Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(9): 1556-1571.e18, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35447072

RESUMEN

SARS-CoV-2 Omicron is highly transmissible and has substantial resistance to neutralization following immunization with ancestral spike-matched vaccines. It is unclear whether boosting with Omicron-matched vaccines would enhance protection. Here, nonhuman primates that received mRNA-1273 at weeks 0 and 4 were boosted at week 41 with mRNA-1273 or mRNA-Omicron. Neutralizing titers against D614G were 4,760 and 270 reciprocal ID50 at week 6 (peak) and week 41 (preboost), respectively, and 320 and 110 for Omicron. 2 weeks after the boost, titers against D614G and Omicron increased to 5,360 and 2,980 for mRNA-1273 boost and 2,670 and 1,930 for mRNA-Omicron, respectively. Similar increases against BA.2 were observed. Following either boost, 70%-80% of spike-specific B cells were cross-reactive against WA1 and Omicron. Equivalent control of virus replication in lower airways was observed following Omicron challenge 1 month after either boost. These data show that mRNA-1273 and mRNA-Omicron elicit comparable immunity and protection shortly after the boost.


Asunto(s)
COVID-19 , SARS-CoV-2 , Vacuna nCoV-2019 mRNA-1273 , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Macaca , ARN Mensajero
2.
Cell ; 185(1): 113-130.e15, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34921774

RESUMEN

mRNA-1273 vaccine efficacy against SARS-CoV-2 Delta wanes over time; however, there are limited data on the impact of durability of immune responses on protection. Here, we immunized rhesus macaques and assessed immune responses over 1 year in blood and upper and lower airways. Serum neutralizing titers to Delta were 280 and 34 reciprocal ID50 at weeks 6 (peak) and 48 (challenge), respectively. Antibody-binding titers also decreased in bronchoalveolar lavage (BAL). Four days after Delta challenge, the virus was unculturable in BAL, and subgenomic RNA declined by ∼3-log10 compared with control animals. In nasal swabs, sgRNA was reduced by 1-log10, and the virus remained culturable. Anamnestic antibodies (590-fold increased titer) but not T cell responses were detected in BAL by day 4 post-challenge. mRNA-1273-mediated protection in the lungs is durable but delayed and potentially dependent on anamnestic antibody responses. Rapid and sustained protection in upper and lower airways may eventually require a boost.

3.
Cell ; 184(16): 4203-4219.e32, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34242577

RESUMEN

SARS-CoV-2-neutralizing antibodies (NAbs) protect against COVID-19. A concern regarding SARS-CoV-2 antibodies is whether they mediate disease enhancement. Here, we isolated NAbs against the receptor-binding domain (RBD) or the N-terminal domain (NTD) of SARS-CoV-2 spike from individuals with acute or convalescent SARS-CoV-2 or a history of SARS-CoV infection. Cryo-electron microscopy of RBD and NTD antibodies demonstrated function-specific modes of binding. Select RBD NAbs also demonstrated Fc receptor-γ (FcγR)-mediated enhancement of virus infection in vitro, while five non-neutralizing NTD antibodies mediated FcγR-independent in vitro infection enhancement. However, both types of infection-enhancing antibodies protected from SARS-CoV-2 replication in monkeys and mice. Three of 46 monkeys infused with enhancing antibodies had higher lung inflammation scores compared to controls. One monkey had alveolar edema and elevated bronchoalveolar lavage inflammatory cytokines. Thus, while in vitro antibody-enhanced infection does not necessarily herald enhanced infection in vivo, increased lung inflammation can rarely occur in SARS-CoV-2 antibody-infused macaques.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , Anticuerpos Antivirales/inmunología , Líquido del Lavado Bronquioalveolar/química , COVID-19/patología , COVID-19/virología , Citocinas/metabolismo , Femenino , Haplorrinos , Humanos , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Dominios Proteicos , ARN Guía de Kinetoplastida/metabolismo , Receptores de IgG/metabolismo , SARS-CoV-2/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/química , Carga Viral , Replicación Viral
4.
Nat Immunol ; 22(10): 1306-1315, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34417590

RESUMEN

B.1.351 is the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant most resistant to antibody neutralization. We demonstrate how the dose and number of immunizations influence protection. Nonhuman primates received two doses of 30 or 100 µg of Moderna's mRNA-1273 vaccine, a single immunization of 30 µg, or no vaccine. Two doses of 100 µg of mRNA-1273 induced 50% inhibitory reciprocal serum dilution neutralizing antibody titers against live SARS-CoV-2 p.Asp614Gly and B.1.351 of 3,300 and 240, respectively. Higher neutralizing responses against B.1.617.2 were also observed after two doses compared to a single dose. After challenge with B.1.351, there was ~4- to 5-log10 reduction of viral subgenomic RNA and low to undetectable replication in bronchoalveolar lavages in the two-dose vaccine groups, with a 1-log10 reduction in nasal swabs in the 100-µg group. These data establish that a two-dose regimen of mRNA-1273 will be critical for providing upper and lower airway protection against major variants of concern.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Primates/inmunología , SARS-CoV-2/inmunología , Vacuna nCoV-2019 mRNA-1273 , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/virología , Línea Celular , Chlorocebus aethiops , Femenino , Humanos , Macaca mulatta , Masculino , Mesocricetus , Primates/virología , ARN Viral/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunación/métodos , Células Vero , Carga Viral/métodos
5.
Immunity ; 54(8): 1869-1882.e6, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34270939

RESUMEN

Vaccine-associated enhanced respiratory disease (VAERD) was previously observed in some preclinical models of severe acute respiratory syndrome (SARS) and MERS coronavirus vaccines. We used the SARS coronavirus 2 (SARS-CoV-2) mouse-adapted, passage 10, lethal challenge virus (MA10) mouse model of acute lung injury to evaluate the immune response and potential for immunopathology in animals vaccinated with research-grade mRNA-1273. Whole-inactivated virus or heat-denatured spike protein subunit vaccines with alum designed to elicit low-potency antibodies and Th2-skewed CD4+ T cells resulted in reduced viral titers and weight loss post challenge but more severe pathological changes in the lung compared to saline-immunized animals. In contrast, a protective dose of mRNA-1273 induced favorable humoral and cellular immune responses that protected from viral replication in the upper and lower respiratory tract upon challenge. A subprotective dose of mRNA-1273 reduced viral replication and limited histopathological manifestations compared to animals given saline. Overall, our findings demonstrate an immunological signature associated with antiviral protection without disease enhancement following vaccination with mRNA-1273.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , COVID-19/prevención & control , Interacciones Huésped-Patógeno/inmunología , SARS-CoV-2/inmunología , Vacunas Sintéticas/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Biopsia , Vacunas contra la COVID-19/administración & dosificación , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina G , Inmunohistoquímica , Ratones , Evaluación de Resultado en la Atención de Salud , ARN Mensajero , Glicoproteína de la Espiga del Coronavirus/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Vacunas Sintéticas/administración & dosificación , Vacunas de ARNm
6.
Nat Immunol ; 17(5): 538-44, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27043413

RESUMEN

Acidic mammalian chitinase (AMCase) is known to be induced by allergens and helminths, yet its role in immunity is unclear. Using AMCase-deficient mice, we show that AMCase deficiency reduced the number of group 2 innate lymphoid cells during allergen challenge but was not required for establishment of type 2 inflammation in the lung in response to allergens or helminths. In contrast, AMCase-deficient mice showed a profound defect in type 2 immunity following infection with the chitin-containing gastrointestinal nematodes Nippostrongylus brasiliensis and Heligmosomoides polygyrus bakeri. The impaired immunity was associated with reduced mucus production and decreased intestinal expression of the signature type 2 response genes Il13, Chil3, Retnlb, and Clca1. CD103(+) dendritic cells, which regulate T cell homing, were also reduced in mesenteric lymph nodes of infected AMCase-deficient mice. Thus, AMCase functions as a critical initiator of protective type 2 responses to intestinal nematodes but is largely dispensable for allergic responses in the lung.


Asunto(s)
Quitinasas/inmunología , Tracto Gastrointestinal/inmunología , Inmunidad/inmunología , Infecciones por Strongylida/inmunología , Animales , Quitinasas/genética , Quitinasas/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/inmunología , Canales de Cloruro/metabolismo , Citometría de Flujo , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/parasitología , Expresión Génica/inmunología , Hormonas Ectópicas/genética , Hormonas Ectópicas/inmunología , Hormonas Ectópicas/metabolismo , Interacciones Huésped-Parásitos/inmunología , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Inmunidad/genética , Péptidos y Proteínas de Señalización Intercelular , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-13/metabolismo , Lectinas/genética , Lectinas/inmunología , Lectinas/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Nematospiroides dubius/inmunología , Nematospiroides dubius/fisiología , Nippostrongylus/inmunología , Nippostrongylus/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/inmunología , beta-N-Acetilhexosaminidasas/metabolismo
7.
Nature ; 594(7864): 553-559, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33971664

RESUMEN

Betacoronaviruses caused the outbreaks of severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome, as well as the current pandemic of SARS coronavirus 2 (SARS-CoV-2)1-4. Vaccines that elicit protective immunity against SARS-CoV-2 and betacoronaviruses that circulate in animals have the potential to prevent future pandemics. Here we show that the immunization of macaques with nanoparticles conjugated with the receptor-binding domain of SARS-CoV-2, and adjuvanted with 3M-052 and alum, elicits cross-neutralizing antibody responses against bat coronaviruses, SARS-CoV and SARS-CoV-2 (including the B.1.1.7, P.1 and B.1.351 variants). Vaccination of macaques with these nanoparticles resulted in a 50% inhibitory reciprocal serum dilution (ID50) neutralization titre of 47,216 (geometric mean) for SARS-CoV-2, as well as in protection against SARS-CoV-2 in the upper and lower respiratory tracts. Nucleoside-modified mRNAs that encode a stabilized transmembrane spike or monomeric receptor-binding domain also induced cross-neutralizing antibody responses against SARS-CoV and bat coronaviruses, albeit at lower titres than achieved with the nanoparticles. These results demonstrate that current mRNA-based vaccines may provide some protection from future outbreaks of zoonotic betacoronaviruses, and provide a multimeric protein platform for the further development of vaccines against multiple (or all) betacoronaviruses.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Betacoronavirus/inmunología , COVID-19/inmunología , COVID-19/prevención & control , Resfriado Común/prevención & control , Reacciones Cruzadas/inmunología , Pandemias , Vacunas Virales/inmunología , Adyuvantes Inmunológicos , Administración Intranasal , Animales , COVID-19/epidemiología , Vacunas contra la COVID-19/inmunología , Resfriado Común/inmunología , Resfriado Común/virología , Modelos Animales de Enfermedad , Femenino , Humanos , Macaca/inmunología , Masculino , Modelos Moleculares , Nanopartículas/química , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Tráquea , Vacunación
8.
Nature ; 586(7830): 567-571, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32756549

RESUMEN

A vaccine for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is needed to control the coronavirus disease 2019 (COVID-19) global pandemic. Structural studies have led to the development of mutations that stabilize Betacoronavirus spike proteins in the prefusion state, improving their expression and increasing immunogenicity1. This principle has been applied to design mRNA-1273, an mRNA vaccine that encodes a SARS-CoV-2 spike protein that is stabilized in the prefusion conformation. Here we show that mRNA-1273 induces potent neutralizing antibody responses to both wild-type (D614) and D614G mutant2 SARS-CoV-2 as well as CD8+ T cell responses, and protects against SARS-CoV-2 infection in the lungs and noses of mice without evidence of immunopathology. mRNA-1273 is currently in a phase III trial to evaluate its efficacy.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/inmunología , Neumonía Viral/prevención & control , Vacunas Virales/inmunología , Vacuna nCoV-2019 mRNA-1273 , Animales , Anticuerpos Neutralizantes/inmunología , Betacoronavirus/genética , Linfocitos T CD8-positivos/inmunología , COVID-19 , Vacunas contra la COVID-19 , Ensayos Clínicos Fase III como Asunto , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/virología , Femenino , Pulmón/inmunología , Pulmón/virología , Ratones , Mutación , Nariz/inmunología , Nariz/virología , Neumonía Viral/virología , ARN Mensajero/genética , ARN Viral/genética , SARS-CoV-2 , Células TH1/inmunología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/inmunología , Vacunas Virales/química , Vacunas Virales/genética
9.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33888584

RESUMEN

Older age at the time of infection with hepatitis viruses is associated with an increased risk of liver fibrosis progression. We hypothesized that the pace of fibrosis progression may reflect changes in gene expression within the aging liver. We compared gene expression in liver specimens from 54 adult donors without evidence of fibrosis, including 36 over 40 y old and 18 between 18 and 40 y old. Chitinase 3-like 1 (CHI3L1), which encodes chitinase-like protein YKL-40/CHI3L1, was identified as the gene with the greatest age-dependent increase in expression in liver tissue. We investigated the cellular source of CHI3L1 in the liver and its function using liver tissue specimens and in vitro models. CHI3L1 expression was significantly higher in livers of patients with cirrhosis of diverse etiologies compared with controls represented by patients who underwent liver resection for hemangioma. The highest intrahepatic CHI3L1 expression was observed in cirrhosis due to hepatitis D virus, followed by hepatitis C virus, hepatitis B virus, and alcohol-induced cirrhosis. In situ hybridization of CHI3L1 messenger RNA (mRNA) identified hepatocytes as the major producers of CHI3L1 in normal liver and in cirrhotic tissue, wherein hepatocytes adjacent to fibrous septa showed higher CHI3L1 expression than did those in more distal areas. In vitro studies showed that recombinant CHI3L1 promotes proliferation and activation of primary human hepatic stellate cells (HSCs), the major drivers of liver fibrosis. These findings collectively demonstrate that CHI3L1 promotes liver fibrogenesis through a direct effect on HSCs and support a role for CHI3L1 in the increased susceptibility of aging livers to fibrosis progression.


Asunto(s)
Proteína 1 Similar a Quitinasa-3/metabolismo , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Adolescente , Adulto , Envejecimiento/fisiología , Biomarcadores/metabolismo , Proteína 1 Similar a Quitinasa-3/fisiología , Quitinasas/metabolismo , Femenino , Expresión Génica/genética , Hepacivirus/patogenicidad , Células Estrelladas Hepáticas/patología , Hepatitis C/metabolismo , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Hígado/citología , Masculino
10.
Am J Epidemiol ; 192(10): 1669-1677, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37191334

RESUMEN

The severe acute respiratory syndrome (SARS-CoV-2) pandemic and high hospitalization rates placed a tremendous strain on hospital resources, necessitating the use of models to predict hospital volumes and the associated resource requirements. Complex epidemiologic models have been developed and published, but many require continued adjustment of input parameters. We developed a simplified model for short-term bed need predictions that self-adjusts to changing patterns of disease in the community and admission rates. The model utilizes public health data on community new case counts for SARS-CoV-2 and projects anticipated hospitalization rates. The model was retrospectively evaluated after the second wave of SARS-CoV-2 in New York, New York (October 2020-April 2021) for its accuracy in predicting numbers of coronavirus disease 2019 (COVID-19) admissions 3, 5, 7, and 10 days into the future, comparing predicted admissions with actual admissions for each day at a large integrated health-care delivery network. The mean absolute percent error of the model was found to be low when evaluated across the entire health system, for a single region of the health system or for a single large hospital (6.1%-7.6% for 3-day predictions, 9.2%-10.4% for 5-day predictions, 12.4%-13.2% for 7-day predictions, and 17.1%-17.8% for 10-day predictions).


Asunto(s)
COVID-19 , Humanos , COVID-19/epidemiología , SARS-CoV-2 , Estudios Retrospectivos , Hospitalización , Hospitales
11.
N Engl J Med ; 383(16): 1544-1555, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32722908

RESUMEN

BACKGROUND: Vaccines to prevent coronavirus disease 2019 (Covid-19) are urgently needed. The effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines on viral replication in both upper and lower airways is important to evaluate in nonhuman primates. METHODS: Nonhuman primates received 10 or 100 µg of mRNA-1273, a vaccine encoding the prefusion-stabilized spike protein of SARS-CoV-2, or no vaccine. Antibody and T-cell responses were assessed before upper- and lower-airway challenge with SARS-CoV-2. Active viral replication and viral genomes in bronchoalveolar-lavage (BAL) fluid and nasal swab specimens were assessed by polymerase chain reaction, and histopathological analysis and viral quantification were performed on lung-tissue specimens. RESULTS: The mRNA-1273 vaccine candidate induced antibody levels exceeding those in human convalescent-phase serum, with live-virus reciprocal 50% inhibitory dilution (ID50) geometric mean titers of 501 in the 10-µg dose group and 3481 in the 100-µg dose group. Vaccination induced type 1 helper T-cell (Th1)-biased CD4 T-cell responses and low or undetectable Th2 or CD8 T-cell responses. Viral replication was not detectable in BAL fluid by day 2 after challenge in seven of eight animals in both vaccinated groups. No viral replication was detectable in the nose of any of the eight animals in the 100-µg dose group by day 2 after challenge, and limited inflammation or detectable viral genome or antigen was noted in lungs of animals in either vaccine group. CONCLUSIONS: Vaccination of nonhuman primates with mRNA-1273 induced robust SARS-CoV-2 neutralizing activity, rapid protection in the upper and lower airways, and no pathologic changes in the lung. (Funded by the National Institutes of Health and others.).


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/inmunología , Neumonía Viral/prevención & control , Vacunas Virales/inmunología , Vacuna nCoV-2019 mRNA-1273 , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Betacoronavirus/fisiología , Antígenos CD4 , COVID-19 , Vacunas contra la COVID-19 , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/terapia , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Inmunización Pasiva , Pulmón/patología , Pulmón/virología , Macaca mulatta , Neumonía Viral/patología , Neumonía Viral/terapia , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Linfocitos T/inmunología , Carga Viral , Vacunas Virales/administración & dosificación , Replicación Viral , Sueroterapia para COVID-19
12.
J Gen Intern Med ; 38(10): 2298-2307, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36757667

RESUMEN

BACKGROUND: Non-arrivals to scheduled ambulatory visits are common and lead to a discontinuity of care, poor health outcomes, and increased subsequent healthcare utilization. Reducing non-arrivals is important given their association with poorer health outcomes and cost to health systems. OBJECTIVE: To develop and validate a prediction model for ambulatory non-arrivals. DESIGN: Retrospective cohort study. PATIENTS OR SUBJECTS: Patients at an integrated health system who had an outpatient visit scheduled from January 1, 2020, to February 28, 2022. MAIN MEASURES: Non-arrivals to scheduled appointments. KEY RESULTS: There were over 4.3 million ambulatory appointments from 1.2 million adult patients. Patients with appointment non-arrivals were more likely to be single, racial/ethnic minorities, and not having an established primary care provider compared to those who arrived at their appointments. A prediction model using the XGBoost machine learning algorithm had the highest AUC value (0.768 [0.767-0.770]). Using SHAP values, the most impactful features in the model include rescheduled appointments, lead time (number of days from scheduled to appointment date), appointment provider, number of days since last appointment with the same department, and a patient's prior appointment status within the same department. Scheduling visits close to an appointment date is predicted to be less likely to result in a non-arrival. Overall, the prediction model calibrated well for each department, especially over the operationally relevant probability range of 0 to 40%. Departments with fewer observations and lower non-arrival rates generally had a worse calibration. CONCLUSIONS: Using a machine learning algorithm, we developed a prediction model for non-arrivals to scheduled ambulatory appointments usable for all medical specialties. The proposed prediction model can be deployed within an electronic health system or integrated into other dashboards to reduce non-arrivals. Future work will focus on the implementation and application of the model to reduce non-arrivals.


Asunto(s)
Algoritmos , Citas y Horarios , Adulto , Humanos , Estudios Retrospectivos , Factores de Tiempo , Aprendizaje Automático
13.
PLoS Pathog ; 16(4): e1008456, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32282850

RESUMEN

Leishmania donovani causes visceral leishmaniasis (VL), which is typically fatal without treatment. There is substantial variation between individuals in rates of disease progression, response to treatment and incidence of post-treatment sequelae, specifically post-kala-azar dermal leishmaniasis (PKDL). Nevertheless, the majority of infected people are asymptomatic carriers. Hamsters and mice are commonly used as models of fatal and non-fatal VL, respectively. Host and parasite genetics are likely to be important factors, but in general the reasons for heterogeneous disease presentation in humans and animal models are poorly understood. Host microbiota has become established as a factor in cutaneous forms of leishmaniasis but this has not been studied in VL. We induced intestinal dysbiosis in mice and hamsters by long-term treatment with broad-spectrum antibiotics in their drinking water. There were no significant differences in disease presentation in dysbiotic mice. In contrast, dysbiotic hamsters infected with L. donovani had delayed onset and progression of weight loss. Half of control hamsters had a rapid progression phenotype compared with none of the ABX-treated animals and the nine-month survival rate was significantly improved compared to untreated controls (40% vs. 10%). Antibiotic-treated hamsters also had significantly less severe hepatosplenomegaly, which was accompanied by a distinct cytokine gene expression profile. The protective effect was not explained by differences in parasite loads or haematological profiles. We further found evidence that the gut-liver axis is a key aspect of fatal VL progression in hamsters, including intestinal parasitism, bacterial translocation to the liver, malakoplakia and iron sequestration, none of which occurred in non-progressing murine VL. Diverse bacterial genera were cultured from VL affected livers, of which Rodentibacter was specifically absent from ABX-treated hamsters, indicating this pathobiont may play a role in promoting disease progression. The results provide experimental support for antibiotic prophylaxis against secondary bacterial infections as an adjunct therapy in human VL patients.


Asunto(s)
Antibacterianos/administración & dosificación , Infecciones Bacterianas/prevención & control , Coinfección/prevención & control , Parasitosis Intestinales/parasitología , Leishmaniasis Visceral/parasitología , Animales , Profilaxis Antibiótica , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Fenómenos Fisiológicos Bacterianos , Coinfección/microbiología , Cricetinae , Progresión de la Enfermedad , Femenino , Microbioma Gastrointestinal , Humanos , Leishmania donovani/fisiología , Leishmaniasis Visceral/complicaciones , Masculino , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Simbiosis
14.
PLoS Pathog ; 16(8): e1008793, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32866189

RESUMEN

Transmission to chimpanzees of a precore hepatitis B virus (HBV) mutant implicated in acute liver failure (ALF) in humans did not cause ALF nor the classic form of acute hepatitis B (AHB) seen upon infection with the wild-type HBV strain, but rather a severe AHB with distinct disease features. Here, we investigated the viral and host immunity factors responsible for the unusual severity of AHB associated with the precore HBV mutant in chimpanzees. Archived serial serum and liver specimens from two chimpanzees inoculated with a precore HBV mutant implicated in ALF and two chimpanzees inoculated with wild-type HBV were studied. We used phage-display library and next-generation sequencing (NGS) technologies to characterize the liver antibody response. The results obtained in severe AHB were compared with those in classic AHB and HBV-associated ALF in humans. Severe AHB was characterized by: (i) the highest alanine aminotransferase (ALT) peaks ever seen in HBV transmission studies with a significantly shorter incubation period, compared to classic AHB; (ii) earlier HBsAg clearance and anti-HBs seroconversion with transient or undetectable hepatitis B e antigen (HBeAg); (iii) limited inflammatory reaction relative to hepatocellular damage at the ALT peak with B-cell infiltration, albeit less extensive than in ALF; (iv) detection of intrahepatic germline antibodies against hepatitis B core antigen (HBcAg) by phage-display libraries in the earliest disease phase, as seen in ALF; (v) lack of intrahepatic IgM anti-HBcAg Fab, as seen in classic AHB, but at variance with ALF; and (vi) higher proportion of antibodies in germline configuration detected by NGS in the intrahepatic antibody repertoire compared to classic AHB, but lower than in ALF. This study identifies distinct outcome-specific features associated with severe AHB caused by a precore HBV mutant in chimpanzees, which bear closer resemblance to HBV ALF than to classic AHB. Our data suggest that precore HBV mutants carry an inherently higher pathogenicity that, in addition to specific host factors, may play a critical role in determining the severity of acute HBV disease.


Asunto(s)
Anticuerpos contra la Hepatitis B/metabolismo , Virus de la Hepatitis B/metabolismo , Hepatitis B/metabolismo , Inmunoglobulina M/metabolismo , Fallo Hepático Agudo/metabolismo , Animales , Modelos Animales de Enfermedad , Hepatitis B/patología , Antígenos del Núcleo de la Hepatitis B/metabolismo , Humanos , Fallo Hepático Agudo/patología , Pan troglodytes
15.
J Gen Intern Med ; 36(5): 1214-1221, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33469750

RESUMEN

BACKGROUND: Post-hospital discharge follow-up appointments are intended to evaluate patients' recovery following a hospitalization, but it is unclear how appointment statuses are associated with readmissions. OBJECTIVE: To examine the association between post-discharge ambulatory follow-up status, (1) having a scheduled appointment and (2) arriving to said appointment, and 30-day readmission. DESIGN AND SETTING: A retrospective cohort study of patients hospitalized at 12 hospitals in an Integrated Delivery Network and their ambulatory appointments in that same network. PATIENTS AND MAIN MEASURES: We included 50,772 patients who had an ambulatory appointment within 18 months of an inpatient admission in 2018. Primary outcome was readmission within 30 days post-discharge. KEY RESULTS: There were 32,108 (63.2%) patients with scheduled follow-up appointments and 18,664 (36.8%) patients with no follow-up; 28,313 (88.2%) patients arrived, 3149 (9.8%) missed, and 646 (2.0%) were readmitted prior to their scheduled appointments. Overall 30-day readmission rate was 7.3%; 6.0% [5.75-6.31] for those who arrived, 8.8% [8.44-9.25] for those without follow-up, and 10.3% [9.28-11.40] for those who missed a scheduled appointment (p < 0.001). After adjusting for covariates, patients who arrived at their appointment in the first week following discharge were significantly less likely to be readmitted than those not having any follow-up scheduled (medical adjusted hazard ratio (aHR) 0.57 [0.47-0.69], p < 0.001; surgical aHR 0.58 [0.44-0.75], p < 0.001) There was an increased risk at weeks 3 and 4 for medical patients who arrived at a follow-up compared to those with no follow-up scheduled (week 3 aHR 1.29 [1.10-1.51], p = 0.001; week 4 aHR 1.46 [1.26-1.70], p < 0.001). CONCLUSIONS: The benefit of patients arriving to their post-discharge appointments compared with patients who missed their follow-up visits or had no follow-up scheduled, is only significant during first week post-discharge, suggesting that coordination within 1 week of discharge is critical in reducing 30-day readmissions.


Asunto(s)
Alta del Paciente , Readmisión del Paciente , Cuidados Posteriores , Citas y Horarios , Estudios de Seguimiento , Humanos , Estudios Retrospectivos
16.
Proc Natl Acad Sci U S A ; 115(48): E11369-E11378, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30420516

RESUMEN

Hepatitis B virus (HBV)-associated acute liver failure (ALF) is a dramatic clinical syndrome leading to death or liver transplantation in 80% of cases. Due to the extremely rapid clinical course, the difficulties in obtaining liver specimens, and the lack of an animal model, the pathogenesis of ALF remains largely unknown. Here, we performed a comprehensive genetic and functional characterization of the virus and the host in liver tissue from HBV-associated ALF and compared the results with those of classic acute hepatitis B in chimpanzees. In contrast with acute hepatitis B, HBV strains detected in ALF livers displayed highly mutated HBV core antigen (HBcAg), associated with increased HBcAg expression ex vivo, which was independent of viral replication levels. Combined gene and miRNA expression profiling revealed a dominant B cell disease signature, with extensive intrahepatic production of IgM and IgG in germline configuration exclusively targeting HBcAg with subnanomolar affinities, and complement deposition. Thus, HBV ALF appears to be an anomalous T cell-independent, HBV core-driven B cell disease, which results from the rare and unfortunate encounter between a host with an unusual B cell response and an infecting virus with a highly mutated core antigen.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígenos del Núcleo de la Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Inmunidad Humoral , Fallo Hepático Agudo/inmunología , Adulto , Animales , Linfocitos B/inmunología , Femenino , Hepatitis B/inmunología , Hepatitis B/patología , Hepatitis B/virología , Antígenos del Núcleo de la Hepatitis B/genética , Virus de la Hepatitis B/genética , Humanos , Hígado/inmunología , Hígado/virología , Fallo Hepático Agudo/patología , Fallo Hepático Agudo/virología , Masculino , Persona de Mediana Edad , Pan troglodytes , Linfocitos T/inmunología
17.
PLoS Pathog ; 13(8): e1006565, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28817732

RESUMEN

The Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic betacoronavirus that was first detected in humans in 2012 as a cause of severe acute respiratory disease. As of July 28, 2017, there have been 2,040 confirmed cases with 712 reported deaths. While many infections have been fatal, there have also been a large number of mild or asymptomatic cases discovered through monitoring and contact tracing. New Zealand white rabbits are a possible model for asymptomatic infection with MERS-CoV. In order to discover more about non-lethal infections and to learn whether a single infection with MERS-CoV would protect against reinfection, we inoculated rabbits with MERS-CoV and monitored the antibody and inflammatory response. Following intranasal infection, rabbits developed a transient dose-dependent pulmonary infection with moderately high levels of viral RNA, viral antigen, and perivascular inflammation in multiple lung lobes that was not associated with clinical signs. The rabbits developed antibodies against viral proteins that lacked neutralizing activity and the animals were not protected from reinfection. In fact, reinfection resulted in enhanced pulmonary inflammation, without an associated increase in viral RNA titers. Interestingly, passive transfer of serum from previously infected rabbits to naïve rabbits was associated with enhanced inflammation upon infection. We further found this inflammation was accompanied by increased recruitment of complement proteins compared to primary infection. However, reinfection elicited neutralizing antibodies that protected rabbits from subsequent viral challenge. Our data from the rabbit model suggests that people exposed to MERS-CoV who fail to develop a neutralizing antibody response, or persons whose neutralizing antibody titers have waned, may be at risk for severe lung disease on re-exposure to MERS-CoV.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por Coronavirus/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Inflamación/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Pruebas de Neutralización , Reacción en Cadena de la Polimerasa , Conejos
18.
J Virol ; 91(24)2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29046448

RESUMEN

Influenza viruses of the H1N1, H2N2, and H3N2 subtypes have caused previous pandemics. H2 influenza viruses represent a pandemic threat due to continued circulation in wild birds and limited immunity in the human population. In the event of a pandemic, antiviral agents are the mainstay for treatment, but broadly neutralizing antibodies (bNAbs) may be a viable alternative for short-term prophylaxis or treatment. The hemagglutinin stem binding bNAbs CR6261 and CR9114 have been shown to protect mice from severe disease following challenge with H1N1 and H5N1 and with H1N1, H3N2, and influenza B viruses, respectively. Early studies with CR6261 and CR9114 showed weak in vitro activity against human H2 influenza viruses, but the in vivo efficacy against H2 viruses is unknown. Therefore, we evaluated these antibodies against human- and animal-origin H2 viruses A/Ann Arbor/6/1960 (H2N2) (AA60) and A/swine/MO/4296424/06 (H2N3) (Sw06). In vitro, CR6261 neutralized both H2 viruses, while CR9114 only neutralized Sw06. To evaluate prophylactic efficacy, mice were given CR6261 or CR9114 and intranasally challenged 24 h later with lethal doses of AA60 or Sw06. Both antibodies reduced mortality, weight loss, airway inflammation, and pulmonary viral load. Using engineered bNAb variants, antibody-mediated cell cytotoxicity reporter assays, and Fcγ receptor-deficient (Fcer1g-/-) mice, we show that the in vivo efficacy of CR9114 against AA60 is mediated by Fcγ receptor-dependent mechanisms. Collectively, these findings demonstrate the in vivo efficacy of CR6261 and CR9114 against H2 viruses and emphasize the need for in vivo evaluation of bNAbs.IMPORTANCE bNAbs represent a strategy to prevent or treat infection by a wide range of influenza viruses. The evaluation of these antibodies against H2 viruses is important because H2 viruses caused a pandemic in 1957 and could cross into humans again. We demonstrate that CR6261 and CR9114 are effective against infection with H2 viruses of both human and animal origin in mice, despite the finding that CR9114 did not display in vitro neutralizing activity against the human H2 virus. These findings emphasize the importance of in vivo evaluation and testing of bNAbs.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Subtipo H2N2 del Virus de la Influenza A/inmunología , Gripe Humana/prevención & control , Pruebas de Neutralización/normas , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/genética , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/administración & dosificación , Reacciones Cruzadas , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Subtipo H2N2 del Virus de la Influenza A/patogenicidad , Gripe Humana/inmunología , Ratones , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Receptores de IgG/deficiencia , Receptores de IgG/genética , Receptores de IgG/inmunología
19.
Infect Immun ; 85(7)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28461392

RESUMEN

We studied infection and immunity of hysterectomized mice infected with Chlamydia muridarum and Chlamydia trachomatis to determine if there were differences between these species in their ability to infect vaginal squamous epithelial cells in vivo independently of proximal upper genital tract tissues. We found that C. muridarum readily colonized and infected vaginal squamous epithelial cells, whereas C. trachomatis did not. Primary infection of the vaginal epithelium with C. muridarum produced infections of a duration longer than that reported for normal mice. Infection resulted in an inflammatory response in the vagina characterized by neutrophils and infiltrating submucosal plasma cells consisting primarily of T cells. Despite the delayed clearance, rechallenged C. muridarum-infected mice were highly immune. Mice vaginally infected with C. muridarum produced serum and vaginal wash antibodies and an antigen-specific gamma interferon-dominated Th1-biased T cell response. By comparison, mice vaginally infected with C. trachomatis exhibited transient low-burden infections, produced no detectable tissue inflammatory response, and failed to seroconvert. We discuss how these marked differences in the biology of vaginal infection between these otherwise genetically similar species are possibly linked to pathogen-specific virulence genes and how they may influence pathology and immunity in the upper genital tract.


Asunto(s)
Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Chlamydia muridarum/crecimiento & desarrollo , Chlamydia muridarum/inmunología , Chlamydia trachomatis/crecimiento & desarrollo , Histerectomía , Vagina/microbiología , Animales , Anticuerpos Antibacterianos/análisis , Anticuerpos Antibacterianos/sangre , Infecciones por Chlamydia/inmunología , Femenino , Interferón gamma/metabolismo , Ratones Endogámicos C57BL , Linfocitos T/inmunología
20.
J Infect Dis ; 213(10): 1557-61, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-26941283

RESUMEN

With >1600 documented human infections with Middle East respiratory syndrome coronavirus (MERS-CoV) and a case fatality rate of approximately 36%, medical countermeasures are needed to prevent and limit the disease. We examined the in vivo efficacy of the human monoclonal antibody m336, which has high neutralizing activity against MERS-CoV in vitro. m336 was administered to rabbits intravenously or intranasally before infection with MERS-CoV. Prophylaxis with m336 resulted in a reduction of pulmonary viral RNA titers by 40-9000-fold, compared with an irrelevant control antibody with little to no inflammation or viral antigen detected. This protection in rabbits supports further clinical development of m336.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Infecciones por Coronavirus/prevención & control , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Administración Intranasal , Administración Intravenosa , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/sangre , Infecciones por Coronavirus/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoglobulinas Intravenosas , Pulmón/virología , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , ARN Viral/análisis , Conejos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA