Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.443
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nat Immunol ; 19(1): 20-28, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29199281

RESUMEN

Emerging infectious diseases will continue to threaten public health and are sustained by global commerce, travel and disruption of ecological systems. Most pandemic threats are caused by viruses from either zoonotic sources or vector-borne sources. Developing better ways to anticipate and manage the ongoing microbial challenge will be critical for achieving the United Nations Sustainable Development Goals and, conversely, each such goal will affect the ability to control infectious diseases. Here we discuss how technology can be applied effectively to better prepare for and respond to new viral diseases with a focus on new paradigms for vaccine development.


Asunto(s)
Enfermedades Transmisibles Emergentes/inmunología , Vacunación/métodos , Vacunas Virales/inmunología , Virosis/inmunología , Animales , Enfermedades Transmisibles Emergentes/prevención & control , Enfermedades Transmisibles Emergentes/virología , Transmisión de Enfermedad Infecciosa/prevención & control , Humanos , Pandemias/prevención & control , Salud Pública/métodos , Vacunas Virales/uso terapéutico , Virosis/epidemiología , Virosis/prevención & control
2.
Nature ; 623(7985): 132-138, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37853126

RESUMEN

Hospital-based transmission had a dominant role in Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus (SARS-CoV) epidemics1,2, but large-scale studies of its role in the SARS-CoV-2 pandemic are lacking. Such transmission risks spreading the virus to the most vulnerable individuals and can have wider-scale impacts through hospital-community interactions. Using data from acute hospitals in England, we quantify within-hospital transmission, evaluate likely pathways of spread and factors associated with heightened transmission risk, and explore the wider dynamical consequences. We estimate that between June 2020 and March 2021 between 95,000 and 167,000 inpatients acquired SARS-CoV-2 in hospitals (1% to 2% of all hospital admissions in this period). Analysis of time series data provided evidence that patients who themselves acquired SARS-CoV-2 infection in hospital were the main sources of transmission to other patients. Increased transmission to inpatients was associated with hospitals having fewer single rooms and lower heated volume per bed. Moreover, we show that reducing hospital transmission could substantially enhance the efficiency of punctuated lockdown measures in suppressing community transmission. These findings reveal the previously unrecognized scale of hospital transmission, have direct implications for targeting of hospital control measures and highlight the need to design hospitals better equipped to limit the transmission of future high-consequence pathogens.


Asunto(s)
COVID-19 , Infección Hospitalaria , Transmisión de Enfermedad Infecciosa , Pacientes Internos , Pandemias , Humanos , Control de Enfermedades Transmisibles , COVID-19/epidemiología , COVID-19/transmisión , Infección Hospitalaria/epidemiología , Infección Hospitalaria/prevención & control , Infección Hospitalaria/transmisión , Transmisión de Enfermedad Infecciosa/prevención & control , Transmisión de Enfermedad Infecciosa/estadística & datos numéricos , Inglaterra/epidemiología , Hospitales , Pandemias/prevención & control , Pandemias/estadística & datos numéricos , Cuarentena/estadística & datos numéricos , SARS-CoV-2
3.
Nature ; 614(7946): 125-135, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36653448

RESUMEN

The human microbiome is an integral component of the human body and a co-determinant of several health conditions1,2. However, the extent to which interpersonal relations shape the individual genetic makeup of the microbiome and its transmission within and across populations remains largely unknown3,4. Here, capitalizing on more than 9,700 human metagenomes and computational strain-level profiling, we detected extensive bacterial strain sharing across individuals (more than 10 million instances) with distinct mother-to-infant, intra-household and intra-population transmission patterns. Mother-to-infant gut microbiome transmission was considerable and stable during infancy (around 50% of the same strains among shared species (strain-sharing rate)) and remained detectable at older ages. By contrast, the transmission of the oral microbiome occurred largely horizontally and was enhanced by the duration of cohabitation. There was substantial strain sharing among cohabiting individuals, with 12% and 32% median strain-sharing rates for the gut and oral microbiomes, and time since cohabitation affected strain sharing more than age or genetics did. Bacterial strain sharing additionally recapitulated host population structures better than species-level profiles did. Finally, distinct taxa appeared as efficient spreaders across transmission modes and were associated with different predicted bacterial phenotypes linked with out-of-host survival capabilities. The extent of microorganism transmission that we describe underscores its relevance in human microbiome studies5, especially those on non-infectious, microbiome-associated diseases.


Asunto(s)
Bacterias , Transmisión de Enfermedad Infecciosa , Microbioma Gastrointestinal , Ambiente en el Hogar , Microbiota , Boca , Femenino , Humanos , Lactante , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Microbioma Gastrointestinal/genética , Metagenoma , Microbiota/genética , Madres , Boca/microbiología , Transmisión Vertical de Enfermedad Infecciosa , Composición Familiar , Envejecimiento , Factores de Tiempo , Viabilidad Microbiana
5.
Nature ; 595(7866): 205-213, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34194045

RESUMEN

Social and cultural forces shape almost every aspect of infectious disease transmission in human populations, as well as our ability to measure, understand, and respond to epidemics. For directly transmitted infections, pathogen transmission relies on human-to-human contact, with kinship, household, and societal structures shaping contact patterns that in turn determine epidemic dynamics. Social, economic, and cultural forces also shape patterns of exposure, health-seeking behaviour, infection outcomes, the likelihood of diagnosis and reporting of cases, and the uptake of interventions. Although these social aspects of epidemiology are hard to quantify and have limited the generalizability of modelling frameworks in a policy context, new sources of data on relevant aspects of human behaviour are increasingly available. Researchers have begun to embrace data from mobile devices and other technologies as useful proxies for behavioural drivers of disease transmission, but there is much work to be done to measure and validate these approaches, particularly for policy-making. Here we discuss how integrating local knowledge in the design of model frameworks and the interpretation of new data streams offers the possibility of policy-relevant models for public health decision-making as well as the development of robust, generalizable theories about human behaviour in relation to infectious diseases.


Asunto(s)
Enfermedades Transmisibles/epidemiología , Enfermedades Transmisibles/transmisión , Transmisión de Enfermedad Infecciosa , Modelos Biológicos , Condiciones Sociales/estadística & datos numéricos , Clima , Cultura , Conjuntos de Datos como Asunto , Epidemias , Femenino , Humanos , Locomoción , Masculino , Reproducibilidad de los Resultados , Medición de Riesgo , Tiempo (Meteorología)
6.
Mol Biol Evol ; 41(1)2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38168711

RESUMEN

In recent times, pathogen genome sequencing has become increasingly used to investigate infectious disease outbreaks. When genomic data is sampled densely enough amongst infected individuals, it can help resolve who infected whom. However, transmission analysis cannot rely solely on a phylogeny of the genomes but must account for the within-host evolution of the pathogen, which blurs the relationship between phylogenetic and transmission trees. When only a single genome is sampled for each host, the uncertainty about who infected whom can be quite high. Consequently, transmission analysis based on multiple genomes of the same pathogen per host has a clear potential for delivering more precise results, even though it is more laborious to achieve. Here, we present a new methodology that can use any number of genomes sampled from a set of individuals to reconstruct their transmission network. Furthermore, we remove the need for the assumption of a complete transmission bottleneck. We use simulated data to show that our method becomes more accurate as more genomes per host are provided, and that it can infer key infectious disease parameters such as the size of the transmission bottleneck, within-host growth rate, basic reproduction number, and sampling fraction. We demonstrate the usefulness of our method in applications to real datasets from an outbreak of Pseudomonas aeruginosa amongst cystic fibrosis patients and a nosocomial outbreak of Klebsiella pneumoniae.


Asunto(s)
Enfermedades Transmisibles , Humanos , Filogenia , Enfermedades Transmisibles/genética , Enfermedades Transmisibles/epidemiología , Brotes de Enfermedades , Genómica , Mapeo Cromosómico , Transmisión de Enfermedad Infecciosa
7.
N Engl J Med ; 386(24): 2283-2294, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35704480

RESUMEN

BACKGROUND: In June 2019, the Bolivian Ministry of Health reported a cluster of cases of hemorrhagic fever that started in the municipality of Caranavi and expanded to La Paz. The cause of these cases was unknown. METHODS: We obtained samples for next-generation sequencing and virus isolation. Human and rodent specimens were tested by means of virus-specific real-time quantitative reverse-transcriptase-polymerase-chain-reaction assays, next-generation sequencing, and virus isolation. RESULTS: Nine cases of hemorrhagic fever were identified; four of the patients with this illness died. The etiologic agent was identified as Mammarenavirus Chapare mammarenavirus, or Chapare virus (CHAPV), which causes Chapare hemorrhagic fever (CHHF). Probable nosocomial transmission among health care workers was identified. Some patients with CHHF had neurologic manifestations, and those who survived had a prolonged recovery period. CHAPV RNA was detected in a variety of human body fluids (including blood; urine; nasopharyngeal, oropharyngeal, and bronchoalveolar-lavage fluid; conjunctiva; and semen) and in specimens obtained from captured small-eared pygmy rice rats (Oligoryzomys microtis). In survivors of CHHF, viral RNA was detected up to 170 days after symptom onset; CHAPV was isolated from a semen sample obtained 86 days after symptom onset. CONCLUSIONS: M. Chapare mammarenavirus was identified as the etiologic agent of CHHF. Both spillover from a zoonotic reservoir and possible person-to-person transmission were identified. This virus was detected in a rodent species, O. microtis. (Funded by the Bolivian Ministry of Health and others.).


Asunto(s)
Arenavirus del Nuevo Mundo , Fiebre Hemorrágica Americana , ARN Viral , Roedores , Animales , Arenavirus del Nuevo Mundo/genética , Arenavirus del Nuevo Mundo/aislamiento & purificación , Bolivia/epidemiología , Infección Hospitalaria/transmisión , Infección Hospitalaria/virología , Transmisión de Enfermedad Infecciosa , Fiebre Hemorrágica Americana/complicaciones , Fiebre Hemorrágica Americana/genética , Fiebre Hemorrágica Americana/transmisión , Fiebre Hemorrágica Americana/virología , Fiebres Hemorrágicas Virales/genética , Fiebres Hemorrágicas Virales/transmisión , Fiebres Hemorrágicas Virales/virología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Reacción en Cadena de la Polimerasa , ARN Viral/genética , ARN Viral/aislamiento & purificación , Ratas/virología , Roedores/virología , Zoonosis Virales/transmisión , Zoonosis Virales/virología
8.
N Engl J Med ; 386(8): 744-756, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-34986294

RESUMEN

BACKGROUND: Before the emergence of the B.1.617.2 (delta) variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), vaccination reduced transmission of SARS-CoV-2 from vaccinated persons who became infected, potentially by reducing viral loads. Although vaccination still lowers the risk of infection, similar viral loads in vaccinated and unvaccinated persons who are infected with the delta variant call into question the degree to which vaccination prevents transmission. METHODS: We used contact-testing data from England to perform a retrospective observational cohort study involving adult contacts of SARS-CoV-2-infected adult index patients. We used multivariable Poisson regression to investigate associations between transmission and the vaccination status of index patients and contacts and to determine how these associations varied with the B.1.1.7 (alpha) and delta variants and time since the second vaccination. RESULTS: Among 146,243 tested contacts of 108,498 index patients, 54,667 (37%) had positive SARS-CoV-2 polymerase-chain-reaction (PCR) tests. In index patients who became infected with the alpha variant, two vaccinations with either BNT162b2 or ChAdOx1 nCoV-19 (also known as AZD1222), as compared with no vaccination, were independently associated with reduced PCR positivity in contacts (adjusted rate ratio with BNT162b2, 0.32; 95% confidence interval [CI], 0.21 to 0.48; and with ChAdOx1 nCoV-19, 0.48; 95% CI, 0.30 to 0.78). Vaccine-associated reductions in transmission of the delta variant were smaller than those with the alpha variant, and reductions in transmission of the delta variant after two BNT162b2 vaccinations were greater (adjusted rate ratio for the comparison with no vaccination, 0.50; 95% CI, 0.39 to 0.65) than after two ChAdOx1 nCoV-19 vaccinations (adjusted rate ratio, 0.76; 95% CI, 0.70 to 0.82). Variation in cycle-threshold (Ct) values (indicative of viral load) in index patients explained 7 to 23% of vaccine-associated reductions in transmission of the two variants. The reductions in transmission of the delta variant declined over time after the second vaccination, reaching levels that were similar to those in unvaccinated persons by 12 weeks in index patients who had received ChAdOx1 nCoV-19 and attenuating substantially in those who had received BNT162b2. Protection in contacts also declined in the 3-month period after the second vaccination. CONCLUSIONS: Vaccination was associated with a smaller reduction in transmission of the delta variant than of the alpha variant, and the effects of vaccination decreased over time. PCR Ct values at diagnosis of the index patient only partially explained decreased transmission. (Funded by the U.K. Government Department of Health and Social Care and others.).


Asunto(s)
Vacuna BNT162 , COVID-19/transmisión , ChAdOx1 nCoV-19 , Transmisión de Enfermedad Infecciosa/prevención & control , SARS-CoV-2 , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/diagnóstico , COVID-19/virología , Prueba de Ácido Nucleico para COVID-19 , Inglaterra , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Carga Viral
9.
PLoS Biol ; 20(6): e3001685, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35731837

RESUMEN

Historically, emerging and reemerging infectious diseases have caused large, deadly, and expensive multinational outbreaks. Often outbreak investigations aim to identify who infected whom by reconstructing the outbreak transmission tree, which visualizes transmission between individuals as a network with nodes representing individuals and branches representing transmission from person to person. We compiled a database, called OutbreakTrees, of 382 published, standardized transmission trees consisting of 16 directly transmitted diseases ranging in size from 2 to 286 cases. For each tree and disease, we calculated several key statistics, such as tree size, average number of secondary infections, the dispersion parameter, and the proportion of cases considered superspreaders, and examined how these statistics varied over the course of each outbreak and under different assumptions about the completeness of outbreak investigations. We demonstrated the potential utility of the database through 2 short analyses addressing questions about superspreader epidemiology for a variety of diseases, including Coronavirus Disease 2019 (COVID-19). First, we found that our transmission trees were consistent with theory predicting that intermediate dispersion parameters give rise to the highest proportion of cases causing superspreading events. Additionally, we investigated patterns in how superspreaders are infected. Across trees with more than 1 superspreader, we found preliminary support for the theory that superspreaders generate other superspreaders. In sum, our findings put the role of superspreading in COVID-19 transmission in perspective with that of other diseases and suggest an approach to further research regarding the generation of superspreaders. These data have been made openly available to encourage reuse and further scientific inquiry.


Asunto(s)
COVID-19 , Árboles de Decisión , COVID-19/epidemiología , Brotes de Enfermedades , Transmisión de Enfermedad Infecciosa , Humanos
10.
Trends Genet ; 37(1): 8-11, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33020021

RESUMEN

Ticks exist across diverse environments and transmit numerous pathogens. Due to their long and unique life cycles, these arthropods likely evolved robust epigenetic mechanisms that provide sustainable responses and buffers against extreme environmental conditions. Herein, we highlight how the study of the epigenetic basis of tick biology and vectorial capacity will enrich our knowledge of tick-borne infections.


Asunto(s)
Vectores Artrópodos/crecimiento & desarrollo , Transmisión de Enfermedad Infecciosa , Epigénesis Genética , Interacciones Huésped-Patógeno , Enfermedades por Picaduras de Garrapatas/epidemiología , Garrapatas/fisiología , Animales , Vectores Artrópodos/clasificación , Vectores Artrópodos/genética , Humanos , Enfermedades por Picaduras de Garrapatas/genética , Enfermedades por Picaduras de Garrapatas/transmisión , Garrapatas/microbiología , Garrapatas/parasitología , Garrapatas/virología
11.
N Engl J Med ; 384(5): 417-427, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33289973

RESUMEN

BACKGROUND: Current strategies for preventing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are limited to nonpharmacologic interventions. Hydroxychloroquine has been proposed as a postexposure therapy to prevent coronavirus disease 2019 (Covid-19), but definitive evidence is lacking. METHODS: We conducted an open-label, cluster-randomized trial involving asymptomatic contacts of patients with polymerase-chain-reaction (PCR)-confirmed Covid-19 in Catalonia, Spain. We randomly assigned clusters of contacts to the hydroxychloroquine group (which received the drug at a dose of 800 mg once, followed by 400 mg daily for 6 days) or to the usual-care group (which received no specific therapy). The primary outcome was PCR-confirmed, symptomatic Covid-19 within 14 days. The secondary outcome was SARS-CoV-2 infection, defined by symptoms compatible with Covid-19 or a positive PCR test regardless of symptoms. Adverse events were assessed for up to 28 days. RESULTS: The analysis included 2314 healthy contacts of 672 index case patients with Covid-19 who were identified between March 17 and April 28, 2020. A total of 1116 contacts were randomly assigned to receive hydroxychloroquine and 1198 to receive usual care. Results were similar in the hydroxychloroquine and usual-care groups with respect to the incidence of PCR-confirmed, symptomatic Covid-19 (5.7% and 6.2%, respectively; risk ratio, 0.86 [95% confidence interval, 0.52 to 1.42]). In addition, hydroxychloroquine was not associated with a lower incidence of SARS-CoV-2 transmission than usual care (18.7% and 17.8%, respectively). The incidence of adverse events was higher in the hydroxychloroquine group than in the usual-care group (56.1% vs. 5.9%), but no treatment-related serious adverse events were reported. CONCLUSIONS: Postexposure therapy with hydroxychloroquine did not prevent SARS-CoV-2 infection or symptomatic Covid-19 in healthy persons exposed to a PCR-positive case patient. (Funded by the crowdfunding campaign YoMeCorono and others; BCN-PEP-CoV2 ClinicalTrials.gov number, NCT04304053.).


Asunto(s)
Antiinfecciosos/uso terapéutico , COVID-19/prevención & control , Hidroxicloroquina/uso terapéutico , SARS-CoV-2 , Adulto , Antiinfecciosos/efectos adversos , COVID-19/transmisión , COVID-19/virología , Transmisión de Enfermedad Infecciosa/prevención & control , Método Doble Ciego , Femenino , Humanos , Hidroxicloroquina/efectos adversos , Masculino , Persona de Mediana Edad , Cooperación del Paciente , Insuficiencia del Tratamiento , Carga Viral
12.
J Virol ; 97(5): e0188822, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-36975793

RESUMEN

Zaïre ebolavirus (EBOV) causes Ebola virus disease (EVD), a devastating viral hemorrhagic fever in humans. Nonhuman primate (NHP) models of EVD traditionally use intramuscular infection with higher case fatality rates and reduced mean time-to-death compared to contact transmission typical of human cases of EVD. A cynomolgus macaque model of oral and conjunctival EBOV was used to further characterize the more clinically relevant contact transmission of EVD. NHPs challenged via the oral route had an overall 50% survival rate. NHPs challenged with a target dose of 1 × 102 PFU or 1 × 104 PFU of EBOV via the conjunctival route had 40% and 100% mortality, respectively. Classic signs of lethal EVD-like disease were observed in all NHPs that succumbed to EBOV infection including viremia, hematological abnormalities, clinical chemistries indicative of hepatic and renal disease, and histopathological findings. Evidence of EBOV viral persistence in the eye was observed in NHPs challenged via the conjunctival route. IMPORTANCE This study is the first to examine the Kikwit strain of EBOV, the most commonly used strain, in the gold-standard macaque model of infection. Additionally, this is the first description of the detection of virus in the vitreous fluid, an immune privileged site that has been proposed as a viral reservoir, following conjunctival challenge. The oral and conjunctival macaque challenge model of EVD described here more faithfully recapitulates the prodrome that has been reported for human EVD. This work paves the way for more advanced studies to model contact transmission of EVD, including early events in mucosal infection and immunity, as well as the establishment of persistent viral infection and the emergence from these reservoirs.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Humanos , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/transmisión , Macaca fascicularis , Modelos Animales de Enfermedad , Conjuntiva/virología , Transmisión de Enfermedad Infecciosa
13.
Nature ; 563(7733): 710-713, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30455422

RESUMEN

Understanding host interactions that lead to pathogen transmission is fundamental to the prediction and control of epidemics1-5. Although the majority of transmissions often occurs within social groups6-9, the contribution of connections that bridge groups and species to pathogen dynamics is poorly understood10-12. These cryptic connections-which are often indirect or infrequent-provide transmission routes between otherwise disconnected individuals and may have a key role in large-scale outbreaks that span multiple populations or species. Here we quantify the importance of cryptic connections in disease dynamics by simultaneously characterizing social networks and tracing transmission dynamics of surrogate-pathogen epidemics through eight communities of bats. We then compared these data to the invasion of the fungal pathogen that causes white-nose syndrome, a recently emerged disease that is devastating North American bat populations13-15. We found that cryptic connections increased links between individuals and between species by an order of magnitude. Individuals were connected, on average, to less than two per cent of the population through direct contact and to only six per cent through shared groups. However, tracing surrogate-pathogen dynamics showed that each individual was connected to nearly fifteen per cent of the population, and revealed widespread transmission between solitarily roosting individuals as well as extensive contacts among species. Connections estimated from surrogate-pathogen epidemics, which include cryptic connections, explained three times as much variation in the transmission of the fungus that causes white-nose syndrome as did connections based on shared groups. These findings show how cryptic connections facilitate the community-wide spread of pathogens and can lead to explosive epidemics.


Asunto(s)
Ascomicetos/patogenicidad , Quirópteros/microbiología , Trazado de Contacto/veterinaria , Transmisión de Enfermedad Infecciosa/veterinaria , Micosis/veterinaria , Sistemas de Identificación Animal , Animales , Control de Enfermedades Transmisibles , Trazado de Contacto/métodos , Transmisión de Enfermedad Infecciosa/estadística & datos numéricos , Polvo/análisis , Hibernación , Humanos , Masculino , Micosis/epidemiología , Micosis/microbiología , Micosis/transmisión , Red Social , Zoonosis/microbiología , Zoonosis/transmisión
14.
Nature ; 564(7736): 415-419, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30546139

RESUMEN

We previously reported1 the presence of amyloid-ß protein (Aß) deposits in individuals with Creutzfeldt-Jakob disease (CJD) who had been treated during childhood with human cadaveric pituitary-derived growth hormone (c-hGH) contaminated with prions. The marked deposition of parenchymal and vascular Aß in these relatively young individuals with treatment-induced (iatrogenic) CJD (iCJD), in contrast to other prion-disease patients and population controls, allied with the ability of Alzheimer's disease brain homogenates to seed Aß deposition in laboratory animals, led us to argue that the implicated c-hGH batches might have been contaminated with Aß seeds as well as with prions. However, this was necessarily an association, and not an experimental, study in humans and causality could not be concluded. Given the public health importance of our hypothesis, we proceeded to identify and biochemically analyse archived vials of c-hGH. Here we show that certain c-hGH batches to which patients with iCJD and Aß pathology were exposed have substantial levels of Aß40, Aß42 and tau proteins, and that this material can seed the formation of Aß plaques and cerebral Aß-amyloid angiopathy in intracerebrally inoculated mice expressing a mutant, humanized amyloid precursor protein. These results confirm the presence of Aß seeds in archived c-hGH vials and are consistent with the hypothesized iatrogenic human transmission of Aß pathology. This experimental confirmation has implications for both the prevention and the treatment of Alzheimer's disease, and should prompt a review of the risk of iatrogenic transmission of Aß seeds by medical and surgical procedures long recognized to pose a risk of accidental prion transmission2,3.


Asunto(s)
Enfermedad de Alzheimer/inducido químicamente , Péptidos beta-Amiloides/metabolismo , Cadáver , Síndrome de Creutzfeldt-Jakob/inducido químicamente , Contaminación de Medicamentos , Hormona del Crecimiento/farmacología , Enfermedad Iatrogénica , Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/análisis , Precursor de Proteína beta-Amiloide/administración & dosificación , Precursor de Proteína beta-Amiloide/efectos adversos , Animales , Estudios de Casos y Controles , Síndrome de Creutzfeldt-Jakob/etiología , Modelos Animales de Enfermedad , Transmisión de Enfermedad Infecciosa/prevención & control , Transmisión de Enfermedad Infecciosa/estadística & datos numéricos , Contaminación de Medicamentos/prevención & control , Contaminación de Medicamentos/estadística & datos numéricos , Femenino , Hormona del Crecimiento/administración & dosificación , Humanos , Masculino , Ratones , Modelos Biológicos , Priones/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Reproducibilidad de los Resultados , Proteínas tau/análisis , Proteínas tau/metabolismo
15.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33397804

RESUMEN

Hepatitis delta virus (HDV) is an unusual RNA agent that replicates using host machinery but exploits hepatitis B virus (HBV) to mobilize its spread within and between hosts. In doing so, HDV enhances the virulence of HBV. How this seemingly improbable hyperparasitic lifestyle emerged is unknown, but it underpins the likelihood that HDV and related deltaviruses may alter other host-virus interactions. Here, we show that deltaviruses diversify by transmitting between mammalian species. Among 96,695 RNA sequence datasets, deltaviruses infected bats, rodents, and an artiodactyl from the Americas but were absent from geographically overrepresented Old World representatives of each mammalian order, suggesting a relatively recent diversification within the Americas. Consistent with diversification by host shifting, both bat and rodent-infecting deltaviruses were paraphyletic, and coevolutionary modeling rejected cospeciation with mammalian hosts. In addition, a 2-y field study showed common vampire bats in Peru were infected by two divergent deltaviruses, indicating multiple introductions to a single host species. One vampire bat-associated deltavirus was detected in the saliva of up to 35% of individuals, formed phylogeographically compartmentalized clades, and infected a sympatric bat, illustrating horizontal transmission within and between species on ecological timescales. Consistent absence of HBV-like viruses in two deltavirus-infected bat species indicated acquisitions of novel viral associations during the divergence of bat and human-infecting deltaviruses. Our analyses support an American zoonotic origin of HDV and reveal prospects for future cross-species emergence of deltaviruses. Given their peculiar life history, deltavirus host shifts will have different constraints and disease outcomes compared to ordinary animal pathogens.


Asunto(s)
Virus de la Hepatitis B/genética , Virus de la Hepatitis Delta/genética , Especificidad del Huésped/genética , Virus Satélites/genética , Animales , Quirópteros/virología , Transmisión de Enfermedad Infecciosa , Variación Genética/genética , Genoma Viral/genética , Hepatitis B/genética , Hepatitis B/transmisión , Hepatitis B/virología , Virus de la Hepatitis B/patogenicidad , Hepatitis D/genética , Hepatitis D/transmisión , Hepatitis D/virología , Virus de la Hepatitis Delta/patogenicidad , Interacciones Huésped-Patógeno/genética , Humanos , Mamíferos/virología , Filogenia , Roedores/virología , Virus Satélites/patogenicidad
16.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33766888

RESUMEN

We estimate the effects of shelter-in-place (SIP) orders during the first wave of the COVID-19 pandemic. We do not find detectable effects of these policies on disease spread or deaths. We find small but measurable effects on mobility that dissipate over time. And we find small, delayed effects on unemployment. We conduct additional analyses that separately assess the effects of expanding versus withdrawing SIP orders and test whether there are spillover effects in other states. Our results are consistent with prior studies showing that SIP orders have accounted for a relatively small share of the mobility trends and economic disruptions associated with the pandemic. We reanalyze two prior studies purporting to show that SIP orders caused large reductions in disease prevalence, and show that those results are not reliable. Our results do not imply that social distancing behavior by individuals, as distinct from SIP policy, is ineffective.


Asunto(s)
COVID-19/epidemiología , Refugio de Emergencia , COVID-19/prevención & control , COVID-19/transmisión , Transmisión de Enfermedad Infecciosa/prevención & control , Refugio de Emergencia/economía , Refugio de Emergencia/tendencias , Humanos , Pandemias , Distanciamiento Físico , Políticas , Prevalencia , SARS-CoV-2/aislamiento & purificación , Factores de Tiempo , Desempleo/estadística & datos numéricos
17.
J Med Internet Res ; 26: e44249, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-37967280

RESUMEN

BACKGROUND: The correlates responsible for the temporal changes of intrahousehold SARS-CoV-2 transmission in the United States have been understudied mainly due to a lack of available surveillance data. Specifically, early analyses of SARS-CoV-2 household secondary attack rates (SARs) were small in sample size and conducted cross-sectionally at single time points. From these limited data, it has been difficult to assess the role that different risk factors have had on intrahousehold disease transmission in different stages of the ongoing COVID-19 pandemic, particularly in children and youth. OBJECTIVE: This study aimed to estimate the transmission dynamic and infectivity of SARS-CoV-2 among pediatric and young adult index cases (age 0 to 25 years) in the United States through the initial waves of the pandemic. METHODS: Using administrative claims, we analyzed 19 million SARS-CoV-2 test records between January 2020 and February 2021. We identified 36,241 households with pediatric index cases and calculated household SARs utilizing complete case information. Using a retrospective cohort design, we estimated the household SARS-CoV-2 transmission between 4 index age groups (0 to 4 years, 5 to 11 years, 12 to 17 years, and 18 to 25 years) while adjusting for sex, family size, quarter of first SARS-CoV-2 positive record, and residential regions of the index cases. RESULTS: After filtering all household records for greater than one member in a household and missing information, only 36,241 (0.85%) of 4,270,130 households with a pediatric case remained in the analysis. Index cases aged between 0 and 17 years were a minority of the total index cases (n=11,484, 11%). The overall SAR of SARS-CoV-2 was 23.04% (95% CI 21.88-24.19). As a comparison, the SAR for all ages (0 to 65+ years) was 32.4% (95% CI 32.1-32.8), higher than the SAR for the population between 0 and 25 years of age. The highest SAR of 38.3% was observed in April 2020 (95% CI 31.6-45), while the lowest SAR of 15.6% was observed in September 2020 (95% CI 13.9-17.3). It consistently decreased from 32% to 21.1% as the age of index groups increased. In a multiple logistic regression analysis, we found that the youngest pediatric age group (0 to 4 years) had 1.69 times (95% CI 1.42-2.00) the odds of SARS-CoV-2 transmission to any family members when compared with the oldest group (18 to 25 years). Family size was significantly associated with household viral transmission (odds ratio 2.66, 95% CI 2.58-2.74). CONCLUSIONS: Using retrospective claims data, the pediatric index transmission of SARS-CoV-2 during the initial waves of the COVID-19 pandemic in the United States was associated with location and family characteristics. Pediatric SAR (0 to 25 years) was less than the SAR for all age other groups. Less than 1% (n=36,241) of all household data were retained in the retrospective study for complete case analysis, perhaps biasing our findings. We have provided measures of baseline household pediatric transmission for tracking and comparing the infectivity of later SARS-CoV-2 variants.


Asunto(s)
COVID-19 , Transmisión de Enfermedad Infecciosa , SARS-CoV-2 , Adolescente , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Adulto Joven , COVID-19/epidemiología , Composición Familiar , Pandemias , Estudios Retrospectivos , Estados Unidos/epidemiología
18.
Emerg Infect Dis ; 29(8): 1627-1629, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37486205

RESUMEN

Streptococcus agalactiae (group B Streptococcus) sequence type 283 bacteremia, found almost exclusively in Southeast Asia, is associated with consuming raw freshwater fish, but some patients deny consumption. We detected fecal carriage in 5/184 (2.7%) persons in northeast Thailand. Human carriers might contribute to transmission or be the original source of this sequence type.


Asunto(s)
Heces , Infecciones Estreptocócicas , Streptococcus agalactiae , Animales , Humanos , Asia Sudoriental , Peces/metabolismo , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/transmisión , Streptococcus agalactiae/genética , Streptococcus agalactiae/aislamiento & purificación , Tailandia/epidemiología , Heces/microbiología , Prevalencia , Transmisión de Enfermedad Infecciosa/estadística & datos numéricos
19.
Annu Rev Med ; 72: 107-118, 2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33502896

RESUMEN

Implementation of the HIV Organ Policy Equity (HOPE) Act marks a new era in transplantation, allowing organ transplantation from HIV+ donors to HIV+ recipients (HIV D+/R+ transplantation). In this review, we discuss major milestones in HIV and transplantation which paved the way for this landmark policy change, including excellent outcomes in HIV D-/R+ recipient transplantation and success in the South African experience of HIV D+/R+ deceased donor kidney transplantation. Under the HOPE Act, from March 2016 to December 2018, there were 56 deceased donors, and 102 organs were transplanted (71 kidneys and 31 livers). In 2019, the first HIV D+/R+ living donor kidney transplants occurred. Reaching the full estimated potential of HIV+ donors will require overcoming challenges at the community, organ procurement organization, and transplant center levels. Multiple clinical trials are ongoing, which will provide clinical and scientific data to further extend the frontiers of knowledge in this field.


Asunto(s)
Transmisión de Enfermedad Infecciosa/estadística & datos numéricos , Infecciones por VIH/epidemiología , VIH , Trasplante de Órganos/métodos , Donantes de Tejidos/provisión & distribución , Receptores de Trasplantes , Comorbilidad , Salud Global , Infecciones por VIH/transmisión , Humanos
20.
Funct Integr Genomics ; 23(1): 36, 2023 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-36631570

RESUMEN

In comparison to previously known severe respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, the newly emerged Omicron (B.1.1.529) variant shows higher infectivity in humans. Exceptionally high infectivity of this variant raises concern of its possible transmission via other intermediate hosts. The SARS-CoV-2 infectivity is established via the association of spike (S) protein receptor binding domain (RBD) with host angiotensin I converting enzyme 2 (hACE2) receptor. In the course of this study, we investigated the interaction between Omicron S protein RBD with the ACE2 receptor of 143 mammalian hosts including human by protein-protein interaction analysis. The goal of this study was to forecast the likelihood that the virus may infect other mammalian species that coexist with or are close to humans in the household, rural, agricultural, or zoological environments. The Omicron RBD was found to interact with higher binding affinity with the ACE2 receptor of 122 mammalian hosts via different amino acid residues from the human ACE2 (hACE2). The rat (Rattus rattus) ACE2 was found to show the strongest interaction with Omicron RBD with a binding affinity of -1393.6 kcal/mol. These distinct strong binding affinity of RBD of Omicron with host ACE2 indicates a greater potential of new host transmissibility and infection via intermediate hosts. Though expected but the phylogenetic position of the mammalian species may not dictate the Omicron RBD binding to the host ACE2 receptor suggesting an involvement of multiple factors in guiding host divergence of the variant.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Transmisión de Enfermedad Infecciosa , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Animales , Humanos , Ratas , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/virología , Mamíferos , Mutación , Filogenia , Unión Proteica , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA