Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(24): 5916-5931.e17, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34767757

RESUMEN

There is increasing interest in the potential contribution of the gut microbiome to autism spectrum disorder (ASD). However, previous studies have been underpowered and have not been designed to address potential confounding factors in a comprehensive way. We performed a large autism stool metagenomics study (n = 247) based on participants from the Australian Autism Biobank and the Queensland Twin Adolescent Brain project. We found negligible direct associations between ASD diagnosis and the gut microbiome. Instead, our data support a model whereby ASD-related restricted interests are associated with less-diverse diet, and in turn reduced microbial taxonomic diversity and looser stool consistency. In contrast to ASD diagnosis, our dataset was well powered to detect microbiome associations with traits such as age, dietary intake, and stool consistency. Overall, microbiome differences in ASD may reflect dietary preferences that relate to diagnostic features, and we caution against claims that the microbiome has a driving role in ASD.


Asunto(s)
Trastorno Autístico/microbiología , Conducta Alimentaria , Microbioma Gastrointestinal , Adolescente , Factores de Edad , Trastorno Autístico/diagnóstico , Conducta , Niño , Preescolar , Heces/microbiología , Femenino , Humanos , Masculino , Fenotipo , Filogenia , Especificidad de la Especie
3.
Nature ; 618(7965): 590-597, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37258672

RESUMEN

Rapidly evolving influenza A viruses (IAVs) and influenza B viruses (IBVs) are major causes of recurrent lower respiratory tract infections. Current influenza vaccines elicit antibodies predominantly to the highly variable head region of haemagglutinin and their effectiveness is limited by viral drift1 and suboptimal immune responses2. Here we describe a neuraminidase-targeting monoclonal antibody, FNI9, that potently inhibits the enzymatic activity of all group 1 and group 2 IAVs, as well as Victoria/2/87-like, Yamagata/16/88-like and ancestral IBVs. FNI9 broadly neutralizes seasonal IAVs and IBVs, including the immune-evading H3N2 strains bearing an N-glycan at position 245, and shows synergistic activity when combined with anti-haemagglutinin stem-directed antibodies. Structural analysis reveals that D107 in the FNI9 heavy chain complementarity-determinant region 3 mimics the interaction of the sialic acid carboxyl group with the three highly conserved arginine residues (R118, R292 and R371) of the neuraminidase catalytic site. FNI9 demonstrates potent prophylactic activity against lethal IAV and IBV infections in mice. The unprecedented breadth and potency of the FNI9 monoclonal antibody supports its development for the prevention of influenza illness by seasonal and pandemic viruses.


Asunto(s)
Anticuerpos Antivirales , Especificidad de Anticuerpos , Virus de la Influenza A , Virus de la Influenza B , Vacunas contra la Influenza , Gripe Humana , Imitación Molecular , Neuraminidasa , Animales , Humanos , Ratones , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/química , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Especificidad de Anticuerpos/inmunología , Arginina/química , Dominio Catalítico , Hemaglutininas Virales/inmunología , Virus de la Influenza A/clasificación , Virus de la Influenza A/enzimología , Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/enzimología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Virus de la Influenza B/clasificación , Virus de la Influenza B/enzimología , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/química , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/inmunología , Gripe Humana/prevención & control , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/química , Neuraminidasa/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Estaciones del Año , Ácidos Siálicos/química
5.
Infection ; 51(2): 475-482, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36201152

RESUMEN

PURPOSE: This case series describes real-world utilization of cefiderocol and associated clinical outcomes in the setting of carbapenem-resistant Gram-negative bacterial infections. METHODS: Adult hospitalized patients administered at least 5 days of cefiderocol as definitive treatment from October 1, 2020 to September 16, 2021 were included in this retrospective cohort analysis. The primary outcome was clinical success defined as a composite of 30 day survival, resolution of infection, and absence of 30 day recurrence of the same organism. RESULTS: Among 24 patients, pneumonia (19, 79%) was the most common source of infection with Acinetobacter baumannii (14, 58%) and P. aeruginosa (10, 42%) as the predominant organisms isolated. Cefiderocol monotherapy was used as definitive treatment in 16 (67%) patients. Eleven patients (46%) met clinical success. Thirty-day mortality occurred in ten (42%) patients while seven (29%) patients had recurrence of infection. Thirteen out of 21 total isolates (62%) tested for susceptibility were deemed susceptible. Of the 16 patients with available susceptibility, 9 (56%) had an infection where all isolated organisms were susceptible to cefiderocol. CONCLUSIONS: Our results provide additional insight into the in vivo activity of cefiderocol. Cefiderocol remains a salvage option for carbapenem-resistant Gram-negative organisms.


Asunto(s)
Antibacterianos , Carbapenémicos , Adulto , Humanos , Carbapenémicos/farmacología , Carbapenémicos/uso terapéutico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Terapia Recuperativa , Estudios Retrospectivos , Bacterias Gramnegativas , Farmacorresistencia Bacteriana Múltiple , Cefalosporinas/farmacología , Cefalosporinas/uso terapéutico , Pruebas de Sensibilidad Microbiana , Cefiderocol
6.
Am J Emerg Med ; 71: 123-128, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37390608

RESUMEN

INTRO: Influenza shares common symptoms with bacterial pneumonia, which may result in unnecessary antibiotic prescriptions in the emergency department (ED) when the diagnosis is unknown. Rapid influenza polymerase chain reaction (PCR) tests have reduced turnaround times compared to standard multiplex PCR respiratory panels allowing for earlier diagnosis, which may improve antimicrobial stewardship outcomes in the ED. This study aims to compare antibiotic and antiviral use before and after deployment of the rapid influenza PCR in the ED. METHODS: This single-center, retrospective, cohort study included pediatric and adult patients discharged from the ED with a positive influenza test using a standard multiplex PCR respiratory panel (January 2017 - July 2019) or rapid PCR (July 2019 - February 2020). The primary endpoint was number of antibiotic prescriptions pre- and post-implementation of the rapid influenza PCR in the ED. Secondary endpoints included number of antiviral prescriptions, duration of antimicrobial therapy, test turnaround time, ED length of stay, 30-day readmission, and adverse events. A multivariable logistic regression evaluated patient factors associated with antimicrobial prescribing. RESULTS: A total of 620 positive influenza results were identified with 280 patients (standard multiplex PCR = 33; rapid PCR = 247) meeting inclusion criteria. Patients were less likely to be prescribed antibiotics (39.4% vs 8.9%, OR 0.15, 95% CI 0.067-0.34) and more likely to be prescribed antivirals (24.2% vs 61.1%, OR 4.92, 95% CI 2.13-11.34) with the rapid influenza PCR. Rapid influenza PCR significantly reduced ED length of stay (4.9 vs 3.4 h, p < 0.01) and test turnaround time (27 h vs 3.5 h, p < 0.01). Patients at high risk for complications associated with influenza were more likely to be prescribed antiviral therapy (22.7% vs 67.8%, OR 7.16, 95% CI 2.52-20.40). Based on the regression analysis conducted, asthma, (OR 3.5, 95% CI 1.48-8.26), immunosuppression (OR 9.6, 95% CI 1.18-78.2), and age <5 years old (OR 3.1, 95% CI 1.80-5.45) were predictors of antiviral prescribing. CONCLUSION: Implementation of a rapid influenza PCR in the ED reduced antibiotic use and optimized antiviral therapy for patients with influenza including those at higher risk of complications.


Asunto(s)
Antiinfecciosos , Gripe Humana , Adulto , Humanos , Niño , Preescolar , Gripe Humana/diagnóstico , Gripe Humana/tratamiento farmacológico , Estudios Retrospectivos , Estudios de Cohortes , Antiinfecciosos/uso terapéutico , Antibacterianos/uso terapéutico , Antivirales/uso terapéutico , Reacción en Cadena de la Polimerasa Multiplex/métodos , Servicio de Urgencia en Hospital
7.
Emerg Infect Dis ; 26(5): 876-880, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32310077

RESUMEN

Candida auris is an emerging fungal pathogen that is typically resistant to fluconazole and is known to cause healthcare-associated outbreaks. We retrospectively reviewed 28 patients who had >1 positive culture for C. auris within a multisite health system in Illinois, USA, during May 2018-April 2019. Twelve of these patients were treated as inpatients for C. auris infections; 10 (83%) met criteria for clinical success, defined as absence of all-cause mortality, C. auris recurrence, and infection-related readmission at 30 days from the first positive culture. The other 2 patients (17%) died within 30 days. Most patients (92%) were empirically treated with micafungin. Four (14%) of 28 total isolates were resistant to fluconazole, 1 (3.6%) was resistant to amphotericin B, and 1 (3.6%) was resistant to echinocandins. Our findings describe low rates of antifungal resistance and favorable clinical outcomes for most C. auris patients.


Asunto(s)
Antifúngicos , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Candidiasis Invasiva , Humanos , Illinois/epidemiología , Pruebas de Sensibilidad Microbiana , Estudios Retrospectivos
8.
Antimicrob Agents Chemother ; 64(11)2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32839223

RESUMEN

Fluoroquinolones (FQs) are often preferred as oral step-down therapy for bloodstream infections (BSIs) due to favorable pharmacokinetic parameters; however, they are also associated with serious adverse events. The objective of this study was to compare clinical outcomes for patients who received an oral FQ versus an oral beta-lactam (BL) as step-down therapy for uncomplicated streptococcal BSIs. This multicenter, retrospective cohort study analyzed adult patients who completed therapy with an oral FQ or BL with at least one blood culture positive for a Streptococcus species from 1 January 2014 to 30 June 2019. The primary outcome was clinical success, defined as the lack of all-cause mortality, recurrent BSI with the same organism, and infection-related readmission at 90 days. A multivariable logistic regression model for predictors of clinical failure was conducted. A total of 220 patients were included, with 87 (40%) receiving an FQ and 133 (60%) receiving a BL. Step-down therapy with an oral BL was noninferior to an oral FQ (93.2% versus 92.0%; mean difference, 1.2%; 90% confidence interval [CI], -5.2 to 7.8). No differences were seen in 90-day mortality, 90-day recurrent BSI, 90-day infection-related readmission, or 90-day incidence of Clostridioides difficile-associated diarrhea. Predictors of clinical failure included oral step-down transition before day 3 (odds ratio [OR] = 5.18; 95% CI, 1.21, 22.16) and low-dose oral step-down therapy (OR = 2.74; 95% CI, 0.95, 7.90). Our results suggest that oral step-down therapy for uncomplicated streptococcal BSI with a BL is noninferior to an FQ.


Asunto(s)
Bacteriemia , Sepsis , Adulto , Antibacterianos/uso terapéutico , Bacteriemia/tratamiento farmacológico , Fluoroquinolonas/uso terapéutico , Humanos , Estudios Retrospectivos , Sepsis/tratamiento farmacológico , Streptococcus , beta-Lactamas/uso terapéutico
9.
Am J Emerg Med ; 38(6): 1153-1158, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31495521

RESUMEN

OBJECTIVES: Traditional antibiograms use local resistance patterns and susceptibility data to guide empiric antimicrobial therapy selection. However, antibiograms are rarely unit-specific and do not account for patient-specific risk factors. METHODS: This retrospective, single-center descriptive study used culture and susceptibility data from January 1 to December 31, 2016 to develop an Emergency Department (ED)-specific antibiogram and compare the antimicrobial susceptibilities of the most commonly identified organisms to the hospital antibiogram. All ED isolates were further stratified by the following risk factors that may influence antimicrobial susceptibility: age, disposition from ED, previous antimicrobial use and/or hospitalization within 30 days, and presenting location (i.e. healthcare facility residence versus community). RESULTS: A total of 2158 isolates from the ED were included: Escherichia coli (n = 1244), Klebsiella pneumoniae (n = 232), Proteus mirabilis (n = 131), Pseudomonas aeruginosa (n = 103), Staphylococcus aureus (n = 303), and Enterococcus faecalis (n = 145). There were no statistically significant differences between the ED and hospital antibiogram (n = 5739) with the exception of Escherichia coli. The hospital antibiogram overestimated Escherichia coli resistance rates for cefazolin (20% vs 15.6%, p = 0.049), ceftriaxone (9.6% vs 6.4%, p < 0.033), and ciprofloxacin (23.7% vs 15.4%, p < 0.006). There were significantly more risk factors present in patients admitted versus discharged from the ED (p < 0.001). Healthcare facility residence had the greatest influence on susceptibility, especially Escherichia coli (81.8% vs 34.9%, p < 0.001) and Proteus mirabilis (75.3% vs 33%, p < 0.001) ciprofloxacin susceptibility. CONCLUSIONS: There were no statistically significant differences between the ED and hospital antibiogram with the exception of Escherichia coli. However, development of an ED-specific antibiogram can aid physicians in prescribing appropriate empiric therapy when risk factors are included.


Asunto(s)
Antibacterianos/uso terapéutico , Bacterias/aislamiento & purificación , Infecciones Bacterianas/tratamiento farmacológico , Servicio de Urgencia en Hospital , Hospitalización/estadística & datos numéricos , Medición de Riesgo/métodos , Anciano , Bacterias/efectos de los fármacos , Infecciones Bacterianas/epidemiología , Infecciones Bacterianas/microbiología , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Masculino , Pruebas de Sensibilidad Microbiana , Estudios Retrospectivos , Factores de Riesgo , Estados Unidos/epidemiología
10.
PLoS Pathog ; 12(5): e1005652, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27191591

RESUMEN

The interaction of Mycobacterium tuberculosis (Mtb) with host cell death signaling pathways is characterized by an initial anti-apoptotic phase followed by a pro-necrotic phase to allow for host cell exit of the bacteria. The bacterial modulators regulating necrosis induction are poorly understood. Here we describe the identification of a transcriptional repressor, Rv3167c responsible for regulating the escape of Mtb from the phagosome. Increased cytosolic localization of MtbΔRv3167c was accompanied by elevated levels of mitochondrial reactive oxygen species and reduced activation of the protein kinase Akt, and these events were critical for the induction of host cell necrosis and macroautophagy. The increase in necrosis led to an increase in bacterial virulence as reflected in higher bacterial burden and reduced survival of mice infected with MtbΔRv3167c. The regulon of Rv3167c thus contains the bacterial mediators involved in escape from the phagosome and host cell necrosis induction, both of which are crucial steps in the intracellular lifecycle and virulence of Mtb.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/patogenicidad , Factores de Transcripción/metabolismo , Tuberculosis/metabolismo , Virulencia/fisiología , Animales , Modelos Animales de Enfermedad , Cobayas , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa , Factores de Virulencia/metabolismo
12.
PLoS Pathog ; 11(5): e1004855, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25974076

RESUMEN

Malaria, caused by Plasmodium parasite infection, continues to be one of the leading causes of worldwide morbidity and mortality. Development of an effective vaccine has been encumbered by the complex life cycle of the parasite that has distinct pre-erythrocytic and erythrocytic stages of infection in the mammalian host. Historically, malaria vaccine development efforts have targeted each stage in isolation. An ideal vaccine, however, would target multiple life cycle stages with multiple arms of the immune system and be capable of eliminating initial infection in the liver, the subsequent blood stage infection, and would prevent further parasite transmission. We have previously shown that immunization of mice with Plasmodium yoelii genetically attenuated parasites (GAP) that arrest late in liver stage development elicits stage-transcending protection against both a sporozoite challenge and a direct blood stage challenge. Here, we show that this immunization strategy engenders both T- and B-cell responses that are essential for stage-transcending protection, but the relative importance of each is determined by the host genetic background. Furthermore, potent anti-blood stage antibodies elicited after GAP immunization rely heavily on FC-mediated functions including complement fixation and FC receptor binding. These protective antibodies recognize the merozoite surface but do not appear to recognize the immunodominant merozoite surface protein-1. The antigen(s) targeted by stage-transcending immunity are present in both the late liver stages and blood stage parasites. The data clearly show that GAP-engendered protective immune responses can target shared antigens of pre-erythrocytic and erythrocytic parasite life cycle stages. As such, this model constitutes a powerful tool to identify novel, protective and stage-transcending T and B cell targets for incorporation into a multi-stage subunit vaccine.


Asunto(s)
Inmunización , Hígado/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Animales , Linfocitos B/inmunología , Femenino , Inmunización/métodos , Malaria/parasitología , Vacunas contra la Malaria/genética , Ratones , Plasmodium yoelii/crecimiento & desarrollo , Esporozoítos/inmunología , Linfocitos T/inmunología , Vacunación
13.
J Immunol ; 194(5): 2268-79, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25646303

RESUMEN

The costimulatory molecule CD40 enhances immunity through several distinct roles in T cell activation and T cell interaction with other immune cells. In a mouse model of immunity to liver stage Plasmodium infection, CD40 was critical for the full maturation of liver dendritic cells, accumulation of CD8(+) T cells in the liver, and protective immunity induced by immunization with the Plasmodium yoelii fabb/f(-) genetically attenuated parasite. Using mixed adoptive transfers of polyclonal wild-type and CD40-deficient CD8(+) T cells into wild-type and CD40-deficient hosts, we evaluated the contributions to CD8(+) T cell immunity of CD40 expressed on host tissues including APC, compared with CD40 expressed on the CD8(+) T cells themselves. Most of the effects of CD40 could be accounted for by expression in the T cells' environment, including the accumulation of large numbers of CD8(+) T cells in the livers of immunized mice. Thus, protective immunity generated during immunization with fabb/f(-) was largely dependent on effective APC licensing via CD40 signaling.


Asunto(s)
Antígenos CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Hígado/inmunología , Vacunas contra la Malaria/administración & dosificación , Malaria/prevención & control , Plasmodium yoelii/inmunología , Esporozoítos/inmunología , Traslado Adoptivo , Animales , Antígenos CD40/deficiencia , Antígenos CD40/genética , Linfocitos T CD8-positivos/parasitología , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/trasplante , Células Dendríticas/inmunología , Células Dendríticas/parasitología , Células Dendríticas/patología , Femenino , Eliminación de Gen , Expresión Génica , Hepatocitos/inmunología , Hepatocitos/parasitología , Hepatocitos/patología , Inmunidad Innata , Hígado/parasitología , Hígado/patología , Activación de Linfocitos , Malaria/inmunología , Malaria/parasitología , Malaria/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Transducción de Señal , Esporozoítos/química , Vacunas Atenuadas
14.
J Immunol ; 195(6): 2710-21, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26254338

RESUMEN

A CD1d-binding glycolipid, α-Galactosylceramide (αGalCer), activates invariant NK T cells and acts as an adjuvant. We previously identified a fluorinated phenyl ring-modified αGalCer analog, 7DW8-5, displaying nearly 100-fold stronger CD1d binding affinity. In the current study, 7DW8-5 was found to exert a more potent adjuvant effect than αGalCer for a vaccine based on radiation-attenuated sporozoites of a rodent malaria parasite, Plasmodium yoelii, also referred to as irradiated P. yoelii sporozoites (IrPySpz). 7DW8-5 had a superb adjuvant effect only when the glycolipid and IrPySpz were conjointly administered i.m. Therefore, we evaluated the effect of distinctly different biodistribution patterns of αGalCer and 7DW8-5 on their respective adjuvant activities. Although both glycolipids induce a similar cytokine response in sera of mice injected i.v., after i.m. injection, αGalCer induces a systemic cytokine response, whereas 7DW8-5 is locally trapped by CD1d expressed by dendritic cells (DCs) in draining lymph nodes (dLNs). Moreover, the i.m. coadministration of 7DW8-5 with IrPySpz results in the recruitment of DCs to dLNs and the activation and maturation of DCs. These events cause the potent adjuvant effect of 7DW8-5, resulting in the enhancement of the CD8(+) T cell response induced by IrPySpz and, ultimately, improved protection against malaria. Our study is the first to show that the colocalization of a CD1d-binding invariant NK T cell-stimulatory glycolipid and a vaccine, like radiation-attenuated sporozoites, in dLN-resident DCs upon i.m. conjoint administration governs the potency of the adjuvant effect of the glycolipid.


Asunto(s)
Antígenos CD1d/inmunología , Galactosilceramidas/farmacología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Linfocitos T CD8-positivos/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Femenino , Galactosilceramidas/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Células T Asesinas Naturales/inmunología , Plasmodium yoelii/inmunología , Unión Proteica/inmunología , Esporozoítos/inmunología
15.
Infect Immun ; 83(1): 39-47, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25312960

RESUMEN

After transmission by Anopheles mosquitoes, Plasmodium sporozoites travel to the liver, infect hepatocytes, and rapidly develop as intrahepatocytic liver stages (LS). Rodent models of malaria exhibit large differences in the magnitude of liver infection, both between parasite species and between strains of mice. This has been mainly attributed to differences in innate immune responses and parasite infectivity. Here, we report that BALB/cByJ mice are more susceptible to Plasmodium yoelii preerythrocytic infection than BALB/cJ mice. This difference occurs at the level of early hepatocyte infection, but expression levels of reported host factors that are involved in infection do not correlate with susceptibility. Interestingly, BALB/cByJ hepatocytes are more frequently polyploid; thus, their susceptibility converges on the previously observed preference of sporozoites to infect polyploid hepatocytes. Gene expression analysis demonstrates hepatocyte-specific differences in mRNA abundance for numerous genes between BALB/cByJ and BALB/cJ mice, some of which encode hepatocyte surface molecules. These data suggest that a yet-unknown receptor for sporozoite infection, present at elevated levels on BALB/cByJ hepatocytes and also polyploid hepatocytes, might facilitate Plasmodium liver infection.


Asunto(s)
Susceptibilidad a Enfermedades , Endocitosis , Hepatocitos/parasitología , Malaria/inmunología , Malaria/parasitología , Plasmodium yoelii/fisiología , Animales , Femenino , Perfilación de la Expresión Génica , Ratones Endogámicos BALB C
16.
Infect Immun ; 82(2): 808-17, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24478094

RESUMEN

Evidence from clinical trials of malaria vaccine candidates suggests that both cell-mediated and humoral immunity to pre-erythrocytic parasite stages can provide protection against infection. Novel pre-erythrocytic antibody (Ab) targets could be key to improving vaccine formulations, which are currently based on targeting antigens such as the circumsporozoite protein (CSP). However, methods to assess the effects of sporozoite-specific Abs on pre-erythrocytic infection in vivo remain underdeveloped. Here, we combined passive transfer of monoclonal Abs (MAbs) or immune serum with a luciferase-expressing Plasmodium yoelii sporozoite challenge to assess Ab-mediated inhibition of liver infection in mice. Passive transfer of a P. yoelii CSP MAb showed inhibition of liver infection when mice were challenged with sporozoites either intravenously or by infectious mosquito bite. However, inhibition was most potent for the mosquito bite challenge, leading to a more significant reduction of liver-stage burden and even a lack of progression to blood-stage parasitemia. This suggests that Abs provide effective protection against a natural infection. Inhibition of liver infection was also achieved by passive transfer of immune serum from whole-parasite-immunized mice. Furthermore, we demonstrated that passive transfer of a MAb against P. falciparum CSP inhibited liver-stage infection in a humanized mouse/P. falciparum challenge model. Together, these models constitute unique and sensitive in vivo methods to assess serum-transferable protection against Plasmodium sporozoite challenge.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antiprotozoarios/inmunología , Sueros Inmunes/inmunología , Malaria/prevención & control , Plasmodium falciparum/inmunología , Plasmodium yoelii/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunización Pasiva , Hígado/inmunología , Hígado/parasitología , Malaria/inmunología , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones SCID
17.
Cell Microbiol ; 15(9): 1508-1526, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23421981

RESUMEN

Plasmodium falciparum (Pf) blood stages express falstatin, an inhibitor of cysteine proteases (ICP), which is implicated in regulating proteolysis during red blood cell infection. Recent data using the Plasmodium berghei rodent malaria model suggested an additional role for ICP in the infection of hepatocytes by sporozoites and during liver-stage development. Here we further characterize the role of ICP in vivo during infection with Plasmodium yoelii (Py) and Pf. We found that Py-ICP was refractory to targeted gene deletion indicating an essential function during asexual blood-stage replication, but significant downregulation of ICP using a regulated system did not impact blood-stage growth. Py-ICP localized to vesicles within the asexual blood-stage parasite cytoplasm, as well as the parasitophorous vacuole, and was exported to dynamic exomembrane structures in the infected RBC. In sporozoites, expression was observed in rhoptries, in addition to intracellular vesicles distinct from TRAP containing micronemes. During liver-stage development, Py-ICP was confined to the parasite compartment until the final phase of liver-stage development when, after parasitophorous vacuolemembrane breakdown, it was released into the infected hepatocyte. Finally, we identified the cysteine protease yoelipain-2 as a binding partner of Py-ICP during blood-stage infection. These data show that ICP may be important in regulating proteolytic processes during blood-stage development, and is likely playing a role in liver stage-hepatocyte interactions at the time of exoerythrocytic merozoite release.


Asunto(s)
Inhibidores de Cisteína Proteinasa/metabolismo , Eritrocitos/parasitología , Membranas Intracelulares/metabolismo , Plasmodium yoelii/crecimiento & desarrollo , Plasmodium yoelii/metabolismo , Secuencia de Aminoácidos , Animales , Proteasas de Cisteína/metabolismo , Ratones , Datos de Secuencia Molecular , Unión Proteica , Alineación de Secuencia , Vacuolas/química , Vacuolas/parasitología
18.
Proc Natl Acad Sci U S A ; 108(10): 4053-6, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21368106

RESUMEN

The obligate mutualism between leafcutter ants and their Attamyces fungi originated 8 to 12 million years ago in the tropics, but extends today also into temperate regions in South and North America. The northernmost leafcutter ant Atta texana sustains fungiculture during winter temperatures that would harm the cold-sensitive Attamyces cultivars of tropical leafcutter ants. Cold-tolerance of Attamyces cultivars increases with winter harshness along a south-to-north temperature gradient across the range of A. texana, indicating selection for cold-tolerant Attamyces variants along the temperature cline. Ecological niche modeling corroborates winter temperature as a key range-limiting factor impeding northward expansion of A. texana. The northernmost A. texana populations are able to sustain fungiculture throughout winter because of their cold-adapted fungi and because of seasonal, vertical garden relocation (maintaining gardens deep in the ground in winter to protect them from extreme cold, then moving gardens to warmer, shallow depths in spring). Although the origin of leafcutter fungiculture was an evolutionary breakthrough that revolutionized the food niche of tropical fungus-growing ants, the original adaptations of this host-microbe symbiosis to tropical temperatures and the dependence on cold-sensitive fungal symbionts eventually constrained expansion into temperate habitats. Evolution of cold-tolerant fungi within the symbiosis relaxed constraints on winter fungiculture at the northern frontier of the leafcutter ant distribution, thereby expanding the ecological niche of an obligate host-microbe symbiosis.


Asunto(s)
Hormigas/fisiología , Evolución Biológica , Frío , Hongos/fisiología , Estaciones del Año , Simbiosis , Animales , Hormigas/parasitología
19.
Cell Microbiol ; 14(3): 316-24, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22151703

RESUMEN

For those stricken with malaria, the classic clinical symptoms are caused by the parasite's cyclic infection of red blood cells. However, this erythrocytic phase of the parasite's life cycle initiates from an asymptomatic pre-erythrocytic phase: the injection of sporozoites via the bite of a parasite-carrying Anopheline mosquito, and the ensuing infection of the liver. With the increased capabilities of studying liver stages in mice, much progress has been made elucidating the cellular and molecular basis of the parasite's progression through this bottleneck of its life cycle. Here we review relevant findings on how sporozoites prepare for infection of the liver and factors crucial to liver stage development as well as key host/parasite interactions.


Asunto(s)
Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Hígado/parasitología , Malaria/parasitología , Plasmodium/fisiología , Esporozoítos/fisiología , Animales , Culicidae/parasitología , Vectores de Enfermedades , Humanos , Malaria/inmunología , Merozoítos/inmunología , Merozoítos/fisiología , Plasmodium/inmunología , Esporozoítos/inmunología , Vacuolas/parasitología
20.
Sci Data ; 10(1): 195, 2023 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-37031232

RESUMEN

We describe the Queensland Twin Adolescent Brain (QTAB) dataset and provide a detailed methodology and technical validation to facilitate data usage. The QTAB dataset comprises multimodal neuroimaging, as well as cognitive and mental health data collected in adolescent twins over two sessions (session 1: N = 422, age 9-14 years; session 2: N = 304, 10-16 years). The MRI protocol consisted of T1-weighted (MP2RAGE), T2-weighted, FLAIR, high-resolution TSE, SWI, resting-state fMRI, DWI, and ASL scans. Two fMRI tasks were added in session 2: an emotional conflict task and a passive movie-watching task. Outside of the scanner, we assessed cognitive function using standardised tests. We also obtained self-reports of symptoms for anxiety and depression, perceived stress, sleepiness, pubertal development measures, and risk and protective factors. We additionally collected several biological samples for genomic and metagenomic analysis. The QTAB project was established to promote health-related research in adolescence.


Asunto(s)
Desarrollo del Adolescente , Encéfalo , Adolescente , Niño , Humanos , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Estudios Longitudinales , Imagen por Resonancia Magnética , Queensland , Gemelos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA