Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 10.295
Filtrar
Más filtros

Publication year range
1.
Cell ; 185(23): 4280-4297.e12, 2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-36323316

RESUMEN

The gut microbiome has an important role in infant health and development. We characterized the fecal microbiome and metabolome of 222 young children in Dhaka, Bangladesh during the first two years of life. A distinct Bifidobacterium longum clade expanded with introduction of solid foods and harbored enzymes for utilizing both breast milk and solid food substrates. The clade was highly prevalent in Bangladesh, present globally (at lower prevalence), and correlated with many other gut taxa and metabolites, indicating an important role in gut ecology. We also found that the B. longum clades and associated metabolites were implicated in childhood diarrhea and early growth, including positive associations between growth measures and B. longum subsp. infantis, indolelactate and N-acetylglutamate. Our data demonstrate geographic, cultural, seasonal, and ecological heterogeneity that should be accounted for when identifying microbiome factors implicated in and potentially benefiting infant development.


Asunto(s)
Bifidobacterium longum , Lactante , Niño , Femenino , Humanos , Preescolar , Bifidobacterium longum/metabolismo , Bifidobacterium/metabolismo , Destete , Oligosacáridos/metabolismo , Bangladesh , Leche Humana , Heces/microbiología
2.
Cell ; 184(17): 4380-4391.e14, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34147139

RESUMEN

Despite the discovery of animal coronaviruses related to SARS-CoV-2, the evolutionary origins of this virus are elusive. We describe a meta-transcriptomic study of 411 bat samples collected from a small geographical region in Yunnan province, China, between May 2019 and November 2020. We identified 24 full-length coronavirus genomes, including four novel SARS-CoV-2-related and three SARS-CoV-related viruses. Rhinolophus pusillus virus RpYN06 was the closest relative of SARS-CoV-2 in most of the genome, although it possessed a more divergent spike gene. The other three SARS-CoV-2-related coronaviruses carried a genetically distinct spike gene that could weakly bind to the hACE2 receptor in vitro. Ecological modeling predicted the co-existence of up to 23 Rhinolophus bat species, with the largest contiguous hotspots extending from South Laos and Vietnam to southern China. Our study highlights the remarkable diversity of bat coronaviruses at the local scale, including close relatives of both SARS-CoV-2 and SARS-CoV.


Asunto(s)
COVID-19/virología , Quirópteros/virología , Coronavirus/genética , Evolución Molecular , SARS-CoV-2/genética , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Asia Sudoriental , China , Coronavirus/clasificación , Coronavirus/aislamiento & purificación , Fenómenos Ecológicos y Ambientales , Genoma Viral , Humanos , Modelos Moleculares , Filogenia , SARS-CoV-2/fisiología , Alineación de Secuencia , Análisis de Secuencia de ARN , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Zoonosis Virales
3.
Cell ; 179(3): 644-658.e13, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31607511

RESUMEN

Rotavirus (RV) encounters intestinal epithelial cells amidst diverse microbiota, opening possibilities of microbes influencing RV infection. Although RV clearance typically requires adaptive immunity, we unintentionally generated RV-resistant immunodeficient mice, which, we hypothesized, reflected select microbes protecting against RV. Accordingly, such RV resistance was transferred by co-housing and fecal transplant. RV-protecting microbiota were interrogated by heat, filtration, and antimicrobial agents, followed by limiting dilution transplant to germ-free mice and microbiome analysis. This approach revealed that segmented filamentous bacteria (SFB) were sufficient to protect mice against RV infection and associated diarrhea. Such protection was independent of previously defined RV-impeding factors, including interferon, IL-17, and IL-22. Colonization of the ileum by SFB induced changes in host gene expression and accelerated epithelial cell turnover. Incubation of RV with SFB-containing feces reduced infectivity in vitro, suggesting direct neutralization of RV. Thus, independent of immune cells, SFB confer protection against certain enteric viral infections and associated diarrheal disease.


Asunto(s)
Inmunidad Adaptativa/genética , Diarrea/microbiología , Mucosa Intestinal/microbiología , Infecciones por Rotavirus/microbiología , Animales , Antiinfecciosos/farmacología , Bacterias/genética , Bacterias/metabolismo , Diarrea/prevención & control , Diarrea/virología , Heces/microbiología , Regulación de la Expresión Génica/genética , Humanos , Íleon/microbiología , Íleon/patología , Íleon/virología , Interferones/genética , Interleucina-17/genética , Interleucinas/genética , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Ratones , Microbiota/genética , Rotavirus/patogenicidad , Infecciones por Rotavirus/prevención & control , Infecciones por Rotavirus/virología , Interleucina-22
4.
Cell ; 173(7): 1742-1754.e17, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29906449

RESUMEN

Osmotic diarrhea is a prevalent condition in humans caused by food intolerance, malabsorption, and widespread laxative use. Here, we assess the resilience of the gut ecosystem to osmotic perturbation at multiple length and timescales using mice as model hosts. Osmotic stress caused reproducible extinction of highly abundant taxa and expansion of less prevalent members in human and mouse microbiotas. Quantitative imaging revealed decimation of the mucus barrier during osmotic perturbation, followed by recovery. The immune system exhibited temporary changes in cytokine levels and a lasting IgG response against commensal bacteria. Increased osmolality prevented growth of commensal strains in vitro, revealing one mechanism contributing to extinction. Environmental availability of microbiota members mitigated extinction events, demonstrating how species reintroduction can affect community resilience. Our findings (1) demonstrate that even mild osmotic diarrhea can cause lasting changes to the microbiota and host and (2) lay the foundation for interventions that increase system-wide resilience.


Asunto(s)
Diarrea/patología , Microbioma Gastrointestinal/efectos de los fármacos , Polietilenglicoles/farmacología , Animales , Bacteroidetes/efectos de los fármacos , Bacteroidetes/genética , Bacteroidetes/aislamiento & purificación , Ciego/química , Ciego/metabolismo , Ciego/microbiología , Ciego/patología , Colon/química , Colon/microbiología , Colon/patología , Citocinas/metabolismo , Diarrea/inmunología , Diarrea/microbiología , Diarrea/veterinaria , Heces/microbiología , Glicósido Hidrolasas/metabolismo , Humanos , Inmunidad Humoral/efectos de los fármacos , Inmunoglobulina G/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Metagenómica , Ratones , Concentración Osmolar , Polietilenglicoles/metabolismo , Proteoma/análisis , ARN Ribosómico 16S/química , ARN Ribosómico 16S/genética , Verrucomicrobia/efectos de los fármacos , Verrucomicrobia/genética , Verrucomicrobia/aislamiento & purificación
5.
Traffic ; 25(8): e12954, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39187475

RESUMEN

Enterocytes and liver cells fulfill important metabolic and barrier functions and are responsible for crucial vectorial secretive and absorptive processes. To date, genetic diseases affecting metabolic enzymes or transmembrane transporters in the intestine and the liver are better comprehended than mutations affecting intracellular trafficking. In this review, we explore the emerging knowledge on intracellular trafficking defects and their clinical manifestations in both the intestine and the liver. We provide a detailed overview including more investigated diseases such as the canonical, variant and associated forms of microvillus inclusion disease, as well as recently described pathologies, highlighting the complexity and disease relevance of several trafficking pathways. We give examples of how intracellular trafficking hubs, such as the apical recycling endosome system, the trans-Golgi network, lysosomes, or the Golgi-to-endoplasmic reticulum transport are involved in the pathomechanism and lead to disease. Ultimately, understanding these processes could spark novel therapeutic approaches, which would greatly improve the quality of life of the affected patients.


Asunto(s)
Hepatopatías , Transporte de Proteínas , Humanos , Hepatopatías/metabolismo , Hepatopatías/genética , Animales , Retículo Endoplásmico/metabolismo , Mucolipidosis/metabolismo , Mucolipidosis/genética , Red trans-Golgi/metabolismo , Síndromes de Malabsorción , Microvellosidades/patología
6.
Proc Natl Acad Sci U S A ; 120(3): e2119409120, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36623190

RESUMEN

Climate-sensitive infectious diseases are an issue of growing concern due to global warming and the related increase in the incidence of extreme weather and climate events. Diarrhea, which is strongly associated with climatic factors, remains among the leading causes of child death globally, disproportionately affecting populations in low- and middle-income countries (LMICs). We use survey data for 51 LMICs between 2000 and 2019 in combination with gridded climate data to estimate the association between precipitation shocks and reported symptoms of diarrheal illness in young children. We account for differences in exposure risk by climate type and explore the modifying role of various social factors. We find that droughts are positively associated with diarrhea in the tropical savanna regions, particularly during the dry season and dry-to-wet and wet-to-dry transition seasons. In the humid subtropical regions, we find that heavy precipitation events are associated with increased risk of diarrhea during the dry season and the transition from dry-to-wet season. Our analysis of effect modifiers highlights certain social vulnerabilities that exacerbate these associations in the two climate zones and present opportunities for public health intervention. For example, we show that stool disposal practices, child feeding practices, and immunizing against the rotavirus modify the association between drought and diarrhea in the tropical savanna regions. In the humid subtropical regions, household's source of water and water disinfection practices modify the association between heavy precipitation and diarrhea. The evidence of effect modification varies depending on the type and duration of the precipitation shock.


Asunto(s)
Clima , Diarrea , Humanos , Niño , Preescolar , Diarrea/epidemiología , Estaciones del Año , Salud Pública , Agua
7.
J Biol Chem ; 300(3): 105779, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38395305

RESUMEN

The newly discovered zoonotic coronavirus swine acute diarrhea syndrome coronavirus (SADS-CoV) causes acute diarrhea, vomiting, dehydration, and high mortality rates in newborn piglets. Although SADS-CoV uses different strategies to evade the host's innate immune system, the specific mechanism(s) by which it blocks the interferon (IFN) response remains unidentified. In this study, the potential of SADS-CoV nonstructural proteins (nsp) to inhibit the IFN response was detected. The results determined that nsp1 was a potent antagonist of IFN response. SADS-CoV nsp1 efficiently inhibited signal transducer and activator of transcription 1 (STAT1) phosphorylation by inducing Janus kinase 1 (JAK1) degradation. Subsequent research revealed that nsp1 induced JAK1 polyubiquitination through K11 and K48 linkages, leading to JAK1 degradation via the ubiquitin-proteasome pathway. Furthermore, SADS-CoV nsp1 induced CREB-binding protein degradation to inhibit IFN-stimulated gene production and STAT1 acetylation, thereby inhibiting STAT1 dephosphorylation and blocking STAT1 transport out of the nucleus to receive antiviral signaling. In summary, the results revealed the novel mechanisms by which SADS-CoV nsp1 blocks the JAK-STAT signaling pathway via the ubiquitin-proteasome pathway. This study yielded valuable findings on the specific mechanism of coronavirus nsp1 in inhibiting the JAK-STAT signaling pathway and the strategies of SADS-CoV in evading the host's innate immune system.


Asunto(s)
Alphacoronavirus , Infecciones por Coronavirus , Complejo de la Endopetidasa Proteasomal , Enfermedades de los Porcinos , Proteínas no Estructurales Virales , Animales , Acetilación , Alphacoronavirus/fisiología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Porcinos , Ubiquitinas/metabolismo , Enfermedades de los Porcinos/metabolismo , Enfermedades de los Porcinos/virología , Células HEK293 , Células Vero , Humanos , Chlorocebus aethiops , Proteínas no Estructurales Virales/metabolismo
8.
J Biol Chem ; 300(4): 107135, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38447796

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is a highly contagious enteric pathogen of the coronavirus family and caused severe economic losses to the global swine industry. Previous studies have established that p53 is a host restriction factor for PEDV infection, and p53 degradation occurs in PEDV-infected cells. However, the underlying molecular mechanisms through which PEDV viral proteins regulate p53 degradation remain unclear. In this study, we found that PEDV infection or expression of the nucleocapsid protein downregulates p53 through a post-translational mechanism: increasing the ubiquitination of p53 and preventing its nuclear translocation. We also show that the PEDV N protein functions by recruiting the E3 ubiquitin ligase COP1 and suppressing COP1 self-ubiquitination and protein degradation, thereby augmenting COP1-mediated degradation of p53. Additionally, COP1 knockdown compromises N-mediated p53 degradation. Functional mapping using truncation analysis showed that the N-terminal domains of N protein were responsible for interacting with COP1 and critical for COP1 stability and p53 degradation. The results presented here suggest the COP1-dependent mechanism for PEDV N protein to abolish p53 activity. This study significantly increases our understanding of PEDV in antagonizing the host antiviral factor p53 and will help initiate novel antiviral strategies against PEDV.


Asunto(s)
Proteínas de la Nucleocápside , Virus de la Diarrea Epidémica Porcina , Proteolisis , Proteína p53 Supresora de Tumor , Ubiquitina-Proteína Ligasas , Ubiquitinación , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Virus de la Diarrea Epidémica Porcina/metabolismo , Animales , Humanos , Proteínas de la Nucleocápside/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Chlorocebus aethiops , Células HEK293 , Porcinos , Células Vero
9.
J Virol ; 98(1): e0162523, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38084960

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is an enteric coronavirus that causes high mortality in piglets, thus posing a serious threat to the world pig industry. Porcine epidemic diarrhea (PED) is related to the imbalance of sodium absorption by small intestinal epithelial cells; however, the etiology of sodium imbalanced diarrhea caused by PEDV remains unclear. Herein, we first proved that PEDV can cause a significant decrease in Na+/H+ exchanger 3 (NHE3) expression on the cell membrane, in a viral dose-dependent manner. Further study showed that the PEDV nucleocapsid (N) protein participates in the regulation of NHE3 activity through interacting with Ezrin. Flame atomic absorption spectroscopy results indicated a serious imbalance in Na+ concentration inside and outside cells following overexpression of PEDV N. Meanwhile, molecular docking technology identified that the small molecule drug Pemetrexed acts on the PEDV N-Ezrin interaction region. It was confirmed that Pemetrexed can alleviate the imbalanced Na+ concentration in IPEC-J2 cells and the diarrhea symptoms of Rongchang pigs caused by PEDV infection. Overall, our data suggest that the interaction between PEDV N and Ezrin reduces the level of phosphorylated Ezrin, resulting in a decrease in the amount of NHE3 protein on the cell membrane. This leads to an imbalance of intracellular and extracellular Na+, which causes diarrhea symptoms in piglets. Pemetrexed is effective in relieving diarrhea caused by PEDV. Our results provide a reference to screen for anti-PEDV targets and to develop drugs to prevent PED.IMPORTANCEPorcine epidemic diarrhea (PED) has caused significant economic losses to the pig industry since its initial outbreak, and the pathogenic mechanism of porcine epidemic diarrhea virus (PEDV) is still under investigation. Herein, we found that the PEDV nucleocapsid protein interacts with Ezrin to regulate Na+/H+ exchanger 3 activity. In addition, we screened out Pemetrexed, a small molecule drug, which can effectively alleviate pig diarrhea caused by PEDV. These results provide support for further exploration of the pathogenesis of PEDV and the development of drugs to prevent PED.


Asunto(s)
Infecciones por Coronavirus , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Animales , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/veterinaria , Diarrea/tratamiento farmacológico , Diarrea/veterinaria , Simulación del Acoplamiento Molecular , Proteínas de la Nucleocápside/metabolismo , Pemetrexed/metabolismo , Virus de la Diarrea Epidémica Porcina/fisiología , Sodio/metabolismo , Intercambiador 3 de Sodio-Hidrógeno/metabolismo , Porcinos , Enfermedades de los Porcinos/tratamiento farmacológico
10.
J Virol ; 98(2): e0203523, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38299844

RESUMEN

Bovine viral diarrhea virus (BVDV) is prevalent worldwide and causes significant economic losses. Gut microbiota is a large microbial community and has a variety of biological functions. However, whether there is a correlation between gut microbiota and BVDV infection and what kind of relation between them have not been reported. Here, we found that gut microbiota composition changed in normal mice after infecting with BVDV, but mainly the low abundance microbe was affected. Interestingly, BVDV infection significantly reduced the diversity of gut microbiota and changed its composition in gut microbiota-dysbiosis mice. Furthermore, compared with normal mice of BVDV infection, there were more viral loads in the duodenum, jejunum, spleen, and liver of the gut microbiota-dysbiosis mice. However, feces microbiota transplantation (FMT) reversed these effects. The data above indicated that the dysbiosis of gut microbiota was a key factor in the high infection rate of BVDV. It is found that the IFN-I signal was involved by investigating the underlying mechanisms. The inhibition of the proliferation and increase in the apoptosis of peripheral blood lymphocytes (PBL) were also observed. However, FMT treatment reversed these changes by regulating PI3K/Akt, ERK, and Caspase-9/Caspase-3 pathways. Furthermore, the involvement of butyrate in the pathogenesis of BVDV was also further confirmed. Our results showed for the first time that gut microbiota acts as a key endogenous defense mechanism against BVDV infection; moreover, targeting regulation of gut microbiota structure and abundance may serve as a new strategy to prevent and control the disease.IMPORTANCEWhether the high infection rate of BVDV is related to gut microbiota has not been reported. In addition, most studies on BVDV focus on in vitro experiments, which limits the study of its prevention and control strategy and its pathogenic mechanism. In this study, we successfully confirmed the causal relationship between gut microbiota and BVDV infection as well as the potential molecular mechanism based on a mouse model of BVDV infection and a mouse model of gut microbiota dysbiosis. Meanwhile, a mouse model which is more susceptible to BVDV provided in this study lays an important foundation for further research on prevention and control strategy of BVDV and its pathogenesis. In addition, the antiviral effect of butyrate, the metabolites of butyrate-producing bacteria, has been further revealed. Overall, our findings provide a promising prevention and control strategy to treat this infectious disease which is distributed worldwide.


Asunto(s)
Diarrea Mucosa Bovina Viral , Virus de la Diarrea Viral Bovina , Microbioma Gastrointestinal , Animales , Bovinos , Ratones , Diarrea Mucosa Bovina Viral/complicaciones , Diarrea Mucosa Bovina Viral/microbiología , Diarrea Mucosa Bovina Viral/terapia , Diarrea Mucosa Bovina Viral/virología , Butiratos/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Diarrea , Virus de la Diarrea Viral Bovina/patogenicidad , Virus de la Diarrea Viral Bovina/fisiología , Disbiosis/complicaciones , Disbiosis/microbiología , Disbiosis/virología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Trasplante de Microbiota Fecal , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Modelos Animales de Enfermedad
11.
J Virol ; 98(4): e0013924, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38501663

RESUMEN

Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a novel porcine enteric coronavirus, and the broad interspecies infection of SADS-CoV poses a potential threat to human health. This study provides experimental evidence to dissect the roles of distinct domains within the SADS-CoV spike S1 subunit in cellular entry. Specifically, we expressed the S1 and its subdomains, S1A and S1B. Cell binding and invasion inhibition assays revealed a preference for the S1B subdomain in binding to the receptors on the cell surface, and this unknown receptor is not utilized by the porcine epidemic diarrhea virus. Nanoparticle display demonstrated hemagglutination of erythrocytes from pigs, humans, and mice, linking the S1A subdomain to the binding of sialic acid (Sia) involved in virus attachment. We successfully rescued GFP-labeled SADS-CoV (rSADS-GFP) from a recombinant cDNA clone to track viral infection. Antisera raised against S1, S1A, or S1B contained highly potent neutralizing antibodies, with anti-S1B showing better efficiency in neutralizing rSADS-GFP infection compared to anti-S1A. Furthermore, depletion of heparan sulfate (HS) by heparinase treatment or pre-incubation of rSADS-GFP with HS or constituent monosaccharides could inhibit SADS-CoV entry. Finally, we demonstrated that active furin cleavage of S glycoprotein and the presence of type II transmembrane serine protease (TMPRSS2) are essential for SADS-CoV infection. These combined observations suggest that the wide cell tropism of SADS-CoV may be related to the distribution of Sia or HS on the cell surface, whereas the S1B contains the main protein receptor binding site. Specific host proteases also play important roles in facilitating SADS-CoV entry.IMPORTANCESwine acute diarrhea syndrome coronavirus (SADS-CoV) is a novel pathogen infecting piglet, and its unique genetic evolution characteristics and broad species tropism suggest the potential for cross-species transmission. The virus enters cells through its spike (S) glycoprotein. In this study, we identify the receptor binding domain on the C-terminal part of the S1 subunit (S1B) of SADS-CoV, whereas the sugar-binding domain located at the S1 N-terminal part of S1 (S1A). Sialic acid, heparan sulfate, and specific host proteases play essential roles in viral attachment and entry. The dissection of SADS-CoV S1 subunit's functional domains and identification of cellular entry cofactors will help to explore the receptors used by SADS-CoV, which may contribute to exploring the mechanisms behind cross-species transmission and host tropism.


Asunto(s)
Alphacoronavirus , Infecciones por Coronavirus , Glicoproteína de la Espiga del Coronavirus , Animales , Humanos , Ratones , Alphacoronavirus/química , Alphacoronavirus/fisiología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Heparitina Sulfato , Ácido N-Acetilneuramínico/metabolismo , Péptido Hidrolasas , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Porcinos
12.
J Virol ; 98(5): e0031724, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38624231

RESUMEN

Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered swine coronavirus with potential cross-species transmission risk. Although SADS-CoV-induced host cell apoptosis and innate immunity antagonization has been revealed, underlying signaling pathways remain obscure. Here, we demonstrated that infection of SADS-CoV induced apoptosis in vivo and in vitro, and that viral protein NS7a is mainly responsible for SADS-CoV-induced apoptosis in host cells. Furthermore, we found that NS7a interacted with apoptosis-inducing factor mitochondria associated 1 (AIFM1) to activate caspase-3 via caspase-6 in SADS-CoV-infected cells, and enhanced SADS-CoV replication. Importantly, NS7a suppressed poly(I:C)-induced expression of type III interferon (IFN-λ) via activating caspase-3 to cleave interferon regulatory factor 3 (IRF3), and caspase-3 inhibitor protects piglets against SADS-CoV infection in vivo. These findings reveal how SADS-CoV induced apoptosis to inhibit innate immunity and provide a valuable clue to the development of effective drugs for the clinical control of SADS-CoV infection.IMPORTANCEOver the last 20 years, multiple animal-originated coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV), middle east respiratory syndrome coronavirus (MERS-CoV), and SARS-CoV-2, have caused millions of deaths, seriously jeopardized human health, and hindered social development, indicating that the study of animal-originated coronaviruses with potential for cross-species transmission is particularly important. Bat-originated swine acute diarrhea syndrome coronavirus (SADS-CoV), discovered in 2017, can not only cause fatal diarrhea in piglets, but also infect multiple human cells, with a potential risk of cross-species transmission, but its pathogenesis is unclear. In this study, we demonstrated that NS7a of SADS-CoV suppresses IFN-λ production via apoptosis-inducing factor mitochondria associated 1 (AIFM1)-caspase-6-caspase-3-interferon regulatory factor 3 (IRF3) pathway, and caspase-3 inhibitor (Z-DEVD-FMK) can effectively inhibit SADS-CoV replication and protect infected piglets. Our findings in this study contribute to a better understanding of SADS-CoV-host interactions as a part of the coronaviruses pathogenesis and using apoptosis-inhibitor as a drug as potential therapeutic approaches for prevention and control of SADS-CoV infection.


Asunto(s)
Apoptosis , Inmunidad Innata , Factor 3 Regulador del Interferón , Interferones , Proteínas no Estructurales Virales , Animales , Porcinos , Humanos , Interferones/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Interferón lambda , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/metabolismo , Alphacoronavirus/metabolismo , Caspasa 3/metabolismo , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/metabolismo , Células Vero , Transducción de Señal , Chlorocebus aethiops , Células HEK293
13.
Proc Natl Acad Sci U S A ; 119(18): e2118126119, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35476513

RESUMEN

Zoonotic transmission of coronaviruses poses an ongoing threat to human populations. Endemic outbreaks of swine acute diarrhea syndrome coronavirus (SADS-CoV) have caused severe economic losses in the pig industry and have the potential to cause human outbreaks. Currently, there are no vaccines or specific antivirals against SADS-CoV, and our limited understanding of SADS-CoV host entry factors could hinder prompt responses to a potential human outbreak. Using a genomewide CRISPR knockout screen, we identified placenta-associated 8 protein (PLAC8) as an essential host factor for SADS-CoV infection. Knockout of PLAC8 abolished SADS-CoV infection, which was restored by complementing PLAC8 from multiple species, including human, rhesus macaques, mouse, pig, pangolin, and bat, suggesting a conserved infection pathway and susceptibility of SADS-CoV among mammals. Mechanistically, PLAC8 knockout does not affect viral entry; rather, knockout cells displayed a delay and reduction in viral subgenomic RNA expression. In a swine primary intestinal epithelial culture (IEC) infection model, differentiated cultures have high levels of PLAC8 expression and support SADS-CoV replication. In contrast, expanding IECs have low levels of PLAC8 expression and are resistant to SADS-CoV infection. PLAC8 expression patterns translate in vivo; the immunohistochemistry of swine ileal tissue revealed high levels of PLAC8 protein in neonatal compared to adult tissue, mirroring the known SADS-CoV pathogenesis in neonatal piglets. Overall, PLAC8 is an essential factor for SADS-CoV infection and may serve as a promising target for antiviral development for potential pandemic SADS-CoV.


Asunto(s)
Alphacoronavirus , Infecciones por Coronavirus , Enfermedades de los Porcinos , Alphacoronavirus/genética , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Infecciones por Coronavirus/epidemiología , Porcinos
14.
J Infect Dis ; 229(4): 988-998, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-37405406

RESUMEN

BACKGROUND: Bacterial pathogens cause substantial diarrhea morbidity and mortality among children living in endemic settings, yet antimicrobial treatment is only recommended for dysentery or suspected cholera. METHODS: AntiBiotics for Children with severe Diarrhea was a 7-country, placebo-controlled, double-blind efficacy trial of azithromycin in children 2-23 months of age with watery diarrhea accompanied by dehydration or malnutrition. We tested fecal samples for enteric pathogens utilizing quantitative polymerase chain reaction to identify likely and possible bacterial etiologies and employed pathogen-specific cutoffs based on genomic target quantity in previous case-control diarrhea etiology studies to identify likely and possible bacterial etiologies. RESULTS: Among 6692 children, the leading likely etiologies were rotavirus (21.1%), enterotoxigenic Escherichia coli encoding heat-stable toxin (13.3%), Shigella (12.6%), and Cryptosporidium (9.6%). More than one-quarter (1894 [28.3%]) had a likely and 1153 (17.3%) a possible bacterial etiology. Day 3 diarrhea was less common in those randomized to azithromycin versus placebo among children with a likely bacterial etiology (risk difference [RD]likely, -11.6 [95% confidence interval {CI}, -15.6 to -7.6]) and possible bacterial etiology (RDpossible, -8.7 [95% CI, -13.0 to -4.4]) but not in other children (RDunlikely, -0.3% [95% CI, -2.9% to 2.3%]). A similar association was observed for 90-day hospitalization or death (RDlikely, -3.1 [95% CI, -5.3 to -1.0]; RDpossible, -2.3 [95% CI, -4.5 to -.01]; RDunlikely, -0.6 [95% CI, -1.9 to .6]). The magnitude of risk differences was similar among specific likely bacterial etiologies, including Shigella. CONCLUSIONS: Acute watery diarrhea confirmed or presumed to be of bacterial etiology may benefit from azithromycin treatment. CLINICAL TRIALS REGISTRATION: NCT03130114.


Asunto(s)
Infecciones Bacterianas , Criptosporidiosis , Cryptosporidium , Disentería , Shigella , Niño , Humanos , Lactante , Antibacterianos/uso terapéutico , Azitromicina/uso terapéutico , Criptosporidiosis/tratamiento farmacológico , Patología Molecular , Diarrea/epidemiología , Infecciones Bacterianas/tratamiento farmacológico , Bacterias , Disentería/complicaciones , Disentería/tratamiento farmacológico
15.
J Infect Dis ; 230(1): 239-249, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052715

RESUMEN

BACKGROUND: Macrolide antibiotics, including azithromycin, can reduce under 5 years of age mortality rates and treat various infections in children in sub-Saharan Africa. These exposures, however, can select for antibiotic-resistant bacteria in the gut microbiota. METHODS: Our previous randomized controlled trial (RCT) of a rapid-test-and-treat strategy for severe acute diarrheal disease in children in Botswana included an intervention (3-day azithromycin dose) group and a control group that received supportive treatment. In this prospective matched cohort study using stools collected at baseline and 60 days after treatment from RCT participants, the collection of antibiotic resistance genes or resistome was compared between groups. RESULTS: Certain macrolide resistance genes increased in prevalence by 13%-55% at 60 days, without differences in gene presence between the intervention and control groups. These genes were linked to tetracycline resistance genes and mobile genetic elements. CONCLUSIONS: Azithromycin treatment for bacterial diarrhea for young children in Botswana resulted in similar effects on the gut resistome as the supportive treatment and did not provide additional selective pressure for macrolide resistance gene maintenance. The gut microbiota of these children contains diverse macrolide resistance genes that may be transferred within the gut upon repeated exposures to azithromycin or coselected by other antibiotics. CLINICAL TRIALS REGISTRATION: NCT02803827.


Asunto(s)
Antibacterianos , Azitromicina , Diarrea , Microbioma Gastrointestinal , Humanos , Azitromicina/uso terapéutico , Azitromicina/administración & dosificación , Botswana , Diarrea/microbiología , Diarrea/tratamiento farmacológico , Antibacterianos/uso terapéutico , Antibacterianos/farmacología , Preescolar , Lactante , Estudios Prospectivos , Femenino , Masculino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Farmacorresistencia Bacteriana/genética , Heces/microbiología , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/clasificación , Bacterias/aislamiento & purificación
16.
J Infect Dis ; 230(1): e75-e79, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052701

RESUMEN

To evaluate how breakthrough rotavirus disease contributes to transmission, we examined the impact of rotavirus vaccination on fecal shedding and duration of illness. We used multivariable linear regression to analyze rotavirus quantity by RT-qPCR and duration among 184 episodes of rotavirus diarrhea positive by ELISA in the PROVIDE study. Vaccinated children had less fecal viral shedding compared to unvaccinated children (mean difference = -0.59 log copies per gram of stool; 95% confidence interval [CI], -.99 to -.19). Duration of illness was on average 0.47 days (95% CI, -.23 to 1.17 days) shorter among vaccinated children. Rotarix vaccination reduces shedding burden among breakthrough cases of rotavirus gastroenteritis. Clinical Trials Registration . NCT01375647.


Asunto(s)
Heces , Infecciones por Rotavirus , Vacunas contra Rotavirus , Rotavirus , Vacunas Atenuadas , Esparcimiento de Virus , Humanos , Vacunas contra Rotavirus/administración & dosificación , Vacunas contra Rotavirus/inmunología , Infecciones por Rotavirus/prevención & control , Infecciones por Rotavirus/epidemiología , Infecciones por Rotavirus/virología , Lactante , Bangladesh/epidemiología , Rotavirus/inmunología , Heces/virología , Femenino , Masculino , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Gastroenteritis/virología , Gastroenteritis/prevención & control , Gastroenteritis/epidemiología , Vacunación , Diarrea/virología , Diarrea/prevención & control , Diarrea/epidemiología , Administración Oral
17.
J Infect Dis ; 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39110032

RESUMEN

Attributing infectious causes of diarrhea is critical to inform treatment and burden estimates. The attributable fraction (AF) approach based on the association between pathogen quantity and diarrhea has been frequently used but may underestimate incidence. We leveraged data from the multisite birth-cohort Malnutrition and Enteric Disease (MAL-ED) Study, where diarrheal and non-diarrheal stools were collected from 1,715 children from 0-2 years. We compared attribution using a longitudinal AF (LAF) method that considers the temporal association between pathogen quantity and diarrhea symptoms to previously-published AF estimates. For rotavirus and Shigella, attribution did not meaningfully change. For others like adenovirus 40 & 41, astrovirus, norovirus GII, sapovirus, Campylobacter jejuni or C coli, ST ETEC, typical EPEC, and Cryptosporidium, attribution increased, demonstrating longitudinal data may be informative for pathogens with weak associations between quantity and diarrhea. We further derived accuracy-based, pathogen-specific quantity cut-offs that may improve attribution in the absence of longitudinal data.

18.
J Infect Dis ; 229(2): 367-375, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-37531657

RESUMEN

BACKGROUND: This phase 2 extension explored the long-term antibody persistence of an investigational Clostridioides difficile vaccine and the safety, tolerability, and immunogenicity of dose 4 approximately 12 months post-dose 3. METHODS: One year post-dose 3, healthy US 65- to 85-year-olds (N = 300) were randomized to dose 4 of vaccine at previously received antigen levels (100 or 200 µg) or placebo. Assessments included safety and percentages of participants achieving neutralizing antibody titers above prespecified thresholds (≥219 and ≥2586 neutralization units/mL for toxins A and B, respectively). RESULTS: In participants previously given three 200-µg doses and placebo in the extension, toxin A and B neutralizing antibodies were above prevaccination levels 48 months post-dose 3 (36 months after placebo); 24.0% and 26.0% had toxin A and B antibodies at or above prespecified thresholds, respectively. Neutralizing antibodies increased post-dose 4 (12 months post-dose 3) and persisted to 36 months post-dose 4. Thirty days post-dose 4, all participants had toxin A and 86.5% to 100% had toxin B titers at or above prespecified thresholds. Local reactions were more frequent in vaccine recipients. Systemic and adverse event frequencies were similar across groups. CONCLUSIONS: C difficile vaccine immune responses persisted 48 months post-dose 3. Dose 4 was immunogenic and well tolerated, supporting continued development. Clinical Trials Registration. ClinicalTrials.gov NCT02561195.


Asunto(s)
Clostridioides difficile , Adulto , Humanos , Vacunas Bacterianas , Anticuerpos Neutralizantes , Anticuerpos Antibacterianos , Formación de Anticuerpos , Inmunogenicidad Vacunal , Anticuerpos Antivirales , Método Doble Ciego
19.
Am J Physiol Cell Physiol ; 326(3): C829-C842, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38223928

RESUMEN

Loss of function mutations in the SLC26A3 gene cause chloride-losing diarrhea in mice and humans. Although systemic adaptive changes have been documented in these patients and in the corresponding knockout mice, how colonic enterocytes adapt to loss of this highly expressed and highly regulated luminal membrane anion exchanger remains unclear. To address this question, SLC26A3 was deleted in the self-differentiating Caco2BBe colonic cell line by the CRISPR/Cas9 technique. We selected a clone with loss of SLC26A3 protein expression and morphological features indistinguishable from those of the native cell line. Neither growth curves nor development of transepithelial electrical resistance (TEER) differed between wild-type (WT) and SLC26A3 knockout (KO) cells. Real-time qPCR and Western analysis in SLC26A3-KO cells revealed an increase in AE2 expression without significant change in NHE3 expression or localization. Steady-state pHi and apical and basolateral Cl-/HCO3- exchange activities were assessed fluorometrically in a dual perfusion chamber with independent perfusion of luminal and serosal baths. Apical Cl-/HCO3- exchange rates were strongly reduced in SLC26A3-KO cells, accompanied by a surface pH more acidic than that of WT cells. Steady-state pHi was not significantly different from that of WT cells, but basolateral Cl-/HCO3- exchange rates were higher in SLC26A3-KO than in WT cells. The data show that CRISPR/Cas9-mediated SLC26A3 deletion strongly reduced apical Cl-/HCO3- exchange rate and apical surface pH, but sustained a normal steady-state pHi due to increased expression and function of basolateral AE2. The low apical surface pH resulted in functional inhibition of NHE-mediated fluid absorption despite normal expression of NHE3 polypeptide.NEW & NOTEWORTHY SLC26A3 gene mutations cause chloride-losing diarrhea. To understand how colonic enterocytes adapt, SLC26A3 was deleted in Caco2BBe cells using CRISPR/Cas9. In comparison to the wild-type cells, SLC26A3 knockout cells showed similar growth and transepithelial resistance but substantially reduced apical Cl-/HCO3- exchange rates, and an acidic surface pH. Steady-state intracellular pH was comparable between the WT and KO cells due to increased basolateral AE2 expression and function.


Asunto(s)
Cloruros , Diarrea , Humanos , Animales , Ratones , Intercambiador 3 de Sodio-Hidrógeno/genética , Aniones , Enterocitos , Concentración de Iones de Hidrógeno , Transportadores de Sulfato/genética , Antiportadores de Cloruro-Bicarbonato/genética
20.
Clin Infect Dis ; 78(3): 573-581, 2024 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-38097379

RESUMEN

BACKGROUND: Multiplex molecular diagnostic panels have greatly enhanced detection of gastrointestinal pathogens. However, data on the impact of these tests on clinical and patient-centered outcomes are limited. METHODS: We conducted a prospective, multicenter, stepped-wedge trial to determine the impact of multiplex molecular testing at 5 academic children's hospitals on children presenting to the emergency department with acute gastroenteritis. Caregivers were interviewed on enrollment and 7-10 days after enrollment to determine symptoms, risk factors, subsequent medical visits, and impact on family members. During the pre-intervention period, diagnostic testing was performed at the clinician's discretion . During the intervention period, multiplex molecular testing was performed on all children, with results available to clinicians. The primary outcome was return visits to a healthcare provider within 10 days of enrollment. RESULTS: Potential pathogens were identified by clinician-ordered tests in 19 of 571 (3.3%) in the pre-intervention period compared with 434 of 586 (74%) in the intervention period; clinically relevant pathogens were detected in 2.1% and 15%, respectively. In the multivariate model, the intervention was associated with a 21% reduction in the odds of any return visit (odds ratio, 0.79; 95% confidence interval, .70-.90) after adjusting for potential confounders. Appropriate treatment was prescribed in 11.3% compared with 19.6% during the intervention period (P = .22). CONCLUSIONS: Routine molecular multiplex testing for all children who presented to the ED with acute gastroenteritis detected more clinically relevant pathogens and led to a 21% decrease in return visits. Additional research is needed to define patients most likely to benefit from testing. Clinical Trials Registration. NCT02248285.


Asunto(s)
Gastroenteritis , Niño , Humanos , Servicio de Urgencia en Hospital , Gastroenteritis/diagnóstico , Gastroenteritis/tratamiento farmacológico , Técnicas de Diagnóstico Molecular/métodos , Estudios Prospectivos , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda