/microbiologia , Pulmão/imunologia , Linfonodos/imunologia , Tricostrongilose/microbiologia , Tuberculose Bovina/microbiologia , Animais , Antinematódeos/farmacologia , Búfalos/microbiologia , Búfalos/parasitologia , Bovinos , Coinfecção , Progressão da Doença , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Eosinófilos/microbiologia , Eosinófilos/parasitologia , Fezes/parasitologia , Feminino , Fenbendazol/farmacologia , Hemoncose/tratamento farmacológico , Hemoncose/mortalidade , Hemoncose/parasitologia , Haemonchus/efeitos dos fármacos , Haemonchus/genética , Haemonchus/patogenicidade , Imunoglobulina A/sangue , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/parasitologia , Linfonodos/efeitos dos fármacos , Linfonodos/microbiologia , Linfonodos/parasitologia , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Mastócitos/microbiologia , Mastócitos/parasitologia , Mycobacterium bovis/crescimento & desenvolvimento , Mycobacterium bovis/patogenicidade , Índice de Gravidade de Doença , Análise de Sobrevida , Tricostrongilose/tratamento farmacológico , Tricostrongilose/mortalidade , Tricostrongilose/parasitologia , Trichostrongylus/efeitos dos fármacos , Trichostrongylus/genética , Trichostrongylus/patogenicidade , Tuberculose Bovina/tratamento farmacológico , Tuberculose Bovina/mortalidade , Tuberculose Bovina/parasitologia
3.
Mol Ecol ; 32(13): 3733-3746, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37009964

RESUMO

Individual animals in natural populations tend to host diverse parasite species concurrently over their lifetimes. In free-living ecological communities, organismal life histories shape interactions with their environment, which ultimately forms the basis of ecological succession. However, the structure and dynamics of mammalian parasite communities have not been contextualized in terms of primary ecological succession, in part because few datasets track occupancy and abundance of multiple parasites in wild hosts starting at birth. Here, we studied community dynamics of 12 subtypes of protozoan microparasites (Theileria spp.) in a herd of African buffalo. We show that Theileria communities followed predictable patterns of succession underpinned by four different parasite life history strategies. However, in contrast to many free-living communities, network complexity decreased with host age. Examining parasite communities through the lens of succession may better inform the effect of complex within host eco-evolutionary dynamics on infection outcomes, including parasite co-existence through the lifetime of the host.


Assuntos
Interações Hospedeiro-Parasita , Características de História de Vida , Parasitos , Animais , Evolução Biológica , Biota , Mamíferos
4.
J Anim Ecol ; 92(3): 665-676, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36567629

RESUMO

Uncovering drivers of community assembly is a key aspect of learning how biological communities function. Drivers of community similarity can be especially useful in this task as they affect assemblage-level changes that lead to differences in species diversity between habitats. Concepts of ß-diversity originally developed for use in free-living communities have been widely applied to parasite communities to gain insight into how infection risk changes with local conditions by comparing parasite communities across abiotic and biotic gradients. Factors shaping ß-diversity in communities of immature parasites, such as larvae, are largely unknown. This is a key knowledge gap as larvae are frequently the infective life-stage and understanding variation in these larval communities is thus key for disease prevention. Our goal was to uncover links between ß-diversity of parasite communities at different life stages; therefore, we used gastrointestinal nematodes infecting African buffalo in Kruger National Park, South Africa, to investigate within-host and extra-host drivers of adult and larval parasite community similarity. We employed a cross-sectional approach using PERMANOVA that examined each worm community at a single time point to assess independent drivers of ß-diversity in larvae and adults as well as a longitudinal approach with path analysis where adult and larval communities from the same host were compared to better link drivers of ß-diversity between these two life stages. Using the cross-sectional approach, we generally found that intrinsic, within-host traits had significant effects on ß-diversity of adult nematode communities, while extrinsic, extra-host variables had significant effects on ß-diversity of larval nematode communities. However, the longitudinal approach provided evidence that intrinsic, within-host factors affected the larval community indirectly via the adult community. Our results provide key data for the comparison of community-level processes where adult and immature stages inhabit vastly different habitats (i.e. within-host vs. abiotic environment). In the context of parasitism, this helps elucidate host infection risk via larval stages and the drivers that shape persistence of adult parasite assemblages, both of which are useful for predicting and preventing infectious disease.


Assuntos
Nematoides , Parasitos , Animais , Ecossistema , Biota , Trato Gastrointestinal , Interações Hospedeiro-Parasita
5.
J Anim Ecol ; 92(7): 1456-1469, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36637333

RESUMO

Habitat fragmentation is an important driver of biodiversity loss and can be remediated through management actions aimed at maintenance of natural connectivity in metapopulations. Connectivity may protect populations from infectious diseases by preserving immunogenetic diversity and disease resistance. However, connectivity could exacerbate the risk of infectious disease spread across vulnerable populations. We tracked the spread of a novel strain of Mycoplasma ovipneumoniae in a metapopulation of desert bighorn sheep Ovis canadensis nelsoni in the Mojave Desert to investigate how variation in connectivity among populations influenced disease outcomes. M. ovipneumoniae was detected throughout the metapopulation, indicating that the relative isolation of many of these populations did not protect them from pathogen invasion. However, we show that connectivity among bighorn sheep populations was correlated with higher immunogenetic diversity, a protective immune response and lower disease prevalence. Variation in protective immunity predicted infection risk in individual bighorn sheep and was associated with heterozygosity at genetic loci linked to adaptive and innate immune signalling. Together, these findings may indicate that population connectivity maintains immunogenetic diversity in bighorn sheep populations in this system and has direct effects on immune responses in individual bighorn sheep and their susceptibility to infection by a deadly pathogen. Our study suggests that the genetic benefits of population connectivity could outweigh the risk of infectious disease spread and supports conservation management that maintains natural connectivity in metapopulations.


Assuntos
Doenças Transmissíveis , Pneumonia , Doenças dos Ovinos , Carneiro da Montanha , Animais , Ovinos , Pneumonia/veterinária , Variação Genética , Imunidade , Doenças dos Ovinos/epidemiologia
6.
Vet Res ; 53(1): 63, 2022 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-35927724

RESUMO

Foot-and-mouth disease (FMD) is one of the most important livestock diseases restricting international trade. While African buffalo (Syncerus caffer) act as the main wildlife reservoir, viral and immune response dynamics during FMD virus acute infection have not been described before in this species. We used experimental needle inoculation and contact infections with three Southern African Territories serotypes to assess clinical, virological and immunological dynamics for thirty days post infection. Clinical FMD in the needle inoculated buffalo was mild and characterised by pyrexia. Despite the absence of generalised vesicles, all contact animals were readily infected with their respective serotypes within the first two to nine days after being mixed with needle challenged buffalo. Irrespective of the route of infection or serotype, there were positive associations between the viral loads in blood and the induction of host innate pro-inflammatory cytokines and acute phase proteins. Viral loads in blood and tonsil swabs were tightly correlated during the acute phase of the infection, however, viraemia significantly declined after a peak at four days post-infection (dpi), which correlated with the presence of detectable neutralising antibodies. In contrast, infectious virus was isolated in the tonsil swabs until the last sampling point (30 dpi) in most animals. The pattern of virus detection in serum and tonsil swabs was similar for all three serotypes in the direct challenged and contact challenged animals. We have demonstrated for the first time that African buffalo are indeed systemically affected by FMD virus and clinical FMD in buffalo is characterized by a transient pyrexia. Despite the lack of FMD lesions, infection of African buffalo was characterised by high viral loads in blood and oropharynx, rapid and strong host innate and adaptive immune responses and high transmissibility.


Assuntos
Vírus da Febre Aftosa , Febre Aftosa , Animais , Anticorpos Antivirais , Búfalos , Comércio , Febre/veterinária , Vírus da Febre Aftosa/fisiologia , Imunidade , Internacionalidade
7.
Proc Natl Acad Sci U S A ; 116(29): 14645-14650, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31262813

RESUMO

Novel parasites can have wide-ranging impacts, not only on host populations, but also on the resident parasite community. Historically, impacts of novel parasites have been assessed by examining pairwise interactions between parasite species. However, parasite communities are complex networks of interacting species. Here we used multivariate taxonomic and trait-based approaches to determine how parasite community composition changed when African buffalo (Syncerus caffer) acquired an emerging disease, bovine tuberculosis (BTB). Both taxonomic and functional parasite richness increased significantly in animals that acquired BTB than in those that did not. Thus, the presence of BTB seems to catalyze extraordinary shifts in community composition. There were no differences in overall parasite taxonomic composition between infected and uninfected individuals, however. The trait-based analysis revealed an increase in direct-transmitted, quickly replicating parasites following BTB infection. This study demonstrates that trait-based approaches provide insight into parasite community dynamics in the context of emerging infections.


Assuntos
Búfalos/parasitologia , Doenças Transmissíveis Emergentes/veterinária , Interações Hospedeiro-Parasita/genética , Parasitos/genética , Tuberculose Bovina/imunologia , Animais , Búfalos/imunologia , Búfalos/microbiologia , Bovinos , Doenças Transmissíveis Emergentes/imunologia , Doenças Transmissíveis Emergentes/microbiologia , Feminino , Interações Hospedeiro-Parasita/imunologia , Estudos Longitudinais , Mycobacterium bovis/imunologia , Parasitos/imunologia , Parasitos/isolamento & purificação , África do Sul , Tuberculose Bovina/microbiologia
8.
J Anim Ecol ; 90(3): 602-614, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33232513

RESUMO

The dynamics of directly transmitted pathogens in natural populations are likely to result from the combined effects of host traits, pathogen biology, and interactions among pathogens within a host. Discovering how these factors work in concert to shape variation in pathogen dynamics in natural host-multi-pathogen systems is fundamental to understanding population health. Here, we describe temporal variation in incidence and then elucidate the effect of hosts trait, season and pathogen co-occurrence on host infection risk using one of the most comprehensive studies of co-infection in a wild population: a suite of seven directly transmitted viral and bacterial respiratory infections from a 4-year study of 200 free-ranging African buffalo Syncerus caffer. Incidence of upper respiratory infections was common throughout the study-five out of the seven pathogens appeared to be consistently circulating throughout our study population. One pathogen exhibited clear outbreak dynamics in our final study year and another was rarely detected. Co-infection was also common in this system: The strongest indicator of pathogen occurrence for respiratory viruses was in fact the presence of other viral respiratory infections. Host traits had minimal effects on odds of pathogen occurrence but did modify pathogen-pathogen associations. In contrast, only season predicted bacterial pathogen occurrence. Though a combination of environmental, behavioural, and physiological factors work together to shape disease dynamics, we found pathogen associations best determined infection risk. Our study demonstrates that, in the absence of very fine-scale data, the intricate changes among these factors are best represented by co-infection.


Assuntos
Coinfecção , Infecções Respiratórias , Viroses , Animais , Búfalos , Coinfecção/epidemiologia , Coinfecção/veterinária , Suscetibilidade a Doenças , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/veterinária , Viroses/epidemiologia , Viroses/veterinária
9.
Proc Natl Acad Sci U S A ; 115(29): 7545-7550, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-29967175

RESUMO

Coinfecting parasites and pathogens remain a leading challenge for global public health due to their consequences for individual-level infection risk and disease progression. However, a clear understanding of the population-level consequences of coinfection is lacking. Here, we constructed a model that includes three individual-level effects of coinfection: mortality, fecundity, and transmission. We used the model to investigate how these individual-level consequences of coinfection scale up to produce population-level infection patterns. To parameterize this model, we conducted a 4-y cohort study in African buffalo to estimate the individual-level effects of coinfection with two bacterial pathogens, bovine tuberculosis (bTB) and brucellosis, across a range of demographic and environmental contexts. At the individual level, our empirical results identified bTB as a risk factor for acquiring brucellosis, but we found no association between brucellosis and the risk of acquiring bTB. Both infections were associated with reductions in survival and neither infection was associated with reductions in fecundity. The model reproduced coinfection patterns in the data and predicted opposite impacts of coinfection at individual and population scales: Whereas bTB facilitated brucellosis infection at the individual level, our model predicted the presence of brucellosis to have a strong negative impact on bTB at the population level. In modeled populations where brucellosis was present, the endemic prevalence and basic reproduction number ([Formula: see text]) of bTB were lower than in populations without brucellosis. Therefore, these results provide a data-driven example of competition between coinfecting pathogens that occurs when one pathogen facilitates secondary infections at the individual level.


Assuntos
Brucelose , Búfalos/microbiologia , Coinfecção , Modelos Biológicos , Tuberculose Bovina , Animais , Brucelose/epidemiologia , Brucelose/microbiologia , Brucelose/transmissão , Brucelose/veterinária , Bovinos , Coinfecção/epidemiologia , Coinfecção/microbiologia , Coinfecção/transmissão , Coinfecção/veterinária , Feminino , Tuberculose Bovina/epidemiologia , Tuberculose Bovina/microbiologia , Tuberculose Bovina/transmissão
10.
Emerg Infect Dis ; 26(7): 1521-1525, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32568048

RESUMO

We screened nonequine animals with unexplained neurologic signs or death in South Africa during 2010-2018 for Shuni virus (SHUV). SHUV was detected in 3.3% of wildlife, 1.1% of domestic, and 2.0% of avian species. Seropositivity was also demonstrated in wildlife. These results suggest a range of possible SHUV hosts in Africa.


Assuntos
Animais Selvagens , Infecções por Bunyaviridae , Animais , Animais Domésticos , Orthobunyavirus , África do Sul/epidemiologia
11.
Biol Lett ; 16(2): 20190811, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32070234

RESUMO

Newborn mammals have an immature immune system that cannot sufficiently protect them against infectious diseases. However, variation in the effectiveness of maternal immunity against different parasites may couple with temporal trends in parasite exposure to influence disparities in the timing of infection risk. Determining the relationship between age and infection risk is critical in identifying the portion of a host population that contributes to parasite dynamics, as well as the parasites that regulate host recruitment. However, there are no data directly identifying timing of first infection among parasites in wildlife. Here, we took advantage of a longitudinal dataset, tracking infection status by viruses, bacteria, protists and gastro-intestinal worms in a herd of African buffalo (Syncerus caffer) to ask: how does age of first infection differ among parasite taxa? We found distinct differences in the age of first infection among parasites that aligned with the mode of transmission and parasite taxonomy. Specifically, we found that tick-borne and environmentally transmitted protists were acquired earlier than directly transmitted bacteria and viruses. These results emphasize the importance of understanding infection risk in juveniles, especially in host species where juveniles are purported to sustain parasite persistence and/or where mortality rates of juveniles influence population dynamics.


Assuntos
Parasitos , Carrapatos , Animais , Animais Selvagens , Especificidade de Hospedeiro , Interações Hospedeiro-Parasita , Humanos , Recém-Nascido , Mamíferos
12.
Proc Biol Sci ; 286(1907): 20190914, 2019 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-31311473

RESUMO

Integrating biological processes across scales remains a central challenge in disease ecology. Genetic variation drives differences in host immune responses, which, along with environmental factors, generates temporal and spatial infection patterns in natural populations that epidemiologists seek to predict and control. However, genetics and immunology are typically studied in model systems, whereas population-level patterns of infection status and susceptibility are uniquely observable in nature. Despite obvious causal connections, organizational scales from genes to host outcomes to population patterns are rarely linked explicitly. Here we identify two loci near genes involved in macrophage (phagocyte) activation and pathogen degradation that additively increase risk of bovine tuberculosis infection by up to ninefold in wild African buffalo. Furthermore, we observe genotype-specific variation in IL-12 production indicative of variation in macrophage activation. Here, we provide measurable differences in infection resistance at multiple scales by characterizing the genetic and inflammatory variation driving patterns of infection in a wild mammal.


Assuntos
Búfalos , Genótipo , Mycobacterium bovis/fisiologia , Tuberculose/veterinária , Alelos , Animais , Feminino , África do Sul , Tuberculose/genética , Tuberculose/microbiologia
13.
Proc Biol Sci ; 286(1912): 20191401, 2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31575363

RESUMO

Immunity is one of the most variable phenotypic traits in animals; however, some individuals may show less fluctuation in immune traits, resulting in stable patterns of immune variation over time. It is currently unknown whether immune variation has consequences for infectious disease risk. In this study, we identified moderately stable immune traits in wild African buffalo and asked whether the stability of these traits affected bovine tuberculosis (TB) infection risk. We found that adaptive immune traits such as the level of interferon-γ (IFN-γ) released after white blood cell stimulation, the number of circulating lymphocytes and the level of antibodies against bovine adenovirus-3 were moderately repeatable (i.e. stable) over time, whereas parameters related to innate immunity either had low repeatability (circulating eosinophil numbers) or were not repeatable (e.g. neutrophil numbers, plasma bacteria killing capacity). Intriguingly, individuals with more repeatable IFN-γ and lymphocyte levels were at a significantly higher risk of acquiring TB infection. In stark contrast, average IFN-γ and lymphocyte levels were poor predictors of TB risk, indicating that immune variability rather than absolute response level better captured variation in disease susceptibility. This work highlights the important and under-appreciated role of immune variability as a predictor of infection risk.


Assuntos
Tuberculose/veterinária , Animais , Búfalos/microbiologia , Suscetibilidade a Doenças , Imunidade Inata , Interferon gama/metabolismo , Tuberculose/epidemiologia , Tuberculose/imunologia
14.
Parasitol Res ; 116(5): 1597-1602, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28357577

RESUMO

Accurate measures of nematode fecundity can provide important information for investigating parasite life history evolution, transmission potential, and effects on host health. Understanding differences among fecundity assessment protocols and standardizing methods, where possible, will enable comparisons across different studies and host and parasite species and systems. Using the trichostrongyle nematode Cooperia fuelleborni isolated from wild African buffalo (Syncerus caffer), we compared egg recovery and enumeration between two methods for measuring the fecundity of female worms. The first method, in utero egg count, involves visual enumeration of the eggs via microscopic inspection of the uterine system. The second method, ex utero egg count, involves dissolving the same specimens from above in a sodium chloride solution to release the eggs from the female's uterus, then enumeration under an inverted microscope. On average, the ex utero method resulted in 34% more eggs than the in utero method. However, results indicate that the two methods used to quantify parasitic nematode fecundity are highly correlated. Thus, while both methods are viable options for estimating relative nematode fecundity, we recommend caution in undertaking comparative studies that utilize egg count data collected using different methods.


Assuntos
Búfalos/parasitologia , Fezes/parasitologia , Nematoides/isolamento & purificação , Infecções por Nematoides/veterinária , Contagem de Ovos de Parasitas/métodos , Animais , Feminino , Fertilidade , Infecções por Nematoides/epidemiologia , Infecções por Nematoides/parasitologia , Óvulo/citologia , África do Sul/epidemiologia
15.
J Zoo Wildl Med ; 48(2): 298-311, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28749296

RESUMO

The International Species Information System has set forth an extensive database of reference intervals for zoologic species, allowing veterinarians and game park officials to distinguish normal health parameters from underlying disease processes in captive wildlife. However, several recent studies comparing reference values from captive and free-ranging animals have found significant variation between populations, necessitating the development of separate reference intervals in free-ranging wildlife to aid in the interpretation of health data. Thus, this study characterizes reference intervals for six biochemical analytes, eleven hematologic or immune parameters, and three hormones using samples from 219 free-ranging African lions ( Panthera leo ) captured in Kruger National Park, South Africa. Using the original sample population, exclusion criteria based on physical examination were applied to yield a final reference population of 52 clinically normal lions. Reference intervals were then generated via 90% confidence intervals on log-transformed data using parametric bootstrapping techniques. In addition to the generation of reference intervals, linear mixed-effect models and generalized linear mixed-effect models were used to model associations of each focal parameter with the following independent variables: age, sex, and body condition score. Age and sex were statistically significant drivers for changes in hepatic enzymes, renal values, hematologic parameters, and leptin, a hormone related to body fat stores. Body condition was positively correlated with changes in monocyte counts. Given the large variation in reference values taken from captive versus free-ranging lions, it is our hope that this study will serve as a baseline for future clinical evaluations and biomedical research targeting free-ranging African lions.


Assuntos
Animais Selvagens , Animais de Zoológico , Leões/sangue , Alanina Transaminase/sangue , Fosfatase Alcalina/sangue , Animais , Glicemia , Sedimentação Sanguínea , Nitrogênio da Ureia Sanguínea , Creatinina/sangue , Feminino , Grelina/sangue , Leptina/sangue , Contagem de Leucócitos/veterinária , Leucócitos/fisiologia , Masculino , Neutrófilos , Valores de Referência , África do Sul , Testosterona/sangue
16.
J Anim Ecol ; 85(5): 1222-33, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27174037

RESUMO

Community assembly is a fundamental process that has long been a central focus in ecology. Extending community assembly theory to communities of co-infecting parasites, we used a gastrointestinal nematode removal experiment in free-ranging African buffalo to examine the community assembly patterns and processes. We first asked whether reassembled communities differ from undisturbed communities by comparing anthelmintic-treated and control hosts. Next, we examined the temporal dynamics of assembly using a cross-section of communities that reassembled for different periods of time since last experimental removal. Next, we tested for evidence of assembly processes that might drive such reassembly patterns: environmental filtering based on host traits (i.e. habitat patches), interspecific interactions, priority effects and chance dispersal from the environmental pool of infective stages (i.e. the regional species pool). On average, reassembled parasite communities had lower abundance, but were more diverse and even, and these patterns varied tightly with reassembly time. Over time, the communities within treated hosts progressively resembled controls as diversity and evenness decreased, while total abundance increased. Notably, experimental removal allowed us to attribute observed differences in abundance, diversity and evenness to the process of community assembly. During early reassembly, parasite accumulation was biased towards a subordinate species and, by excluding stochastic assembly processes (i.e. chance dispersal and priority effects), we were able to determine that early assembly is deterministic. Later in the reassembly process, we established that host traits, as well as stochastic dispersal from the environmental pool of infective stages, can affect the community composition. Overall, our results suggest that there is a high degree of resiliency and environmental dependence to the worm communities of buffalo. More generally, our data show that both deterministic and stochastic processes may play a role in the assembly of parasite communities of wild hosts, but their relative importance may vary temporally. Consequently, the best strategy for managing reassembling parasite communities may also need to shift over time.


Assuntos
Biota , Búfalos/parasitologia , Interações Hospedeiro-Parasita , Nematoides/fisiologia , Animais , Antinematódeos/administração & dosagem , Fenbendazol/administração & dosagem , Gastroenteropatias/tratamento farmacológico , Gastroenteropatias/parasitologia , Gastroenteropatias/veterinária , Infecções por Nematoides/tratamento farmacológico , Infecções por Nematoides/parasitologia , Infecções por Nematoides/veterinária , Dinâmica Populacional , Distribuição Aleatória , África do Sul , Processos Estocásticos
17.
J Anim Ecol ; 85(4): 1025-34, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27084785

RESUMO

Experimental studies in laboratory settings have demonstrated a critical role of parasite interactions in shaping parasite communities. The sum of these interactions can produce diverse effects on individual hosts as well as influence disease emergence and persistence at the population level. A predictive framework for the effects of parasite interactions in the wild remains elusive, largely because of limited longitudinal or experimental data on parasite communities of free-ranging hosts. This 4-year study followed a community of haemoparasites in free-ranging African buffalo (Syncerus caffer). We detected infection by 11 haemoparasite species using PCR-based diagnostic techniques, and analyzed drivers of infection patterns using generalized linear mixed models to understand the role of host characteristics and season on infection likelihood. We tested for (i) effects of co-infection by other haemoparasites (within guild) and (ii) effects of parasites infecting different tissue types (across guild). We found that within guild co-infections were the strongest predictors of haemoparasite infections in the buffalo; but that seasonal and host characteristics also had important effects. In contrast, the evidence for across-guild effects of parasites utilizing different tissue on haemoparasite infection was weak. These results provide a nuanced view of the role of co-infections in determining haemoparasite infection patterns in free living mammalian hosts. Our findings suggest a role for interactions among parasites infecting a single tissue type in determining infection patterns.


Assuntos
Búfalos , Coinfecção/veterinária , Theileriose/imunologia , Animais , Sangue/microbiologia , Sangue/parasitologia , Coinfecção/imunologia , Coinfecção/microbiologia , Coinfecção/parasitologia , Feminino , Interações Hospedeiro-Parasita , Estudos Longitudinais , África do Sul , Theileria/fisiologia , Theileriose/parasitologia
18.
Nature ; 468(7324): 647-52, 2010 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-21124449

RESUMO

Current unprecedented declines in biodiversity reduce the ability of ecological communities to provide many fundamental ecosystem services. Here we evaluate evidence that reduced biodiversity affects the transmission of infectious diseases of humans, other animals and plants. In principle, loss of biodiversity could either increase or decrease disease transmission. However, mounting evidence indicates that biodiversity loss frequently increases disease transmission. In contrast, areas of naturally high biodiversity may serve as a source pool for new pathogens. Overall, despite many remaining questions, current evidence indicates that preserving intact ecosystems and their endemic biodiversity should generally reduce the prevalence of infectious diseases.


Assuntos
Biodiversidade , Doenças Transmissíveis/transmissão , Animais , Doenças Transmissíveis/epidemiologia , Doenças Transmissíveis/microbiologia , Doenças Transmissíveis/virologia , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/microbiologia , Doenças Transmissíveis Emergentes/transmissão , Doenças Transmissíveis Emergentes/virologia , Orthohantavírus/fisiologia , Humanos , Doença de Lyme/microbiologia , Doença de Lyme/transmissão , Especificidade da Espécie , Zoonoses/epidemiologia , Zoonoses/transmissão
19.
J Anim Ecol ; 84(3): 637-646, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25631200

RESUMO

Animals' social and movement behaviours can impact the transmission dynamics of infectious diseases, especially for pathogens transmitted through close contact between hosts or through contact with infectious stages in the environment. Estimating pathogen transmission rates and R0 from natural systems can be challenging. Because host behavioural traits that underlie the transmission process vary predictably with body size, one of the best-studied traits among animals, body size might therefore also predict variation in parasite transmission dynamics. Here, we examine how two host behaviours, social group living and the intensity of habitat use, scale allometrically using comparative data from wild primate, carnivore and ungulate species. We use these empirical relationships to parameterize classical compartment models for infectious micro- and macroparasitic diseases, and examine how the risk of pathogen invasion changes as a function of host behaviour and body size. We then test model predictions using comparative data on parasite prevalence and richness from wild mammals. We report a general pattern suggesting that smaller-bodied mammal species utilizing home ranges more intensively experience greater risk for invasion by environmentally transmitted macroparasites. Conversely, larger-bodied hosts exhibiting a high degree of social group living could be more readily invaded by directly transmitted microparasites. These trends were supported through comparison of micro- and macroparasite species richness across a large number of carnivore, primate and ungulate species, but empirical data on carnivore macroparasite prevalence showed mixed results. Collectively, our study demonstrates that combining host behavioural traits with dynamical models of infectious disease scaled against host body size can generate testable predictions for variation in parasite risk across species; a similar approach might be useful in future work focused on predicting parasite distributions in local host communities.


Assuntos
Doenças dos Animais/transmissão , Comportamento Animal , Tamanho Corporal , Doenças Transmissíveis/veterinária , Mamíferos , Doenças dos Animais/microbiologia , Doenças dos Animais/virologia , Animais , Doenças Transmissíveis/microbiologia , Doenças Transmissíveis/transmissão , Doenças Transmissíveis/virologia , Comportamento de Retorno ao Território Vital , Interações Hospedeiro-Patógeno , Modelos Biológicos , Prevalência , Comportamento Social
20.
J Anim Ecol ; 84(4): 999-1009, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25714466

RESUMO

Chronic infections may have negative impacts on wildlife populations, yet their effects are difficult to detect in the absence of long-term population monitoring. Brucella abortus, the bacteria responsible for bovine brucellosis, causes chronic infections and abortions in wild and domestic ungulates, but its impact on population dynamics is not well understood. We report infection patterns and fitness correlates of bovine brucellosis in African buffalo based on (1) 7 years of cross-sectional disease surveys and (2) a 4-year longitudinal study in Kruger National Park (KNP), South Africa. We then used a matrix population model to translate these observed patterns into predicted population-level effects. Annual brucellosis seroprevalence ranged from 8·7% (95% CI = 1·8-15·6) to 47·6% (95% CI = 35·1-60·1) increased with age until adulthood (>6) and varied by location within KNP. Animals were on average in worse condition after testing positive for brucellosis (F = -5·074, P < 0·0001), and infection was associated with a 2·0 (95% CI = 1·1-3·7) fold increase in mortality (χ(2)  = 2·039, P = 0·036). Buffalo in low body condition were associated with lower reproductive success (F = 2·683, P = 0·034), but there was no association between brucellosis and pregnancy or being observed with a calf. For the range of body condition scores observed in the population, the model-predicted growth rate was λ = 1·11 (95% CI = 1·02-1·21) in herds without brucellosis and λ = 1·00 (95% CI = 0·85-1·16) when brucellosis seroprevalence was 30%. Our results suggest that brucellosis infection can potentially result in reduced population growth rates, but because these effects varied with demographic and environmental conditions, they may remain unseen without intensive, longitudinal monitoring.


Assuntos
Brucella abortus/patogenicidade , Brucelose/veterinária , Búfalos/microbiologia , Fertilidade , Animais , Brucelose/epidemiologia , Brucelose/microbiologia , Estudos Transversais , Feminino , Estudos Longitudinais , Masculino , Dinâmica Populacional , Gravidez , Estudos Soroepidemiológicos , África do Sul , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA