Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Nat Med ; 26(8): 1183-1192, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32770165

RESUMO

Digital technologies are being harnessed to support the public-health response to COVID-19 worldwide, including population surveillance, case identification, contact tracing and evaluation of interventions on the basis of mobility data and communication with the public. These rapid responses leverage billions of mobile phones, large online datasets, connected devices, relatively low-cost computing resources and advances in machine learning and natural language processing. This Review aims to capture the breadth of digital innovations for the public-health response to COVID-19 worldwide and their limitations, and barriers to their implementation, including legal, ethical and privacy barriers, as well as organizational and workforce barriers. The future of public health is likely to become increasingly digital, and we review the need for the alignment of international strategies for the regulation, evaluation and use of digital technologies to strengthen pandemic management, and future preparedness for COVID-19 and other infectious diseases.


Assuntos
Infecções por Coronavirus/prevenção & controle , Pandemias/estatística & dados numéricos , Pneumonia Viral/prevenção & controle , Vigilância da População , Saúde Pública/estatística & dados numéricos , Betacoronavirus/patogenicidade , COVID-19 , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/virologia , Humanos , Aprendizado de Máquina , Processamento de Linguagem Natural , Pandemias/prevenção & controle , Pneumonia Viral/epidemiologia , Pneumonia Viral/virologia , Privacidade , SARS-CoV-2
4.
Sci Rep ; 7(1): 11971, 2017 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-28931860

RESUMO

The development is reported of an ultra-rapid, point-of-care diagnostic device which harnesses surface acoustic wave (SAW) biochips, to detect HIV in a finger prick of blood within 10 seconds (sample-in-result-out). The disposable quartz biochip, based on microelectronic components found in every consumer smartphone, is extremely fast because no complex labelling, amplification or wash steps are needed. A pocket-sized control box reads out the SAW signal and displays results electronically. High analytical sensitivity and specificity are found with model and real patient blood samples. The findings presented here open up the potential of consumer electronics to cut lengthy test waiting times, giving patients on the spot access to potentially life-saving treatment and supporting more timely public health interventions to prevent disease transmission.


Assuntos
Técnicas Biossensoriais/métodos , Infecções por HIV/diagnóstico , Sistemas Automatizados de Assistência Junto ao Leito , Smartphone , Humanos , Sensibilidade e Especificidade , Tempo
6.
PLoS Pathog ; 13(4): e1006299, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28406982

RESUMO

Human cytomegalovirus (CMV) infection is a substantial cause of morbidity and mortality in immunocompromised hosts and globally is one of the most important congenital infections. The nucleoside analogue ganciclovir (GCV), which requires initial phosphorylation by the viral UL97 kinase, is the mainstay for treatment. To date, CMV decay kinetics during GCV therapy have not been extensively investigated and its clinical implications not fully appreciated. We measured CMV DNA levels in the blood of 92 solid organ transplant recipients with CMV disease over the initial 21 days of ganciclovir therapy and identified four distinct decay patterns, including a new pattern exhibiting a transient viral rebound (Hump) following initial decline. Since current viral dynamics models were unable to account for this Hump profile, we developed a novel multi-level model, which includes the intracellular role of UL97 in the continued activation of ganciclovir, that successfully described all the decline patterns observed. Fitting the data allowed us to estimate ganciclovir effectiveness in vivo (mean 92%), infected cell half-life (mean 0.7 days), and other viral dynamics parameters that determine which of the four kinetic patterns will ensue. An important clinical implication of our results is that the virological efficacy of GCV operates over a broad dose range. The model also raises the possibility that GCV can drive replication to a new lower steady state but ultimately cannot fully eradicate it. This model is likely to be generalizable to other anti-CMV nucleoside analogs that require activation by viral enzymes such as UL97 or its homologues.


Assuntos
Antivirais/metabolismo , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/efeitos dos fármacos , Farmacorresistência Viral/genética , Ganciclovir/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Ativação Metabólica , Antivirais/farmacologia , Antivirais/uso terapêutico , Citomegalovirus/genética , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/virologia , Ganciclovir/farmacologia , Ganciclovir/uso terapêutico , Meia-Vida , Humanos , Hospedeiro Imunocomprometido , Modelos Teóricos , Mutação , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Replicação Viral/efeitos dos fármacos
7.
F1000Res ; 6: 138, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28299191

RESUMO

Congenital cytomegalovirus (CMV) remains a leading cause of disability in children. Understanding the pathogenesis of infection from the mother via the placenta to the neonate is crucial if we are to produce new interventions and provide supportive mechanisms to improve the outcome of congenitally infected children. In recent years, some major goals have been achieved, including the diagnosis of primary maternal CMV infection in pregnant women by using the anti-CMV IgG avidity test and the diagnosis and prognosis of foetal CMV infection by using polymerase chain reaction real-time tests to detect and quantify the virus in amniotic fluid. This review summarises recent advances in our understanding and highlights where challenges remain, especially in vaccine development and anti-viral therapy of the pregnant woman and the neonate. Currently, no therapeutic options during pregnancy are available except those undergoing clinical trials, whereas valganciclovir treatment is recommended for congenitally infected neonates with moderately to severely symptomatic disease.

8.
AIDS Res Hum Retroviruses ; 33(4): 332-338, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27796131

RESUMO

To evaluate the impact of highly active antiretroviral therapy (HAART) on CMV transmission and breast milk level in the context of maternal HIV. Specimens from a randomized trial conducted in Nairobi, Kenya between 2003-2005 were used to compare CMV transmission and breast milk levels between mother-infant pairs randomized to HAART versus short-course antenatal zidovudine plus single-dose nevirapine (ZDV/sdNVP) for prevention of mother-to-child HIV transmission (PMTCT). Fifty-one antiretroviral-naïve women ≤32 weeks gestation, and CD4 between 200-500 cells/mm3 were randomized at 34 weeks to begin either antenatal ZDV/sdNVP, or HAART through 6 months postpartum. Mean breast milk CMV levels and transmission were compared between arms. Age, sociodemographics, CD4%, and HIV plasma RNA viral load were similar between arms at baseline. CMV viral loads were measured from 243 infant plasma and 185 breast milk specimens during the first year postpartum. The probability of infant CMV infection at 12 months was 19% lower in the HAART arm compared to ZDV/sdNVP (75% vs. 94%, p = .04). All women had CMV detected in breast milk, with 72%, 98%, and 97% testing positive during the first, second, and third weeks postpartum, respectively. There was a trend for early higher mean breast milk CMV level in the HAART arm at 1 week (p = .08), and there was significantly slower decline in breast milk CMV levels (area under the curve, p = .01). HAART started during the third trimester may decrease infant CMV infections, by mechanisms independent of breast milk CMV levels. CLINICAL TRIALS REGISTRATION: NCT00167674.


Assuntos
Terapia Antirretroviral de Alta Atividade , Infecções por Citomegalovirus/prevenção & controle , Citomegalovirus/isolamento & purificação , Infecções por HIV/complicações , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Leite Humano/virologia , Carga Viral , Adulto , Infecções por Citomegalovirus/transmissão , Feminino , Infecções por HIV/tratamento farmacológico , Humanos , Lactente , Recém-Nascido , Quênia , Gravidez , Complicações Infecciosas na Gravidez/tratamento farmacológico , Adulto Jovem
9.
PLoS One ; 11(9): e0163722, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27684379

RESUMO

BACKGROUND: To help decide when to start and when to stop pre-emptive therapy for cytomegalovirus infection, we conducted two open-label randomized controlled trials in renal, liver and bone marrow transplant recipients in a single centre where pre-emptive therapy is indicated if viraemia exceeds 3000 genomes/ml (2520 IU/ml) of whole blood. METHODS: Patients with two consecutive viraemia episodes each below 3000 genomes/ml were randomized to continue monitoring or to immediate treatment (Part A). A separate group of patients with viral load greater than 3000 genomes/ml was randomized to stop pre-emptive therapy when two consecutive levels less than 200 genomes/ml (168 IU/ml) or less than 3000 genomes/ml were obtained (Part B). For both parts, the primary endpoint was the occurrence of a separate episode of viraemia requiring treatment because it was greater than 3000 genomes/ml. RESULTS: In Part A, the primary endpoint was not significantly different between the two arms; 18/32 (56%) in the monitor arm had viraemia greater than 3000 genomes/ml compared to 10/27 (37%) in the immediate treatment arm (p = 0.193). However, the time to developing an episode of viraemia greater than 3000 genomes/ml was significantly delayed among those randomized to immediate treatment (p = 0.022). In Part B, the primary endpoint was not significantly different between the two arms; 19/55 (35%) in the less than 200 genomes/ml arm subsequently had viraemia greater than 3000 genomes/ml compared to 23/51 (45%) among those randomized to stop treatment in the less than 3000 genomes/ml arm (p = 0.322). However, the duration of antiviral treatment was significantly shorter (p = 0.0012) in those randomized to stop treatment when viraemia was less than 3000 genomes/ml. DISCUSSION: The results illustrate that patients have continuing risks for CMV infection with limited time available for intervention. We see no need to alter current rules for stopping or starting pre-emptive therapy.

10.
J Infect Dis ; 213(6): 992-8, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26518046

RESUMO

BACKGROUND: Cytomegalovirus (CMV) is associated with morbidity and mortality in human immunodeficiency virus (HIV)-exposed infants. We assessed the effect of and relative contribution of breastfeeding to CMV acquisition among infants delivered by HIV-infected mothers. METHODS: Between 1993 and 1998 pregnant, HIV-infected women in Nairobi, Kenya, were randomly assigned to breastfeed or formula-feed their infants in an HIV transmission study. Women were allocated equally between treatment arms, and the study was not blinded. The primary endpoint of this nested study was time to infant CMV infection. RESULTS: CMV infection was assessed in 138 breastfed and 134 formula-fed infants. Baseline characteristics were similar between arms. Breastfed infants acquired CMV earlier than formula-fed infants (median age of acquisition, 4.26 vs 9.87 months; P < .001) and had a higher 1-year probability of CMV infection (0.89 vs 0.69; P < .001). Breastfeeding was associated with a 1.6-fold increased risk of infant CMV acquisition independent of infant HIV status (multivariable hazard ratio, 1.61; 95% confidence interval, 1.20-2.16; P = .002). Approximately one third of CMV infections occurred during the peripartum period, with 40% acquired through breastfeeding and the remainder acquired through modes other than breast milk. CONCLUSIONS: Preventing CMV acquisition may be a priority for HIV-exposed infants, but there is a narrow window of opportunity for intervention. Approaches that reduce maternal cervical and breast milk CMV reactivation may help delay infant infection.


Assuntos
Infecções por Citomegalovirus/transmissão , Infecções por HIV/complicações , Transmissão Vertical de Doenças Infecciosas , Adulto , Aleitamento Materno , Estudos de Coortes , Infecções por Citomegalovirus/complicações , DNA Viral/sangue , DNA Viral/isolamento & purificação , Feminino , Humanos , Lactente , Fórmulas Infantis , Recém-Nascido , Gravidez , Fatores de Risco , Adulto Jovem
12.
J Gen Virol ; 96(Pt 1): 131-143, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25312585

RESUMO

Graft rejection in transplant patients is managed clinically by suppressing T-cell function with immunosuppressive drugs such as prednisolone and methylprednisolone. In such immunocompromised hosts, human cytomegalovirus (HCMV) is an important opportunistic pathogen and can cause severe morbidity and mortality. Currently, the effect of glucocorticosteroids (GCSs) on the HCMV life cycle remains unclear. Previous reports showed enhanced lytic replication of HCMV in vitro in the presence of GCSs. In the present study, we explored the implications of steroid exposure on latency and reactivation. We observed a direct effect of several GCSs used in the clinic on the activation of a quiescent viral major immediate-early promoter in stably transfected THP-1 monocytic cells. This activation was prevented by the glucocorticoid receptor (GR) antagonist Ru486 and by shRNA-mediated knockdown of the GR. Consistent with this observation, prednisolone treatment of latently infected primary monocytes resulted in HCMV reactivation. Analysis of the phenotype of these cells showed that treatment with GCSs was correlated with differentiation to an anti-inflammatory macrophage-like cell type. On the basis that these observations may be pertinent to HCMV reactivation in post-transplant settings, we retrospectively evaluated the incidence, viral kinetics and viral load of HCMV in liver transplant patients in the presence or absence of GCS treatment. We observed that combination therapy of baseline prednisolone and augmented methylprednisolone, upon organ rejection, significantly increased the incidence of HCMV infection in the intermediate risk group where donor and recipient are both HCMV seropositive (D+R+) to levels comparable with the high risk D+R- group.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Glucocorticoides/metabolismo , Fígado/virologia , Células Mieloides/virologia , Ativação Viral/fisiologia , Latência Viral/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Diferenciação Celular/fisiologia , Linhagem Celular , Doenças Transmissíveis/metabolismo , Doenças Transmissíveis/virologia , Infecções por Citomegalovirus/virologia , Feminino , Humanos , Hospedeiro Imunocomprometido/fisiologia , Fígado/metabolismo , Transplante de Fígado/métodos , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Monócitos/virologia , Células Mieloides/metabolismo , Receptores de Glucocorticoides/metabolismo , Carga Viral/fisiologia , Adulto Jovem
14.
PLoS One ; 9(2): e87855, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24504006

RESUMO

BACKGROUND: Studies in HIV-1-infected infants and HIV-1-exposed, uninfected infants link early cytomegalovirus (CMV) acquisition with growth delay and cognitive impairment. We investigated maternal valacyclovir to delay infant acquisition of CMV. METHODS: Pregnant women with HIV-1, HSV-2 and CD4 count >250 cells/µl were randomized at 34 weeks gestation to 500 mg twice-daily valacyclovir or placebo for 12 months. Maternal CMV DNA was measured in plasma at 34 weeks gestation, in cervical secretions at 34 and 38 weeks gestation, and in breast milk at 7 postpartum timepoints; infant CMV DNA was measured in dried blood spots at 8 timepoints including birth. RESULTS: Among 148 women, 141 infants were compared in intent-to-treat analyses. Maternal and infant characteristics were similar between study arms. Infant CMV acquisition did not differ between study arms, with 46/70 infants (66%) in placebo arm and 47/71 infants (66%) in the valacyclovir arm acquiring CMV; median time to CMV detection did not differ. CMV DNA was detected in 92% of 542 breast milk specimens with no difference in CMV level between study arms. Change in cervical shedding of CMV DNA between baseline and 38 weeks was 0.40-log greater in the placebo arm than the valacyclovir arm (p = 0.05). CONCLUSIONS: In this cohort of HIV-1-seropositive mothers, two-thirds of infants acquired CMV by one year. Maternal valacyclovir had no effect on timing of infant CMV acquisition or breast milk CMV viral loads, although it modestly reduced cervical CMV shedding. Maternal prophylaxis to reduce infant CMV acquisition warrants further evaluation in trials with antiviral agents. TRIALS REGISTRATION: ClinicalTrials.gov NCT00530777.


Assuntos
Aciclovir/análogos & derivados , Antivirais/uso terapêutico , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus , Soropositividade para HIV , Transmissão Vertical de Doenças Infecciosas , Valina/análogos & derivados , Aciclovir/administração & dosagem , Aciclovir/farmacocinética , Aciclovir/uso terapêutico , Antivirais/administração & dosagem , Antivirais/farmacocinética , Infecções por Citomegalovirus/prevenção & controle , Infecções por Citomegalovirus/virologia , Feminino , Humanos , Lactente , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Leite Humano/virologia , Gravidez , Pré-Medicação , Resultado do Tratamento , Valaciclovir , Valina/administração & dosagem , Valina/farmacocinética , Valina/uso terapêutico , Carga Viral , Eliminação de Partículas Virais
15.
Clin Infect Dis ; 58(4): 564-72, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24192386

RESUMO

BACKGROUND: Cytomegalovirus (CMV) infection is associated with adverse outcomes in human immunodeficiency virus (HIV)-exposed infants. Determinants of vertical CMV transmission in the setting of maternal HIV-1 infection are not well-defined. METHODS: CMV and HIV-1 levels were measured in plasma, cervical secretions, and breast milk of 147 HIV-1-infected women to define correlates of maternal CMV replication and infant CMV acquisition. RESULTS: Although few women had detectable CMV in plasma (4.8%), the majority had detectable CMV DNA in cervical secretions (66%) and breast milk (99%). There was a strong association between cervical CMV detection during pregnancy and later breast milk levels (ß = 0.47; P = .005). Plasma HIV-1 level and CD4 counts were associated with CMV in the cervix and breast milk. However HIV-1 levels within the cervix and breast milk were not associated with CMV within these compartments. Maternal breast milk CMV levels (hazard ratio [HR], 1.4; P = .003) and maternal CD4 < 450 cells/mm(3) (HR, 1.8; P = .008) were independently associated with infant CMV acquisition; each log10 increase in breast milk CMV was associated with a 40% increase in infant infection. The breast milk CMV level required to attain a 50% probability of CMV transmission increased with higher maternal CD4 counts, increasing from 3.55 log10 CMV DNA copies/mL at a CD4 count of 350 cells/mm(3) to 5.50 log10 CMV DNA copies/mL at a CD4 count of 1000 cells/mm(3). CONCLUSIONS: Breast milk CMV levels and maternal CD4 count are major determinants of CMV transmission in the setting of maternal HIV-1. Maternal immune reconstitution or lowering breast milk CMV levels may reduce vertical CMV transmission.


Assuntos
Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/transmissão , Citomegalovirus/isolamento & purificação , Infecções por HIV/complicações , Transmissão Vertical de Doenças Infecciosas , Complicações Infecciosas na Gravidez/virologia , Contagem de Linfócito CD4 , Colo do Útero/virologia , Infecções por Citomegalovirus/virologia , Feminino , Infecções por HIV/virologia , HIV-1/isolamento & purificação , Humanos , Lactente , Recém-Nascido , Leite Humano/virologia , Plasma/virologia , Gravidez , Carga Viral
17.
Eur J Immunol ; 43(4): 886-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23592382

RESUMO

Cytomegalovirus (CMV) infection is widespread in the human population. Normally, in adolescents and adults, prior exposure to CMV can readily be determined by IgG assays. However, in individuals where antibody production is impaired, such as patients with common variable immunodeficiency disease or in the very young where maternal antibodies are present, diagnosis of CMV infection is problematic using such assays. In this issue of the European Journal of Immunology, a study by Sester and colleagues [Eur J Immunol 2013. 43: 1099-1108] using CD4(+) T-cell immunity as a marker of infection clearly differentiates young children with prior exposure to CMV from those who only have passive maternal antibody. This information will quickly find application in the pretransplant screening of young children for CMV infection and help with the stratification of these children to identify those who are truly CMV negative and are therefore at risk for future CMV infection and disease if receiving an organ from a CMV-positive donor as discussed in this Commentary.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Imunidade Materno-Adquirida , Subpopulações de Linfócitos T/imunologia , Feminino , Humanos , Masculino
18.
Clin Microbiol Rev ; 26(1): 86-102, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23297260

RESUMO

Human cytomegalovirus (CMV) is a leading cause of congenital infections worldwide. In the developed world, following the virtual elimination of circulating rubella, it is the commonest nongenetic cause of childhood hearing loss and an important cause of neurodevelopmental delay. The seroprevalence of CMV in adults and the incidence of congenital CMV infection are highest in developing countries (1 to 5% of births) and are most likely driven by nonprimary maternal infections. However, reliable estimates of prevalence and outcome from developing countries are not available. This is largely due to the dogma that maternal preexisting seroimmunity virtually eliminates the risk for sequelae. However, recent data demonstrating similar rates of sequelae, especially hearing loss, following primary and nonprimary maternal infection have underscored the importance of congenital CMV infection in resource-poor settings. Although a significant proportion of congenital CMV infections are attributable to maternal primary infection in well-resourced settings, the absence of specific interventions for seronegative mothers and uncertainty about fetal prognosis have discouraged routine maternal antibody screening. Despite these challenges, encouraging results from prototype vaccines have been reported, and the first randomized phase III trials of prenatal interventions and prolonged postnatal antiviral therapy are under way. Successful implementation of strategies to prevent or reduce the burden of congenital CMV infection will require heightened global awareness among clinicians and the general population. In this review, we highlight the global epidemiology of congenital CMV and the implications of growing knowledge in areas of prevention, diagnosis, prognosis, and management for both low (50 to 70%)- and high (>70%)-seroprevalence settings.


Assuntos
Infecções por Citomegalovirus/congênito , Infecções por Citomegalovirus/epidemiologia , Antivirais/uso terapêutico , Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/administração & dosagem , Vacinas contra Citomegalovirus/imunologia , Saúde Global , Perda Auditiva/epidemiologia , Perda Auditiva/etiologia , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto
19.
Rev Med Virol ; 23(2): 97-125, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23165654

RESUMO

Recent evidence suggesting a potential anti-CMV effect of mTORis is of great interest to the transplant community. However, the concept of an immunosuppressant with antiviral properties is not new, with many accounts of the antiviral properties of several agents over the years. Despite these reports, to date, there has been little effort to collate the evidence into a fuller picture. This manuscript was developed to gather the evidence of antiviral activity of the agents that comprise a typical immunosuppressive regimen against viruses that commonly reactivate following transplant (HHV1 and 2, VZV, EBV, CMV and HHV6, 7, and 8, HCV, HBV, BKV, HIV, HPV, and parvovirus). Appropriate immunosuppressive regimens posttransplant that avoid acute rejection while reducing risk of viral reactivation are also reviewed. The existing literature was disparate in nature, although indicating a possible stimulatory effect of tacrolimus on BKV, potentiation of viral reactivation by steroids, and a potential advantage of mammalian target of rapamycin (mTOR) inhibition in several viral infections, including BKV, HPV, and several herpesviruses.


Assuntos
Imunossupressores/administração & dosagem , Ativação Viral/efeitos dos fármacos , Viroses/induzido quimicamente , Viroses/prevenção & controle , Humanos , Imunossupressores/efeitos adversos , Imunossupressores/farmacologia , Transplante , Viroses/virologia
20.
J Virol ; 86(20): 11373-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22875969

RESUMO

Cytomegalovirus (CMV) coinfection is associated with infant HIV-1 disease progression and mortality. In a cohort of Kenyan HIV-infected infants, the frequencies of activated (CD38(+) HLA-DR(+)) and apoptosis-vulnerable (CD95(+) Bcl-2(-)) CD4(+) and CD8(+) T cells increased substantially during acute CMV infection. The frequency of activated CD4(+) T cells was strongly associated with both concurrent CMV coinfection (P = 0.001) and HIV-1 viral load (P = 0.05). The frequency of apoptosis-vulnerable cells was also associated with CMV coinfection in the CD4 (P = 0.02) and CD8 (P < 0.001) T cell subsets. Similar observations were made in HIV-exposed uninfected infants. CMV-induced increases in T cell activation and apoptosis may contribute to the rapid disease progression in coinfected infants.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/imunologia , Infecções por HIV/complicações , Infecções por HIV/imunologia , HIV-1 , Ativação Linfocitária , ADP-Ribosil Ciclase 1/análise , Apoptose , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Coinfecção , Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Progressão da Doença , Infecções por HIV/virologia , HIV-1/imunologia , Antígenos HLA-DR/análise , Humanos , Lactente , Quênia , Proteínas Proto-Oncogênicas c-bcl-2/análise , Carga Viral , Receptor fas/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...