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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Biol ; 14(1): 117, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-28034300

RESUMEN

BACKGROUND: Increasing evidence suggests that influenza reassortment not only contributes to the emergence of new human pandemics but also plays an important role in seasonal influenza epidemics, disease severity, evolution, and vaccine efficacy. We studied this process within 2091 H3N2 full genomes utilizing a combination of the latest reassortment detection tools and more conventional phylogenetic analyses. RESULTS: We found that the amount of H3N2 intra-subtype reassortment depended on the number of sampled genomes, occurred with a steady frequency of 3.35%, and was not affected by the geographical origins, evolutionary patterns, or previous reassortment history of the virus. We identified both single reassortant genomes and reassortant clades, each clade representing one reassortment event followed by successful spread of the reassorted variant in the human population. It was this spread that was mainly responsible for the observed high presence of H3N2 intra-subtype reassortant genomes. The successfully spread variants were generally sampled within one year of their formation, highlighting the risk of their rapid spread but also presenting an opportunity for their rapid detection. Simultaneous spread of several different reassortant lineages was observed, and despite their limited average lifetime, second and third generation reassortment was detected, as well as reassortment between viruses belonging to different vaccine-associated clades, likely displaying differing antigenic properties. Some of the spreading reassortants remained confined to certain geographical regions, while others, sharing common properties in amino acid positions of the HA, NA, and PB2 segments, were found throughout the world. CONCLUSIONS: Detailed surveillance of seasonal influenza reassortment patterns and variant properties may provide unique information needed for prediction of spread and construction of future influenza vaccines.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A/genética , Evolución Molecular , Genoma Viral/genética , Humanos , Subtipo H3N2 del Virus de la Influenza A/clasificación , Gripe Humana/transmisión , Gripe Humana/virología , Filogenia
2.
Malar J ; 15(1): 377, 2016 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-27448805

RESUMEN

BACKGROUND: In this phase 1 clinical trial, healthy adult, malaria-naïve subjects were immunized with radiation-attenuated Plasmodium falciparum sporozoites (PfRAS) by mosquito bite and then underwent controlled human malaria infection (CHMI). The PfRAS model for immunization against malaria had previously induced >90 % sterile protection against homologous CHMI. This study was to further explore the safety, tolerability and protective efficacy of the PfRAS model and to provide biological specimens to characterize protective immune responses and identify protective antigens in support of malaria vaccine development. METHODS: Fifty-seven subjects were screened, 41 enrolled and 30 received at least one immunization. The true-immunized subjects received PfRAS via mosquito bite and the mock-immunized subjects received mosquito bites from irradiated uninfected mosquitoes. Sera and peripheral blood mononuclear cells (PBMCs) were collected before and after PfRAS immunizations. RESULTS: Immunization with PfRAS was generally safe and well tolerated, and repeated immunization via mosquito bite did not appear to increase the risk or severity of AEs. Local adverse events (AEs) of true-immunized and mock-immunized groups consisted of erythaema, papules, swelling, and induration and were consistent with reactions from mosquito bites seen in nature. Two subjects, one true- and one mock-immunized, developed large local reactions that completely resolved, were likely a result of mosquito salivary antigens, and were withdrawn from further participation as a safety precaution. Systemic AEs were generally rare and mild, consisting of headache, myalgia, nausea, and low-grade fevers. Two true-immunized subjects experienced fever, malaise, myalgia, nausea, and rigours approximately 16 h after immunization. These symptoms likely resulted from pre-formed antibodies interacting with mosquito salivary antigens. Ten subjects immunized with PfRAS underwent CHMI and five subjects (50 %) were sterilely protected and there was a significant delay to parasitaemia in the other five subjects. All ten subjects developed humoral immune responses to whole sporozoites and to the circumsporozoite protein prior to CHMI, although the differences between protected and non-protected subjects were not statistically significant for this small sample size. CONCLUSIONS: The protective efficacy of this clinical trial (50 %) was notably less than previously reported (>90 %). This may be related to differences in host genetics or the inherent variability in mosquito biting behavior and numbers of sporozoites injected. Differences in trial procedures, such as the use of leukapheresis prior to CHMI and of a longer interval between the final immunization and CHMI in these subjects compared to earlier trials, may also have reduced protective efficacy. This trial has been retrospectively registered at ISRCTN ID 17372582, May 31, 2016.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Culicidae/fisiología , Mordeduras y Picaduras de Insectos , Vacunas contra la Malaria/efectos adversos , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Adolescente , Adulto , Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Humanos , Vacunas contra la Malaria/administración & dosificación , Masculino , Persona de Mediana Edad , Plasmodium falciparum/efectos de la radiación , Esporozoítos/inmunología , Esporozoítos/efectos de la radiación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/inmunología , Adulto Joven
3.
MMWR Morb Mortal Wkly Rep ; 63(42): 947-9, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25340911

RESUMEN

On February 10, 2014, the USS Ardent, a U.S. Navy minesweeper, was moored in San Diego, California, while conducting training. Over the course of 3 days, 25 of 102 crew members sought medical care because of influenza-like illness (ILI). Nasal swab specimens were collected from each patient, and initial rapid influenza testing indicated 16 cases of influenza A. Ultimately, polymerase chain reaction (PCR) testing conducted by the Naval Health Research Center determined that 20 specimens were influenza A, of which 18 were subtype H3N2. Two specimens could not be subtyped. The HA gene sequence of an outbreak isolate was 99% identical to strains circulating during the 2013-14 influenza season and antigenically similar to the H3N2 component of the 2013-14 influenza vaccine. At the time of the outbreak, 99% of the crew had received influenza vaccine. Through the duration of the outbreak, the minesweeper squadron medical officer collaborated with Navy Environmental and Preventive Medicine Unit Five, higher-level Navy authorities, and County of San Diego Public Health Services to implement the outbreak response, which included disseminating outbreak information to surrounding Navy units, disinfecting the ship, sending home infected crew members, identifying family members at high risk, and providing antiviral medications and guidance. No crew member had onset of symptoms >6 days after the first crew member became ill. This outbreak highlights the risk for an H3N2 influenza outbreak among vaccinated and otherwise healthy young persons.


Asunto(s)
Brotes de Enfermedades , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/epidemiología , Personal Militar , Adulto , California/epidemiología , Trazado de Contacto , Brotes de Enfermedades/prevención & control , Humanos , Gripe Humana/prevención & control , Gripe Humana/virología , Masculino , Personal Militar/estadística & datos numéricos , Navíos , Adulto Joven
4.
Eur J Immunol ; 40(2): 437-42, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20063313

RESUMEN

Chronic helminth infections induce T-cell hyporesponsiveness, which may affect immune responses to other pathogens or to vaccines. This study investigates the influence of Treg activity on proliferation and cytokine responses to BCG and Plasmodium falciparum-parasitized RBC in Indonesian schoolchildren. Geohelminth-infected children's in vitro T-cell proliferation to either BCG or pRBC was reduced compared to that of uninfected children. Although the frequency of CD4(+)CD25(hi)FOXP3(+) T cells was similar regardless of infection status, the suppressive activity differed between geohelminth-infected and geohelminth-uninfected groups: Ag-specific proliferative responses increased upon CD4(+)CD25(hi) T-cell depletion in geohelminth-infected subjects only. In addition, IFN-gamma production in response to both BCG and parasitized RBC was increased after removal of CD4(+)CD25(hi) T cells. These data demonstrate that geohelminth-associated Treg influence immune responses to bystander Ag of mycobacteria and plasmodia. Geohelminth-induced immune modulation may have important consequences for co-endemic infections and vaccine trials.


Asunto(s)
Helmintiasis/inmunología , Malaria Falciparum/inmunología , Linfocitos T Reguladores/inmunología , Tuberculosis/inmunología , Vacuna BCG/inmunología , Vacuna BCG/farmacología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Niño , Eritrocitos/inmunología , Eritrocitos/parasitología , Helmintiasis/parasitología , Humanos , Indonesia , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Recuento de Linfocitos , Malaria Falciparum/microbiología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Tuberculosis/microbiología
5.
BMC Public Health ; 11 Suppl 2: S4, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21388564

RESUMEN

Capacity-building initiatives related to public health are defined as developing laboratory infrastructure, strengthening host-country disease surveillance initiatives, transferring technical expertise and training personnel. These initiatives represented a major piece of the Armed Forces Health Surveillance Center, Division of Global Emerging Infections Surveillance and Response System (AFHSC-GEIS) contributions to worldwide emerging infectious disease (EID) surveillance and response. Capacity-building initiatives were undertaken with over 80 local and regional Ministries of Health, Agriculture and Defense, as well as other government entities and institutions worldwide. The efforts supported at least 52 national influenza centers and other country-specific influenza, regional and U.S.-based EID reference laboratories (44 civilian, eight military) in 46 countries worldwide. Equally important, reference testing, laboratory infrastructure and equipment support was provided to over 500 field sites in 74 countries worldwide from October 2008 to September 2009. These activities allowed countries to better meet the milestones of implementation of the 2005 International Health Regulations and complemented many initiatives undertaken by other U.S. government agencies, such as the U.S. Department of Health and Human Services, the U.S. Agency for International Development and the U.S. Department of State.


Asunto(s)
Gripe Humana/epidemiología , Personal Militar , Salud Pública , Infecciones del Sistema Respiratorio/epidemiología , Vigilancia de Guardia , Salud Global , Agencias Gubernamentales , Humanos , Cooperación Internacional , Laboratorios , Estados Unidos
6.
Microbes Infect ; 9(12-13): 1439-46, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17913540

RESUMEN

Effective vaccines against infectious diseases and biological warfare agents remain an urgent public health priority. Studies have characterized the differentiation of effector and memory T cells and identified a subset of T cells capable of conferring enhanced protective immunity against pathogen challenge. We hypothesized that the kinetics of T cell differentiation influences the immunogenicity and protective efficacy of plasmid DNA vaccines, and tested this hypothesis in the Plasmodium yoelii murine model of malaria. We found that increasing the interval between immunizations significantly enhanced the frequency and magnitude of CD8+ and CD4+ T cell responses as well as protective immunity against sporozoite challenge. Moreover, the interval between immunizations was more important than the total number of immunizations. Immunization interval had a significantly greater impact on T cell responses and protective immunity than on antibody responses. With prolonged immunization intervals, T cell responses induced by homologous DNA only regimens achieved levels similar to those induced by heterologous DNA prime/ virus boost immunization at standard intervals. Our studies establish that the dosing interval significantly impacts the immunogenicity and protective efficacy of plasmid DNA vaccines.


Asunto(s)
Esquemas de Inmunización , Malaria/inmunología , Malaria/prevención & control , Plásmidos/inmunología , Plasmodium yoelii/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Inmunización , Inmunización Secundaria , Malaria/parasitología , Vacunas contra la Malaria/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Plasmodium yoelii/patogenicidad
7.
Influenza Other Respir Viruses ; 10(3): 220-3, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26920652
8.
Vaccine ; 34(33): 3907-12, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27265447

RESUMEN

BACKGROUND: Factors, such as age, comorbidities, vaccine type, herd immunity, previous influenza exposure, and antigenic shift may impact the immune response to the influenza vaccine, protection against circulating strains, and antibody waning. Evaluating vaccine effectiveness (VE) is important for informing timing of vaccine administration and evaluating overall vaccine benefit. METHODS: VE was assessed using febrile respiratory illness surveillance among Department of Defense non-active duty beneficiaries from influenza seasons 2010-2011 through 2013-2014. Respiratory specimens were taken from participants meeting the case definition and tested by polymerase chain reaction for influenza. VE was calculated using logistic regression and by taking 1 minus the odds ratio of being vaccinated in the laboratory confirmed positive influenza cases versus laboratory confirmed negative controls. RESULTS: This study included 1486 participants. We found an overall adjusted VE that provided significant and fairly consistent protection ranging from 54% to 67% during 0-180days postvaccination. This VE dropped to -11% (95% confidence interval: -102% to 39%) during 181-365days. CONCLUSIONS: Our study found moderate VE up to 6months postvaccination. Since the influenza season starts at different times each year, optimal timing is difficult to predict. Consequently, early influenza vaccination may still offer the best overall protection.


Asunto(s)
Vacunas contra la Influenza/uso terapéutico , Gripe Humana/prevención & control , Potencia de la Vacuna , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Vacunas contra la Influenza/inmunología , Modelos Logísticos , Masculino , Persona de Mediana Edad , Enfermedades Respiratorias/diagnóstico , Adulto Joven
9.
J Leukoc Biol ; 72(6): 1164-71, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12488498

RESUMEN

Efficient activation of specific immune responses requires a concerted interaction between T cells and antigen-presenting cells. A requirement for bystander participation of CD4+ T cells for expansion and maintenance of memory CD8+ T cells has been noted in several models, but a role with regard to effector CD8+ T responses has not been well-defined. In this report, the requirement of bystander participation for optimal induction of antigen-specific CD8+ T cell effector function was determined by directly quantitating antigen-specific interferon-gamma (IFN-gamma) CD8+ T cell responses by enzyme-linked immunospot assays, and by indirectly evaluating induction of the chemokine monokine induced by IFN-gamma as a marker for IFN-gamma-mediated effector function. Our results demonstrate that bystander cell participation, mediated by CD4+ T cell and natural killer (NK) cells, is required for optimal induction of antigen-specific CD8+ T cell effector responses. Our data further establish a novel role for NK cells in the activation of antigen-specific immune responses.


Asunto(s)
Antígenos Virales/inmunología , Efecto Espectador/inmunología , Linfocitos T CD8-positivos/inmunología , Péptidos y Proteínas de Señalización Intercelular , Adulto , Linfocitos T CD4-Positivos/inmunología , Quimiocina CXCL9 , Quimiocinas CXC/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Células Asesinas Naturales/inmunología , Persona de Mediana Edad
10.
PLoS One ; 10(3): e0121037, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25816244

RESUMEN

BACKGROUND: In 2011, a new variant of influenza A(H3N2) emerged that contained a recombination of genes from swine H3N2 viruses and the matrix (M) gene of influenza A(H1N1)pdm09 virus. New combinations and variants of pre-existing influenza viruses are worrisome if there is low or nonexistent immunity in a population, which increases chances for an outbreak or pandemic. METHODS: Sera collected in 2011 were obtained from US Department of Defense service members in three age groups: 19-21 years, 32-33 years, and 47-48 years. Pre- and post-vaccination samples were available for the youngest age group, and postvaccination samples for the two older groups. Specimens were tested using microneutralization assays for antibody titers against H3N2v (A/Indiana/10/2011) and seasonal H3N2 virus (A/Perth/16/2009). RESULTS: The youngest age group had significantly (p<0.05) higher geometric mean titers for H3N2v with 165 (95% confidence interval [CI]: 105-225) compared with the two older groups, aged 32-33 and 47-48 years, who had geometric mean titers of 68 (95% CI: 55-82) and 46 (95% CI: 24-65), respectively. Similarly, the youngest age group also had the highest geometric mean titers for seasonal H3N2. In the youngest age group, the proportion of patients who seroconverted after vaccination was 12% for H3N2v and 27% for seasonal H3N2. DISCUSSION: Our results were similar to previous studies that found highest seroprotection among young adults and decreasing titers among older adults. The proportion of 19- to 21-year-olds who seroconverted after seasonal vaccination was low and similar to previous findings. Improving our understanding of H3N2v immunity among different age groups in the United States can help inform vaccination plans if H3N2v becomes more transmissible in the future.


Asunto(s)
Anticuerpos Antivirales/sangre , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Adulto , Humanos , Subtipo H3N2 del Virus de la Influenza A/genética , Gripe Humana/inmunología , Persona de Mediana Edad , Pruebas de Neutralización , Estados Unidos , United States Department of Defense , Adulto Joven
11.
PLoS One ; 9(12): e114871, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25549089

RESUMEN

BACKGROUND: Diagnostic tests for respiratory infections can be costly and time-consuming. Improved characterization of specific respiratory pathogens by identifying frequent signs, symptoms and demographic characteristics, along with improving our understanding of coinfection rates and seasonality, may improve treatment and prevention measures. METHODS: Febrile respiratory illness (FRI) and severe acute respiratory infection (SARI) surveillance was conducted from October 2011 through March 2013 among three US populations: civilians near the US-Mexico border, Department of Defense (DoD) beneficiaries, and military recruits. Clinical and demographic questionnaire data and respiratory swabs were collected from participants, tested by PCR for nine different respiratory pathogens and summarized. Age stratified characteristics of civilians positive for influenza and recruits positive for rhinovirus were compared to other and no/unknown pathogen. Seasonality and coinfection rates were also described. RESULTS: A total of 1444 patients met the FRI or SARI case definition and were enrolled in this study. Influenza signs and symptoms varied across age groups of civilians. Recruits with rhinovirus had higher percentages of pneumonia, cough, shortness of breath, congestion, cough, less fever and longer time to seeking care and were more likely to be male compared to those in the no/unknown pathogen group. Coinfections were found in 6% of all FRI/SARI cases tested and were most frequently seen among children and with rhinovirus infections. Clear seasonal trends were identified for influenza, rhinovirus, and respiratory syncytial virus. CONCLUSIONS: The age-stratified clinical characteristics associated with influenza suggest that age-specific case definitions may improve influenza surveillance and identification. Improving identification of rhinoviruses, the most frequent respiratory infection among recruits, may be useful for separating out contagious individuals, especially when larger outbreaks occur. Overall, describing the epidemiology of pathogen specific respiratory diseases can help improve clinical diagnoses, establish baselines of infection, identify outbreaks, and help prioritize the development of new vaccines and treatments.


Asunto(s)
Infecciones del Sistema Respiratorio/epidemiología , Estaciones del Año , Encuestas y Cuestionarios , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología , Estados Unidos/epidemiología
12.
PLoS One ; 8(3): e57899, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23526959

RESUMEN

BACKGROUND: Helminth infections are proposed to have immunomodulatory activities affecting health outcomes either detrimentally or beneficially. We evaluated the effects of albendazole treatment, every three months for 21 months, on STH, malarial parasitemia and allergy. METHODS AND FINDINGS: A household-based cluster-randomized, double-blind, placebo-controlled trial was conducted in an area in Indonesia endemic for STH. Using computer-aided block randomization, 481 households (2022 subjects) and 473 households (1982 subjects) were assigned to receive placebo and albendazole, respectively, every three months. The treatment code was concealed from trial investigators and participants. Malarial parasitemia and malaria-like symptoms were assessed in participants older than four years of age while skin prick test (SPT) to allergens as well as reported symptoms of allergy in children aged 5-15 years. The general impact of treatment on STH prevalence and body mass index (BMI) was evaluated. Primary outcomes were prevalence of malarial parasitemia and SPT to any allergen. Analysis was by intention to treat. At 9 and 21 months post-treatment 80.8% and 80.1% of the study subjects were retained, respectively. The intensive treatment regiment resulted in a reduction in the prevalence of STH by 48% in albendazole and 9% in placebo group. Albendazole treatment led to a transient increase in malarial parasitemia at 6 months post treatment (OR 4.16(1.35-12.80)) and no statistically significant increase in SPT reactivity (OR 1.18(0.74-1.86) at 9 months or 1.37 (0.93-2.01) 21 months). No effect of anthelminthic treatment was found on BMI, reported malaria-like- and allergy symptoms. No adverse effects were reported. CONCLUSIONS: The study indicates that intensive community treatment of 3 monthly albendazole administration for 21 months over two years leads to a reduction in STH. This degree of reduction appears safe without any increased risk of malaria or allergies. TRIAL REGISTRATION: Controlled-Trials.com ISRCTN83830814.


Asunto(s)
Albendazol/administración & dosificación , Albendazol/efectos adversos , Antihelmínticos/administración & dosificación , Antihelmínticos/efectos adversos , Helmintiasis/prevención & control , Malaria/etiología , Malaria/inmunología , Parasitemia/etiología , Parasitemia/inmunología , Adolescente , Adulto , Niño , Preescolar , Método Doble Ciego , Esquema de Medicación , Femenino , Helmintiasis/transmisión , Humanos , Hipersensibilidad/etiología , Indonesia , Masculino , Persona de Mediana Edad , Suelo/parasitología , Adulto Joven
17.
J Infect Dis ; 195(2): 174-84, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17191162

RESUMEN

BACKGROUND: Bioterrorism-related anthrax exposures occurred at the US Capitol in 2001. Exposed individuals received antibiotics and anthrax vaccine adsorbed immunization. METHODS: A prospective longitudinal study of 124 subjects--stratified on the basis of spore exposure, nasopharyngeal culture results, and immunization status from inside and outside an epidemiologically defined exposure zone--was performed to describe clinical outcome and immune responses after Bacillus anthracis exposure. Antibody and cell-mediated immune (CMI) responses to protective antigen (PA) and lethal factor were assayed by enzyme-linked immunosorbent assay and fluorescence-activated cell sorting. RESULTS: Antibody and CMI dose-exposure responses, albeit generally of low magnitude, were seen for unimmunized subjects from inside, within the perimeter, and outside the exposure zone and in nonexposed control subjects. Anti-PA antibody and CMI responses were detected in 94% and 86% of immunized subjects. No associations were seen between symptoms and exposure levels or immune responses. CONCLUSIONS: Anthrax spores primed cellular and possibly antibody immune responses in a dose-dependent manner and may have enhanced vaccine boost and recall responses. Immune responses were detected inside the perimeter and outside the exposure zone, which implies more-extensive spore exposure than was predicted. Despite postexposure prophylaxis with antibiotics, inhalation of B. anthracis spores resulted in stimulation of the immune system and possibly subclinical infection, and the greater the exposure, the more complete the immune response. The significance of low-level exposure should not be underestimated.


Asunto(s)
Vacunas contra el Carbunco/administración & dosificación , Carbunco/epidemiología , Carbunco/inmunología , Antibacterianos/administración & dosificación , Bacillus anthracis/patogenicidad , Bioterrorismo , Carbunco/fisiopatología , Carbunco/prevención & control , Vacunas contra el Carbunco/inmunología , Antibacterianos/uso terapéutico , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Bacillus anthracis/crecimiento & desarrollo , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , District of Columbia/epidemiología , Humanos , Esquemas de Inmunización , Exposición por Inhalación , Linfocitos/inmunología , Monocitos/inmunología , Esporas Bacterianas/inmunología , Resultado del Tratamiento
18.
Blood ; 99(2): 590-9, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11781243

RESUMEN

Adoptive transfer of autologous preinfection-collected peripheral blood mononuclear cells (PBMCs) or activated CD4(+) T cells was performed in simian immunodeficiency virus (SIVmac239)-infected monkeys following short-term antiviral therapy with PMPA (9-R-[2-phosphonylmethoxypropyl] adenine). Short-term chemotherapy alone led to a transient decrease in plasma and cellular proviral DNA loads and transient rescue of gag/pol and env cytotoxic T-lymphocyte precursors (pCTLs). However, cessation of therapy allowed for SIV infection to resume its clinical course. PMPA chemotherapy coupled with infusions of either autologous pre-SIV infection-collected PBMCs or activated CD4(+) T cells led to extended control of plasma and cellular proviral DNA loads after infusion, in spite of the fact that the transfused cells were not primed against SIV. However, qualitatively different antiviral defenses were induced by infusion of unfractionated and unmanipulated PBMCs versus purified and activated CD4(+) T cells: PBMC infusions significantly favored development of SIVenv-specific pCTLs, neutralizing antibodies, and secretion of soluble noncytotoxic suppressor factors of SIV replication. In contrast, activated CD4(+) T cells predominantly promoted CTL responses to SIVgag/pol and SIVenv. In addition, infusion of influenza-primed activated CD4(+) T cells markedly enhanced influenza-specific pCTL responses, whereas infusion of similarly influenza-primed unfractionated PBMCs enhanced such pCTL responses only modestly, suggesting that the predominant immune defect after SIV infection lies in the T helper cell compartment rather than the effector cell compartment. Thus, adoptive immunotherapy with autologous "SIV naïve" CD4(+) lymphocytes was sufficient to rescue cell-mediated immune responses and induce long-term anti-SIV control and immune responses in the absence of continued antiviral chemotherapy.


Asunto(s)
Adenina/análogos & derivados , Linfocitos T CD4-Positivos/trasplante , Inmunoterapia Adoptiva , Organofosfonatos , Síndrome de Inmunodeficiencia Adquirida del Simio/terapia , Virus de la Inmunodeficiencia de los Simios/inmunología , Subgrupos de Linfocitos T/trasplante , Adenina/uso terapéutico , Animales , Anticuerpos Antivirales/sangre , Antivirales/uso terapéutico , Transfusión de Sangre Autóloga , Linfocitos T CD4-Positivos/inmunología , Terapia Combinada , ADN Viral/sangre , Progresión de la Enfermedad , Leucocitos Mononucleares/trasplante , Macaca mulatta , Pruebas de Neutralización , Compuestos Organofosforados/uso terapéutico , Provirus/aislamiento & purificación , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Tenofovir , Carga Viral
19.
Antimicrob Agents Chemother ; 48(7): 2455-63, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15215094

RESUMEN

CEL-1000 (DGQEEKAGVVSTGLIGGG) is a novel potential preventative and therapeutic agent. We report that CEL-1000 confers a high degree of protection against Plasmodium sporozoite challenge in a murine model of malaria, as shown by the total absence of blood stage infection following challenge with 100 sporozoites (100% protection) and by a substantial reduction (400-fold) of liver stage parasite RNA following challenge with 50,000 sporozoites. CEL-1000 protection was demonstrated in A/J (H-2(a)) and C3H/HeJ (H-2(k)) mice but not in BALB/c (H-2(d)) or CAF1 (A/J x BALB/c F(1) hybrid) mice. In CEL-1000-treated and protected mice, high levels of gamma interferon (IFN-gamma) in serum and elevated frequencies of hepatic and splenic CD4+ IFN-gamma-positive T cells were detected 24 h after administration of an additional dose of CEL-1000. Treatment of A/J mice that received CEL-1000 with antibodies against IFN-gamma just prior to challenge abolished the protection, and a similar treatment with antibodies against CD4+ T cells partially reduced the level of protection, while treatment with control antibodies or antibodies specific for interleukin-12 (IL-12), CD8+ T cells, or NK cells had no effect. Our data establish that the protection induced by CEL-1000 is dependent on IFN-gamma and is partially dependent on CD4+ T cells but is independent of CD8+ T cells, NK cells, and IL-12 at the effector phase and does not induce a detectable antibody response.


Asunto(s)
Antígenos de Protozoos/inmunología , Antimaláricos/farmacología , Genes MHC Clase II/genética , Malaria/prevención & control , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Interferón gamma/inmunología , Cinética , Hígado/química , Hígado/parasitología , Malaria/inmunología , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Plasmodium berghei/inmunología , Plasmodium yoelii/inmunología , ARN/química , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Infect Immun ; 70(7): 3493-9, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12065488

RESUMEN

The persistence of immunity to malaria induced in mice by a heterologous DNA priming and poxvirus boosting regimen was characterized. Mice were immunized by priming with DNA vaccine plasmids encoding the Plasmodium yoelii circumsporozoite protein (PyCSP) and murine granulocyte-macrophage colony-stimulating factor and boosting with recombinant vaccinia encoding PyCSP. BALB/c mice immunized with either high-dose (100 microg of p PyCSP plus 30 microg of pGM-CSF) or low-dose (1 microg of p PyCSP plus 1 microg of pGM-CSF DNA) priming were protected against challenge with 50 P. yoelii sporozoites. Protection 2 weeks after immunization was 70 to 100%, persisted at this level for at least 20 weeks, and declined to 30 to 40% by 28 weeks. Eight of eight mice protected at 20 weeks were still protected when rechallenged at 40 weeks. The antigen (Ag)-specific effector CD8(+)-T-cell population present 2 weeks after boosting had ex vivo Ag-specific cytolytic activity, expressed both gamma interferon (IFN-gamma) and tumor necrosis factor alpha, and constituted 12 to 20% of splenic CD8(+) T cells. In contrast, the memory CD8(+)-Ag-specific-cell population at 28 weeks lacked cytolytic activity and constituted only 6% of splenic CD8(+) T cells, but at the single-cell level it produced significantly higher levels of IFN-gamma than the effectors. High levels of Ag- or parasite-specific antibodies present 2 weeks after boosting had declined three- to sevenfold by 28 weeks. Low-dose priming was similarly immunogenic and as protective as high-dose priming against a 50-, but not a 250-, sporozoite challenge. These results demonstrate that a heterologous priming and boosting vaccination can provide lasting protection against malaria in this model system.


Asunto(s)
Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , ADN Protozoario/inmunología , Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Plasmodium yoelii/inmunología , Proteínas Protozoarias/inmunología , Vacunas de ADN/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/genética , Femenino , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Inmunización Secundaria , Memoria Inmunológica/inmunología , Malaria/inmunología , Vacunas contra la Malaria/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Plasmodium yoelii/genética , Proteínas Protozoarias/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/inmunología , Factores de Tiempo , Vacunas de ADN/genética , Virus Vaccinia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA