Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.995
Filtrar
1.
Infect Dis Poverty ; 13(1): 40, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38822386

RESUMO

BACKGROUND: Opisthorchiid flukes, particularly Opisthorchis viverrini, Opisthorchis felineus, Clonorchis sinensis, and Metorchis spp. are the most common fish-borne zoonotic human liver flukes (hLFs). Liver fluke infections are more prevalent in resource-deprived and underprivileged areas. We herein estimated the prevalence of the metacercariae (MC) of major hLFs in common large freshwater fishes (lFWF) marketed for human consumption from some selected areas of Bangladesh along with detection of their molluscan vectors and reservoirs. METHODS: The current status of fish-borne zoonotic hLF infections in lFWF was investigated along with their molluscan vectors and mammalian reservoir hosts in Mymensingh and Kishoreganj in Bangladesh from July 2018-June 2022 using conventional and multiple molecular techniques, such as PCR, PCR-restriction fragment length polymorphism (RFLP), sequencing, and bioinformatic analyses. The infection rate of fishes was analyzed using the Z-test and the loads of MC were compared using the chi-squared (χ2) test. RESULTS: The MC of C. sinensis, Opisthorchis spp., and Metorchis spp. were detected in 11 species of common and popular lFWF. In lFWF, the estimated prevalence was 18.7% and the mean load was 137.4 ± 149.8 MC per 100 g of fish. The prevalence was the highest (P < 0.05) in spotted snakehead fishes (Channa punctata, 63.6%). The highest rate of infection (P < 0.05) was observed with the MC of C. sinensis (11.8%). Metacercariae were almost equally (P > 0.05) distributed between the head and body of fishes. The infection rate was slightly higher in cultured (19.6%) fishes. The MC of C. sinensis, O. felineus, O. viverrini, and Metorchis orientalis in fishes were confirmed using PCR, PCR-RFLP and bioinformatics. The cercariae of opisthorchiid (Pleurolophocercus cercariae) flukes were only recovered from Bithynia spp. (3.9%, 42 out of 1089). The ova of hLFs from dogs (4.3%, 5 out of 116) and cats (6.0%, 6 out of 100), and adult flukes (M. orientalis) from ducks (41.1% 113 out of 275) were detected. CONCLUSIONS: The MC of hLFs are highly prevalent in fresh water fishes in Bangladesh. Reservoir hosts, such as street dogs, cats, and ducks carried the patent infection, and residents of Bangladesh are at risk.


Assuntos
Reservatórios de Doenças , Doenças dos Peixes , Peixes , Água Doce , Zoonoses , Animais , Bangladesh/epidemiologia , Peixes/parasitologia , Água Doce/parasitologia , Doenças dos Peixes/parasitologia , Doenças dos Peixes/epidemiologia , Humanos , Reservatórios de Doenças/parasitologia , Reservatórios de Doenças/veterinária , Zoonoses/parasitologia , Zoonoses/epidemiologia , Zoonoses/transmissão , Vetores de Doenças , Prevalência , Opisthorchis/genética , Opisthorchis/isolamento & purificação , Metacercárias/genética , Metacercárias/isolamento & purificação , Clonorchis sinensis/genética , Clonorchis sinensis/isolamento & purificação , Moluscos/parasitologia
2.
Methods Mol Biol ; 2807: 93-110, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38743223

RESUMO

Correlative light-electron microscopy (CLEM) has evolved in the last decades, especially after significant developments in sample preparation, imaging acquisition, software, spatial resolution, and equipment, including confocal, live-cell, super-resolution, and electron microscopy (scanning, transmission, focused ion beam, and cryo-electron microscopy). However, the recent evolution of different laser-related techniques, such as mass spectrometry imaging (MSI) and laser capture microdissection, could further expand spatial imaging capabilities into high-resolution OMIC approaches such as proteomic, lipidomics, small molecule, and drug discovery. Here, we will describe a protocol to integrate the detection of rare viral reservoirs with imaging mass spectrometry.


Assuntos
Infecções por HIV , Humanos , Infecções por HIV/virologia , HIV-1/fisiologia , Espectrometria de Massas/métodos , Microscopia Eletrônica/métodos , Imagem Molecular/métodos , Reservatórios de Doenças/virologia
3.
J Math Biol ; 89(1): 7, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38772937

RESUMO

Malaria is a vector-borne disease that exacts a grave toll in the Global South. The epidemiology of Plasmodium vivax, the most geographically expansive agent of human malaria, is characterised by the accrual of a reservoir of dormant parasites known as hypnozoites. Relapses, arising from hypnozoite activation events, comprise the majority of the blood-stage infection burden, with implications for the acquisition of immunity and the distribution of superinfection. Here, we construct a novel model for the transmission of P. vivax that concurrently accounts for the accrual of the hypnozoite reservoir, (blood-stage) superinfection and the acquisition of immunity. We begin by using an infinite-server queueing network model to characterise the within-host dynamics as a function of mosquito-to-human transmission intensity, extending our previous model to capture a discretised immunity level. To model transmission-blocking and antidisease immunity, we allow for geometric decay in the respective probabilities of successful human-to-mosquito transmission and symptomatic blood-stage infection as a function of this immunity level. Under a hybrid approximation-whereby probabilistic within-host distributions are cast as expected population-level proportions-we couple host and vector dynamics to recover a deterministic compartmental model in line with Ross-Macdonald theory. We then perform a steady-state analysis for this compartmental model, informed by the (analytic) distributions derived at the within-host level. To characterise transient dynamics, we derive a reduced system of integrodifferential equations, likewise informed by our within-host queueing network, allowing us to recover population-level distributions for various quantities of epidemiological interest. In capturing the interplay between hypnozoite accrual, superinfection and acquired immunity-and providing, to the best of our knowledge, the most complete population-level distributions for a range of epidemiological values-our model provides insights into important, but poorly understood, epidemiological features of P. vivax.


Assuntos
Malária Vivax , Conceitos Matemáticos , Mosquitos Vetores , Plasmodium vivax , Superinfecção , Humanos , Plasmodium vivax/imunologia , Plasmodium vivax/fisiologia , Superinfecção/imunologia , Superinfecção/transmissão , Superinfecção/parasitologia , Malária Vivax/transmissão , Malária Vivax/imunologia , Malária Vivax/epidemiologia , Malária Vivax/parasitologia , Animais , Mosquitos Vetores/parasitologia , Mosquitos Vetores/imunologia , Reservatórios de Doenças/parasitologia , Modelos Biológicos , Simulação por Computador , Anopheles/parasitologia , Anopheles/imunologia
4.
Adv Exp Med Biol ; 1451: 75-90, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38801572

RESUMO

The current multicounty outbreak of monkeypox virus (MPXV) posed an emerging and continued challenge to already strained public healthcare sector, around the globe. Since its first identification, monkeypox disease (mpox) remained enzootic in Central and West African countries where reports of human cases are sporadically described. Recent trends in mpox spread outside the Africa have highlighted increased incidence of spillover of the MPXV from animal to humans. While nature of established animal reservoirs remained undefined, several small mammals including rodents, carnivores, lagomorphs, insectivores, non-human primates, domestic/farm animals, and several species of wildlife are proposed to be carrier of the MPXV infection. There are established records of animal-to-human (zoonotic) spread of MPXV through close interaction of humans with animals by eating bushmeat, contracting bodily fluids or trading possibly infected animals. In contrast, there are reports and increasing possibilities of human-to-animal (zooanthroponotic) spread of the MPXV through petting and close interaction with pet owners and animal care workers. We describe here the rationales and molecular factors which predispose the spread of MPXV not only amongst humans but also from animals to humans. A range of continuing opportunities for the spread and evolution of MPXV are discussed to consider risks beyond the currently identified groups. With the possibility of MPXV establishing itself in animal reservoirs, continued and broad surveillance, investigation into unconventional transmissions, and exploration of spillover events are warranted.


Assuntos
Monkeypox virus , Mpox , Zoonoses , Animais , Mpox/transmissão , Mpox/epidemiologia , Mpox/virologia , Humanos , Monkeypox virus/patogenicidade , Monkeypox virus/genética , Zoonoses/transmissão , Zoonoses/virologia , Zoonoses/epidemiologia , Reservatórios de Doenças/virologia , Surtos de Doenças , Animais Selvagens/virologia
5.
Sci Rep ; 14(1): 11171, 2024 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-38750049

RESUMO

White-tailed deer (Odocoileus virginianus) have emerged as a reservoir host for SARS-CoV-2 given their susceptibility to infection and demonstrated high rates of seroprevalence and infection across the United States. As SARS-CoV-2 circulates within free-ranging white-tailed deer populations, there is the risk of transmission to other wildlife species and even back to the human population. The goal of this study was to determine the susceptibility, shedding, and immune response of North American elk (Cervus elaphus canadensis) to experimental infection with SARS-CoV-2, to determine if another wide-ranging cervid species could potentially serve as a reservoir host for the virus. Here we demonstrate that while North American elk do not develop clinical signs of disease, they do develop a neutralizing antibody response to infection, suggesting the virus is capable of replicating in this mammalian host. Additionally, we demonstrate SARS-CoV-2 RNA presence in the medial retropharyngeal lymph nodes of infected elk three weeks after experimental infection. Consistent with previous observations in humans, these data may highlight a mechanism of viral persistence for SARS-CoV-2 in elk.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19 , Cervos , RNA Viral , SARS-CoV-2 , Animais , Cervos/virologia , SARS-CoV-2/genética , SARS-CoV-2/fisiologia , COVID-19/virologia , RNA Viral/genética , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Eliminação de Partículas Virais , Reservatórios de Doenças/virologia , Feminino
6.
Infect Genet Evol ; 121: 105604, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38754756

RESUMO

Bats are known reservoirs of various emerging pathogens, and have recently been found to host a novel hantavirus, named Brno loanvirus (BRNV), from the Mammantavirinae subfamily (family Hantaviridae, order Bunyavirales). Here we report BRNV detection in bats from the urban area of Brno, Czech Republic in March 2022. Specifically, we uncovered a high prevalence of BRNV (8.8%, 5/57) among hibernating bats (Nyctalus noctula) in urban area, which poses a risk of human exposure. The positive bats included adult females (3/9 positive), a juvenile female (1/32 positive), and an adult male (1/6 positive). All 10 juvenile males were negative. We used RT-qPCR to quantify the BRNV RNA levels in various bat organs, which yielded positive results for viral RNA in organs, including the kidneys, heart, spleen, brain, liver, lung, and gut, and in body cavity fluid. Among all tested organs, the liver showed the highest levels of viral RNA in 4 out of 5 animals examined (average Ct value of 20.8 ± 7.4).


Assuntos
Quirópteros , Animais , República Tcheca/epidemiologia , Quirópteros/virologia , Feminino , Masculino , Orthohantavírus/genética , Orthohantavírus/isolamento & purificação , Orthohantavírus/classificação , RNA Viral/genética , Filogenia , Reservatórios de Doenças/virologia , Infecções por Hantavirus/veterinária , Infecções por Hantavirus/epidemiologia , Infecções por Hantavirus/virologia
7.
Prev Vet Med ; 228: 106231, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38788301

RESUMO

Wild lagomorphs play a key epidemiological role as reservoirs of Leishmania infantum, causative agent of the largest outbreak of human leishmaniosis in Europe to date. A large-scale survey study was conducted on wild rabbit (Oryctolagus cuniculus) and Iberian hare (Lepus granatensis) populations in Spanish Mediterranean ecosystems to evaluate the exposure of L. infantum and investigate potential risk factors associated with exposure to this zoonotic parasite. Between 2018 and 2021, a total of 631 wild lagomorphs (471 wild rabbits and 160 Iberian hares) were collected in Andalusia (southern Spain) and tested for antibodies against L. infantum using the indirect fluorescent antibody test (IFAT). Spleen samples from 563 of the wild lagomorphs sampled (441 wild rabbits and 122 Iberian hares) were also evaluated by real-time quantitative PCR (qPCR) for detection of Leishmania kDNA. Exposure to L. infantum (positive by IFAT and/or qPCR) was detected in 56.4 % (356/631; 95 %CI: 52.3-60.3) of the lagomorphs analyzed. Anti-Leishmania antibodies were found in 12.8 % (81/631; 95 %CI: 10.2-15.5) of the animals, and L. infantum kDNA was detected in 59.0 % (332/563; 95 %CI: 54.9-63.0) of the spleen samples tested. Phylogenetic analysis revealed high homology (99.9-100 %) between L. infantum sequences obtained and strains previously isolated from humans in Spain. While apparent seroprevalence was significantly higher in Iberian hares (19.4 %; 95 %CI: 13.3-25.5) compared to wild rabbits (10.6 %; 95 %CI: 7.9-13.4), no significant differences in prevalence were found between wild rabbits (61.0 %; 95 %CI: 56.5-65.6) and Iberian hares (51.6 %; 95 %CI: 42.8-60.5). At least one positive animal was found on 64.8 % (70/108) of the hunting grounds sampled, and a high-risk spatial cluster (P < 0.001) was also identified in central Andalusia. The multivariable analysis identified bioclimatic level (meso-Mediterranean climate) and the presence of goats on hunting grounds as risk factors potentially associated with L. infantum exposure in wild lagomorphs. This study shows high, widespread exposure, but heterogeneous distribution of L. infantum in wild lagomorph populations in Mediterranean ecosystems in southern Spain. The results point to the need to promote integrated surveillance programs for the detection of Leishmania spp. in wild lagomorphs in order to establish effective control measures against human leishmaniosis under a One Health approach.


Assuntos
Leishmania infantum , Leishmaniose Visceral , Animais , Leishmania infantum/isolamento & purificação , Espanha/epidemiologia , Leishmaniose Visceral/veterinária , Leishmaniose Visceral/epidemiologia , Leishmaniose Visceral/parasitologia , Animais Selvagens/parasitologia , Lebres/parasitologia , Coelhos , Prevalência , Monitoramento Epidemiológico/veterinária , Ecossistema , Feminino , Masculino , Reservatórios de Doenças/veterinária , Reservatórios de Doenças/parasitologia , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/análise , Estudos Soroepidemiológicos , Lagomorpha/parasitologia
8.
Ecohealth ; 21(1): 71-82, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38727761

RESUMO

Mycobacterium bovis, a member of the Mycobacterium tuberculosis complex (MTC) and non-tuberculous Mycobacteria (NTM), may infect wild and domestic mammals, including humans. Although cattle are the main hosts and spreaders of M. bovis, many wildlife hosts play an important role worldwide. In Argentina, wild boar and domestic pigs are considered important links in mammalian tuberculosis (mTB) transmission. The aim of this work was to investigate the presence of M. bovis in wild pigs from different regions of Argentina, to characterize isolates of M. bovis obtained, and to compare those with other previously found in vertebrate hosts. A total of 311 samples from wild pigs were obtained, and bacteriological culture, molecular identification and genotyping were performed, obtaining 63 isolates (34 MTC and 29 NTM). Twelve M. bovis spoligotypes were detected. Our findings suggest that wild pigs have a prominent role as reservoirs of mTB in Argentina, based on an estimated prevalence of 11.2 ± 1.8% (95% CI 8.0-14.8) for MTC and the frequency distribution of spoligotypes shared by cattle (75%), domestic pigs (58%) and wildlife (50%). Argentina has a typical scenario where cattle and pigs are farm-raised extensively, sharing the environment with wildlife, creating conditions for effective transmission of mTB in the wildlife-livestock-human interface.


Assuntos
Animais Selvagens , Mycobacterium bovis , Doenças dos Suínos , Tuberculose , Animais , Argentina/epidemiologia , Animais Selvagens/microbiologia , Tuberculose/epidemiologia , Tuberculose/veterinária , Tuberculose/microbiologia , Mycobacterium bovis/isolamento & purificação , Mycobacterium bovis/genética , Suínos , Doenças dos Suínos/microbiologia , Doenças dos Suínos/epidemiologia , Sus scrofa/microbiologia , Reservatórios de Doenças/microbiologia , Reservatórios de Doenças/veterinária , Prevalência , Genótipo
10.
Emerg Microbes Infect ; 13(1): 2341141, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38597241

RESUMO

The Natal multimammate mouse (Mastomys natalensis) is the host of Lassa mammarenavirus, causing Lassa haemorrhagic fever in West Africa. As there is currently no operational vaccine and therapeutic drugs are limited, we explored rodent control as an alternative to prevent Lassa virus spillover in Upper Guinea, where the disease is highly endemic in rural areas. In a seven-year experiment, we distributed rodenticides for 10-30 days once a year and, in the last year, added intensive snap trapping for three months in all the houses of one village. We also captured rodents both before and after the intervention period to assess their effectiveness by examining alterations in trapping success and infection rates (Lassa virus RNA and IgG antibodies). We found that both interventions reduced the rodent population by 74-92% but swiftly rebounded to pre-treatment levels, even already six months after the last snap-trapping control. Furthermore, while we observed that chemical control modestly decreased Lassa virus infection rates annually (a reduction of 5% in seroprevalence per year), the intensive trapping unexpectedly led to a significantly higher infection rate (from a seroprevalence of 28% before to 67% after snap trapping control). After seven years, we conclude that annual chemical control, alone or with intensive trapping, is ineffective and sometimes counterproductive in preventing Lassa virus spillover in rural villages. These unexpected findings may result from density-dependent breeding compensation following culling and the survival of a small percentage of chronically infected rodents that may spread the virus to a new susceptible generation of mice.


Assuntos
Febre Lassa , Vírus Lassa , Camundongos , Animais , Vírus Lassa/genética , Guiné/epidemiologia , Controle de Roedores , Estudos Soroepidemiológicos , Reservatórios de Doenças , Febre Lassa/epidemiologia , Febre Lassa/prevenção & controle , Murinae , África Ocidental/epidemiologia
11.
Viruses ; 16(4)2024 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-38675988

RESUMO

Sosuga virus (SOSV), a rare human pathogenic paramyxovirus, was first discovered in 2012 when a person became ill after working in South Sudan and Uganda. During an ecological investigation, several species of bats were sampled and tested for SOSV RNA and only one species, the Egyptian rousette bat (ERBs; Rousettus aegyptiacus), tested positive. Since that time, multiple other species have been sampled and ERBs in Uganda have continued to be the only species of bat positive for SOSV infection. Subsequent studies of ERBs with SOSV demonstrated that ERBs are a competent host for SOSV and shed this infectious virus while exhibiting only minor infection-associated pathology. Following the 2014 Ebola outbreak in West Africa, surveillance efforts focused on discovering reservoirs for zoonotic pathogens resulted in the capture and testing of many bat species. Here, SOSV RNA was detected by qRT-PCR only in ERBs captured in the Moyamba District of Sierra Leone in the central region of the country. These findings represent a substantial range extension from East Africa to West Africa for SOSV, suggesting that this paramyxovirus may occur in ERB populations throughout its sub-Saharan African range.


Assuntos
Quirópteros , Animais , Quirópteros/virologia , Serra Leoa/epidemiologia , Infecções por Paramyxoviridae/veterinária , Infecções por Paramyxoviridae/virologia , Infecções por Paramyxoviridae/epidemiologia , RNA Viral/genética , Filogenia , Reservatórios de Doenças/virologia , Humanos
12.
Nat Commun ; 15(1): 3589, 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38678025

RESUMO

The black rat (Rattus rattus) is a globally invasive species that has been widely introduced across Africa. Within its invasive range in West Africa, R. rattus may compete with the native rodent Mastomys natalensis, the primary reservoir host of Lassa virus, a zoonotic pathogen that kills thousands annually. Here, we use rodent trapping data from Sierra Leone and Guinea to show that R. rattus presence reduces M. natalensis density within the human dwellings where Lassa virus exposure is most likely to occur. Further, we integrate infection data from M. natalensis to demonstrate that Lassa virus zoonotic spillover risk is lower at sites with R. rattus. While non-native species can have numerous negative effects on ecosystems, our results suggest that R. rattus invasion has the indirect benefit of decreasing zoonotic spillover of an endemic pathogen, with important implications for invasive species control across West Africa.


Assuntos
Reservatórios de Doenças , Espécies Introduzidas , Febre Lassa , Vírus Lassa , Murinae , Zoonoses , Animais , Vírus Lassa/patogenicidade , Vírus Lassa/fisiologia , Febre Lassa/transmissão , Febre Lassa/epidemiologia , Febre Lassa/virologia , Febre Lassa/veterinária , Reservatórios de Doenças/virologia , Humanos , Ratos , Murinae/virologia , Zoonoses/virologia , Zoonoses/transmissão , Zoonoses/epidemiologia , Serra Leoa/epidemiologia , Guiné/epidemiologia , Ecossistema , Doenças dos Roedores/virologia , Doenças dos Roedores/epidemiologia , Doenças dos Roedores/transmissão
13.
J Vector Borne Dis ; 61(1): 43-50, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38648405

RESUMO

BACKGROUND OBJECTIVES: Leptospirosis is an important zoonotic infection that has caused significant mortality and morbidity worldwide. This disease is endemic in Malaysia and as a developing tropical country, leptospirosis is concerning as it threatens Malaysian public health and the country's economic sectors. However, there is limited information on leptospirosis in Malaysia, especially regarding leptospiral seroepidemiology among carriers in Malaysia. Therefore, more epidemiological information on the source of the disease and reservoir are needed for better disease control and source intervention. The objectives of this study are to gather information on Leptospira infection and the carrier status of rats captured from selected wet markets of Kuala Lumpur metropolitan city in Malaysia. METHODS: Live rat trappings were performed in four major wet markets in Kuala Lumpur, namely, Pudu, Chow Kit, Datuk Keramat, and Petaling Street. Animal samplings were performed for 12 months in 2017, where blood and kidney samples were collected and tested for anti-leptospiral antibodies via Microscopic Agglutination Test (MAT) and pathogenic Leptospira screening via Polymerase Chain Reaction (PCR) amplification offlaB gene. RESULTS: MAT showed that 34.7% (n = 50/144) of the captured rats were positive for anti-leptospiral antibody of which the most prominent serovar was Malaya followed by a local strain, IMR LEP 175. In parallel, 50 rats were also positive for pathogenic Leptospira DNA. INTERPRETATION CONCLUSION: This study showed that there are persistent Leptospira infections among rats in Kuala Lumpur wet markets and these rats are important reservoir hosts for the bacteria.


Assuntos
Anticorpos Antibacterianos , Leptospira , Leptospirose , Animais , Malásia/epidemiologia , Leptospirose/epidemiologia , Leptospirose/veterinária , Leptospirose/microbiologia , Ratos , Leptospira/genética , Leptospira/isolamento & purificação , Anticorpos Antibacterianos/sangue , Portador Sadio/microbiologia , Portador Sadio/epidemiologia , Estudos Soroepidemiológicos , Masculino , Reservatórios de Doenças/microbiologia , Doenças dos Roedores/epidemiologia , Doenças dos Roedores/microbiologia , Feminino , Reação em Cadeia da Polimerase , Testes de Aglutinação
14.
Sci Rep ; 14(1): 9823, 2024 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-38684927

RESUMO

The emergence of infectious diseases with pandemic potential is a major public health threat worldwide. The World Health Organization reports that about 60% of emerging infectious diseases are zoonoses, originating from spillover events. Although the mechanisms behind spillover events remain unclear, mathematical modeling offers a way to understand the intricate interactions among pathogens, wildlife, humans, and their shared environment. Aiming at gaining insights into the dynamics of spillover events and the outcome of an eventual disease outbreak in a population, we propose a continuous time stochastic modeling framework. This framework links the dynamics of animal reservoirs and human hosts to simulate cross-species disease transmission. We conduct a thorough analysis of the model followed by numerical experiments that explore various spillover scenarios. The results suggest that although most epidemic outbreaks caused by novel zoonotic pathogens do not persist in the human population, the rising number of spillover events can avoid long-lasting extinction and lead to unexpected large outbreaks. Hence, global efforts to reduce the impacts of emerging diseases should not only address post-emergence outbreak control but also need to prevent pandemics before they are established.


Assuntos
Doenças Transmissíveis Emergentes , Saúde Pública , Zoonoses , Humanos , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/transmissão , Animais , Zoonoses/epidemiologia , Zoonoses/transmissão , Surtos de Doenças , Modelos Teóricos , Reservatórios de Doenças , Pandemias
15.
Parasit Vectors ; 17(1): 198, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38689318

RESUMO

Canine leishmaniasis is a widespread disease on the American continent, with cases reported from Uruguay to the USA and Canada. While numerous Leishmania spp. have been reported in dogs in this region, Leishmania infantum and Leishmania braziliensis are the most common etiological agents of canine leishmaniasis from a continental perspective. Nonetheless, other species may predominate locally in some countries. The participation of dogs in the transmission cycle of various Leishmania spp. has long been speculated, but evidence indicates that their role as reservoirs of species other than L. infantum is negligible. Various native wildlife (e.g., small rodents, marsupials, sloths, and monkeys) are, in fact, the primary hosts of Leishmania spp. in the Americas. In this review, an updated list of Leishmania spp. infecting dogs in the Americas is presented, along with their distribution and clinical and zoonotic importance.


Assuntos
Doenças do Cão , Leishmaniose , Zoonoses , Animais , Cães , Humanos , América/epidemiologia , Reservatórios de Doenças/parasitologia , Reservatórios de Doenças/veterinária , Doenças do Cão/parasitologia , Doenças do Cão/transmissão , Doenças do Cão/epidemiologia , Leishmania/patogenicidade , Leishmaniose/epidemiologia , Leishmaniose/transmissão , Leishmaniose/veterinária , Zoonoses/transmissão , Zoonoses/parasitologia
16.
Front Cell Infect Microbiol ; 14: 1364002, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38660490

RESUMO

The role of the oral microbiota in the overall health and in systemic diseases has gained more importance in the recent years, mainly due to the systemic effects that are mediated by the chronic inflammation caused by oral diseases, such as periodontitis, through the microbial communities of the mouth. The chronic infection by the human immunodeficiency virus (HIV) interacts at the tissue level (e.g. gut, genital tract, brain) to create reservoirs; the modulation of the gut microbiota by HIV infection is a good example of these interactions. The purpose of the present review is to assess the state of knowledge on the oral microbiota (microbiome, mycobiome and virome) of HIV-infected patients in comparison to that of HIV-negative individuals and to discuss the reciprocal influence of HIV infection and oral microbiota in patients with periodontitis on the potential establishment of a viral gingival reservoir. The influence of different clinical and biological parameters are reviewed including age, immune and viral status, potent antiretroviral therapies, smoking, infection of the airway and viral coinfections, all factors that can modulate the oral microbiota during HIV infection. The analysis of the literature proposed in this review indicates that the comparisons of the available studies are difficult due to their great heterogeneity. However, some important findings emerge: (i) the oral microbiota is less influenced than that of the gut during HIV infection, although some recurrent changes in the microbiome are identified in many studies; (ii) severe immunosuppression is correlated with altered microbiota and potent antiretroviral therapies correct partially these modifications; (iii) periodontitis constitutes a major factor of dysbiosis, which is exacerbated in HIV-infected patients; its pathogenesis can be described as a reciprocal reinforcement of the two conditions, where the local dysbiosis present in the periodontal pocket leads to inflammation, bacterial translocation and destruction of the supporting tissues, which in turn enhances an inflammatory environment that perpetuates the periodontitis cycle. With the objective of curing viral reservoirs of HIV-infected patients in the future years, it appears important to develop further researches aimed at defining whether the inflamed gingiva can serve of viral reservoir in HIV-infected patients with periodontitis.


Assuntos
Gengiva , Infecções por HIV , Microbiota , Humanos , Infecções por HIV/tratamento farmacológico , Infecções por HIV/microbiologia , Infecções por HIV/complicações , Infecções por HIV/virologia , Gengiva/microbiologia , Gengiva/virologia , Boca/microbiologia , Boca/virologia , Reservatórios de Doenças/microbiologia , Reservatórios de Doenças/virologia , Periodontite/microbiologia , Periodontite/virologia , Viroma , Disbiose/microbiologia , Antirretrovirais/uso terapêutico , HIV
17.
Am J Vet Res ; 85(6)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38640951

RESUMO

Antimicrobial resistance (AMR) is a serious health issue shared across all One Health domains. Wildlife species represent a key intersection of the animal and environmental domains. They are a relevant but understudied reservoir and route of spread for AMR throughout the environment. Most wildlife AMR research thus far has focused on avian species, terrestrial mammals, and a selection of aquatic and marine species. Pathogens often identified in terrestrial wildlife include enteric zoonotic organisms such as Eschericia coli and Salmonella spp, in addition to nonenterics such as Staphylococci. Resistances have been commonly identified to antimicrobials important in veterinary and human medicine, including ß-lactams, tetracyclines, aminoglycosides, and macrolides. Our emerging understanding of the dynamics of AMR distribution across life on Earth provides further opportunities for us to assess the risk it poses to veterinary and human health. Future work will require prioritizing which wildlife most exacerbates and indicates AMR in domestic animals. However, decreasing prices and increasing ease for metagenomic sequencing allows for synergies with expanding wildlife viral disease surveillance. Improved understanding of how wildlife impacts veterinary and human healthcare may increase opportunities for related research funding and global equity in such research. The companion Currents in One Health article by Vezeau and Kahn, JAVMA, June 2024, addresses in further detail the routes of spread of AMR across different animal populations and actions that can be taken to mitigate AMR with special consideration for wildlife sources.


Assuntos
Animais Selvagens , Reservatórios de Doenças , Farmacorresistência Bacteriana , Animais , Animais Selvagens/microbiologia , Reservatórios de Doenças/veterinária , Reservatórios de Doenças/microbiologia , Antibacterianos/farmacologia , Humanos , Zoonoses/microbiologia
18.
Viruses ; 16(4)2024 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-38675857

RESUMO

The persistence of the latent viral reservoir is the main hurdle to curing HIV-1 infection. SIV infection of non-human primates (NHPs), namely Indian-origin rhesus macaques, is the most relevant and widely used animal model to evaluate therapies that seek to eradicate HIV-1. The utility of a model ultimately rests on how accurately it can recapitulate human disease, and while reservoirs in the NHP model behave quantitatively very similar to those of long-term suppressed persons with HIV-1 (PWH) in the most salient aspects, recent studies have uncovered key nuances at the clonotypic level that differentiate the two in qualitative terms. In this review, we will highlight differences relating to proviral intactness, clonotypic structure, and decay rate during ART between HIV-1 and SIV reservoirs and discuss the relevance of these distinctions in the interpretation of HIV-1 cure strategies. While these, to some degree, may reflect a unique biology of the virus or host, distinctions among the proviral landscape in SIV are likely to be shaped significantly by the condensed timeframe of NHP studies. ART is generally initiated earlier in the disease course, and animals are virologically suppressed for shorter periods before receiving interventions. Because these are experimental variables dictated by the investigator, we offer guidance on study design for cure-related studies performed in the NHP model. Finally, we highlight the case of GS-9620 (Vesatolimod), an antiviral TLR7 agonist tested in multiple independent pre-clinical studies in which virological outcomes may have been influenced by study-related variables.


Assuntos
Infecções por HIV , HIV-1 , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Latência Viral , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/fisiologia , Animais , HIV-1/genética , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Humanos , Infecções por HIV/virologia , Infecções por HIV/tratamento farmacológico , Latência Viral/efeitos dos fármacos , Modelos Animais de Doenças , Reservatórios de Doenças/virologia , Provírus/genética , Provírus/fisiologia , Carga Viral
19.
Trends Immunol ; 45(3): 198-210, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38453576

RESUMO

Bats are among the most diverse mammalian species, representing over 20% of mammalian diversity. The past two decades have witnessed a disproportionate spillover of viruses from bats to humans compared with other mammalian hosts, attributed to the viral richness within bats, their phylogenetic likeness to humans, and increased human contact with wildlife. Unique evolutionary adaptations in bat genomes, particularly in antiviral protection and immune tolerance genes, enable bats to serve as reservoirs for pandemic-inducing viruses. Here, we discuss current limitations and advances made in understanding the role of bats as drivers of pandemic zoonoses. We also discuss novel technologies that have revealed spatial, dynamic, and physiological factors driving virus and host coevolution.


Assuntos
Quirópteros , Vírus , Animais , Humanos , Reservatórios de Doenças , Filogenia , Zoonoses , Mamíferos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...