Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 212(2): 245-257, 2024 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-38047900

RESUMEN

CD8 T cells are emerging as important mediators in atherosclerosis and cardiovascular disease (CVD). Immune activation may play a particular role in people with HIV (PWH) who are at an increased risk of CVD, even after controlling for known CVD risk factors. Latent CMV infection is associated with increased CVD risk for both PWH and people without HIV, and human CMV-specific CD4 and CD8 T cells are enriched for an immunosenescent phenotype. We previously showed that CMV coinfection in PWH promotes vascular homing and activation of inflammatory CD4 T cells through the CD2-LFA-3 axis. However, the role of CD2/LFA3 costimulation of CD8 T cells in PWH with CMV has yet to be described. In the present study, we demonstrate that CD2 expression on CX3CR1+CD57+CD28- inflammescent CD8 T cells is increased on cells from CMV-seropositive PWH. In vitro CD2/LFA-3 costimulation enhances TCR-mediated activation of these inflammatory CD8 memory T cells. Finally, we show that LFA-3 is highly expressed in aortas of SIV-infected rhesus macaques and in atherosclerotic plaques of people without HIV. Our findings are consistent with a model in which CMV infection enhances CD2 expression on highly proinflammatory CD8 T cells that can then be stimulated by LFA-3 expressed in the vasculature, even in the absence of CD28 costimulation. This model, in which CMV infection exacerbates toxic cytokine and granzyme production by CD8 T cells within the vasculature, highlights a potential therapeutic target in atherosclerosis development and progression, especially for PWH.


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Infecciones por Citomegalovirus , Infecciones por VIH , Animales , Humanos , Antígenos CD28/metabolismo , Infecciones por VIH/tratamiento farmacológico , Citomegalovirus , Antígenos CD58/metabolismo , Macaca mulatta , Linfocitos T CD8-positivos , Linfocitos T CD4-Positivos , Aterosclerosis/metabolismo
2.
EMBO J ; 39(24): e107167, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33438774

RESUMEN

While HIV-1 infects T but not B cells, it nevertheless impairs the function of B cells and thereby contribute to the failure to produce neutralizing antibodies. In this issue, Kaw et al describe the mechanisms leading to this failure and report a key role for the HIV-1 protein NEF in preventing B-cell maturation into antibody-producing plasma cells.


Asunto(s)
Infecciones por VIH , VIH-1 , Linfocitos B , Linfocitos T CD4-Positivos , Humanos
3.
J Infect Dis ; 227(12): 1381-1385, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-36790818

RESUMEN

Plasma extracellular vesicle (EV)-associated cytokines were quantified in people with HIV (PWH) with different virological control status, including elite controllers (EC) who maintain persistent control (PC) or not (TC). Cytokine signatures and pathways were determined for each group. Median EV-associated cytokine levels were higher among PWH than HIV-uninfected. EC showed the highest levels of EV-associated cytokines among PWH with PC levels higher than TC levels. IL-18 levels best distinguished PWH from uninfected controls, and EC from ART-treated, and IL-3 distinguished PC from TC. The role of EV-cytokines in intercellular communication and endogenous control of HIV expression should be investigated further.


Asunto(s)
Vesículas Extracelulares , Infecciones por VIH , Seropositividad para VIH , VIH-1 , Humanos , VIH-1/metabolismo , Interleucina-18/metabolismo , Interleucina-3 , Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Biomarcadores , Vesículas Extracelulares/metabolismo
4.
PLoS Pathog ; 17(5): e1009508, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33984071

RESUMEN

Both gram-negative and gram-positive bacteria release extracellular vesicles (EVs) that contain components from their mother cells. Bacterial EVs are similar in size to mammalian-derived EVs and are thought to mediate bacteria-host communications by transporting diverse bioactive molecules including proteins, nucleic acids, lipids, and metabolites. Bacterial EVs have been implicated in bacteria-bacteria and bacteria-host interactions, promoting health or causing various pathologies. Although the science of bacterial EVs is less developed than that of eukaryotic EVs, the number of studies on bacterial EVs is continuously increasing. This review highlights the current state of knowledge in the rapidly evolving field of bacterial EV science, focusing on their discovery, isolation, biogenesis, and more specifically on their role in microbiota-host communications. Knowledge of these mechanisms may be translated into new therapeutics and diagnostics based on bacterial EVs.


Asunto(s)
Bacterias/metabolismo , Vesículas Extracelulares/metabolismo , Interacciones Microbiota-Huesped , Animales , Humanos
5.
J Infect Dis ; 225(6): 1040-1049, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32603406

RESUMEN

BACKGROUND: The role of extracellular vesicles (EVs) in human immunodeficiency virus (HIV) pathogenesis is unknown. We examine the cellular origin of plasma microvesicles (MVs), a type of ectocytosis-derived EV, the presence of mitochondria in MVs, and their relationship to circulating cell-free mitochondrial deoxyribonucleic acid (ccf-mtDNA) in HIV-infected patients and controls. METHODS: Five participant groups were defined: 30 antiretroviral therapy (ART)-naive; 30 ART-treated with nondetectable viremia; 30 elite controllers; 30 viremic controllers; and 30 HIV-uninfected controls. Microvesicles were quantified and characterized from plasma samples by flow cytometry. MitoTrackerDeepRed identified MVs containing mitochondria and ccf-mtDNA was quantified by real-time polymerase chain reaction. RESULTS: Microvesicle numbers were expanded at least 10-fold in all HIV-infected groups compared with controls. More than 79% were platelet-derived MVs. Proportions of MVs containing mitochondria (22.3% vs 41.6%) and MV mitochondrial density (706 vs 1346) were significantly lower among HIV-infected subjects than controls, lowest levels for those on ART. Microvesicle numbers correlated with ccf-mtDNA levels that were higher among HIV-infected patients. CONCLUSIONS: A massive release of platelet-derived MVs occurs during HIV infection. Some MVs contain mitochondria, but their proportion and mitochondrial densities were lower in HIV infection than in controls. Platelet-derived MVs may be biomarkers of platelet activation, possibly reflecting pathogenesis even in absence of HIV replication.


Asunto(s)
Micropartículas Derivadas de Células , Vesículas Extracelulares , Infecciones por VIH , ADN Mitocondrial , Humanos , Tetraspanina 29 , Viremia
6.
Eur J Immunol ; 51(7): 1857-1859, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33772780

RESUMEN

T-cell accumulation in atherosclerotic plaques contributes to plaque destabilization. We found that several chemokine receptors are differentially expressed on peripheral blood compared to plaque-resident T cells and corresponding ligands are upregulated in plaques. These data indicate that T-cell migration into human atherosclerotic plaques may predominantly occur via CCR5-CCL3 and CX3CR1-CX3CL1 interactions.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Movimiento Celular/fisiología , Quimiocina CCL3/metabolismo , Quimiocina CX3CL1/metabolismo , Placa Aterosclerótica/metabolismo , Receptores CCR5/metabolismo , Linfocitos T/metabolismo , Aterosclerosis/metabolismo , Células Cultivadas , Humanos
7.
PLoS Pathog ; 16(9): e1008885, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32976527

RESUMEN

Atherosclerotic cardiovascular disease (ASCVD) remains an important cause of morbidity in the general population and risk for ASCVD is increased approximately 2-fold in persons living with HIV infection (PLWH). This risk is linked to elevated CD8 T cell counts that are abundant in atherosclerotic plaques and have been implicated in disease pathogenesis yet the mechanisms driving T cell recruitment to and activation within plaques are poorly defined. Here we investigated the role of CD8 T cells in atherosclerosis in a non-human primate model of HIV infection and in the HIV-uninfected elderly; we sought to identify factors that promote the activation, function, and recruitment to endothelium of CX3CR1+ CD8 T cells. We measured elevated expression of CX3CL1 and IL-15, and increased CD8 T cell numbers in the aortas of rhesus macaques infected with SIV or SHIV, and demonstrated similar findings in atherosclerotic vessels of HIV-uninfected humans. We found that recombinant TNF enhanced the production and release of CX3CL1 and bioactive IL-15 from aortic endothelial cells, but not from aortic smooth muscle cells. IL-15 in turn promoted CX3CR1 surface expression on and TNF synthesis by CD8 T cells, and IL-15-treated CD8 T cells exhibited enhanced CX3CL1-dependent chemoattraction toward endothelial cells in vitro. Finally, we show that CD8 T cells in human atherosclerotic plaques have an activated, resident phenotype consistent with in vivo IL-15 and CX3CL1 exposure. In this report, we define a novel model of CD8 T cell involvement in atherosclerosis whereby CX3CL1 and IL-15 operate in tandem within the vascular endothelium to promote infiltration by activated CX3CR1+ memory CD8 T cells that drive further endothelial activation via TNF. We propose that these interactions are prevalent in aging and in PLWH, populations where circulating activated CX3CR1+ CD8 T cell numbers are often expanded.


Asunto(s)
Aterosclerosis/metabolismo , Linfocitos T CD8-positivos/metabolismo , Quimiocina CX3CL1/metabolismo , Infecciones por VIH/metabolismo , Interleucina-15/metabolismo , Anciano , Animales , Células Endoteliales/metabolismo , Humanos , Macaca mulatta/metabolismo , Receptores de Quimiocina/metabolismo
8.
PLoS Biol ; 17(7): e3000363, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31318874

RESUMEN

For many years, double-layer phospholipid membrane vesicles, released by most cells, were not considered to be of biological significance. This stance has dramatically changed with the recognition of extracellular vesicles (EVs) as carriers of biologically active molecules that can traffic to local or distant targets and execute defined biological functions. The dimensionality of the field has expanded with the appreciation of diverse types of EVs and the complexity of vesicle biogenesis, cargo loading, release pathways, targeting mechanisms, and vesicle processing. With the expanded interest in the field and the accelerated rate of publications on EV structure and function in diverse biomedical fields, it has become difficult to distinguish between well-established biological features of EV and the untested hypotheses or speculative assumptions that await experimental proof. With the growing interest despite the limited evidence, we sought in this essay to formulate a set of unsolved mysteries in the field, sort out established data from fascinating hypotheses, and formulate several challenging questions that must be answered for the field to advance.


Asunto(s)
Comunicación Celular/fisiología , Membrana Celular/metabolismo , Vesículas Extracelulares/metabolismo , Modelos Biológicos , Animales , Transporte Biológico/fisiología , Endocitosis/fisiología , Exocitosis/fisiología , Vesículas Extracelulares/clasificación , Humanos , Tamaño de la Partícula
9.
J Immunol ; 204(10): 2722-2733, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32229536

RESUMEN

Cytotoxic CD4 T cells are linked to cardiovascular morbidities and accumulate in both HIV and CMV infections, both of which are associated with increased risk of cardiovascular disease (CVD). In this study, we identify CMV coinfection as a major driver of the cytotoxic phenotype, characterized by elevated CD57 expression and reduced CD28 expression, in circulating CD4 T cells from people living with HIV infection, and investigate potential mechanisms linking this cell population to CVD. We find that human CD57+ CD4 T cells express high levels of the costimulatory receptor CD2 and that CD2/LFA-3 costimulation results in a more robust and polyfunctional effector response to TCR signals, compared with CD28-mediated costimulation. CD57+ CD4 T cells also express the vascular endothelium-homing receptor CX3CR1 and migrate toward CX3CL1-expressing endothelial cells in vitro. IL-15 promotes the cytotoxic phenotype, elevates CX3CR1 expression, and enhances the trafficking of CD57+ CD4 T cells to endothelium and may therefore be important in linking these cells to cardiovascular complications. Finally, we demonstrate the presence of activated CD57+ CD4 T cells and expression of CX3CL1 and LFA-3 in atherosclerotic plaque tissues from HIV-uninfected donors. Our findings are consistent with a model in which cytotoxic CD4 T cells contribute to CVD in HIV/CMV coinfection and in atherosclerosis via CX3CR1-mediated trafficking and CD2/LFA-3-mediated costimulation. This study identifies several targets for therapeutic interventions and may help bridge the gap in understanding how CMV infection and immunity are linked to increased cardiovascular risk in people living with HIV infection.


Asunto(s)
Vasos Sanguíneos/fisiología , Linfocitos T CD4-Positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/fisiología , Infecciones por VIH/inmunología , VIH-1/fisiología , Placa Aterosclerótica/inmunología , Antígenos CD28/metabolismo , Antígenos CD57/metabolismo , Antígenos CD58/metabolismo , Receptor 1 de Quimiocinas CX3C/metabolismo , Movimiento Celular , Quimiocina CX3CL1/metabolismo , Coinfección , Citotoxicidad Inmunológica , Humanos , Receptores CXCR3/metabolismo , Riesgo
10.
Int J Mol Sci ; 23(14)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35887283

RESUMEN

Coronavirus disease 2019 (COVID-19) is characterized by immune activation in response to viral spread, in severe cases leading to the development of cytokine storm syndrome (CSS) and increased mortality. Despite its importance in prognosis, the pathophysiological mechanisms of CSS in COVID-19 remain to be defined. Towards this goal, we analyzed cytokine profiles and their interrelation in regard to anti-cytokine treatment with tocilizumab in 98 hospitalized patients with COVID-19. We performed a multiplex measurement of 41 circulating cytokines in the plasma of patients on admission and 3-5 days after, during the follow-up. Then we analyzed the patient groups separated in two ways: according to the clusterization of their blood cytokines and based on the administration of tocilizumab therapy. Patients with and without CSS formed distinct clusters according to their cytokine concentration changes. However, the tocilizumab therapy, administered based on the standard clinical and laboratory criteria, did not fully correspond to those clusters of CSS. Furthermore, among all cytokines, IL-6, IL-1RA, IL-10, and G-CSF demonstrated the most prominent differences between patients with and without clinical endpoints, while only IL-1RA was prognostically significant in both groups of patients with and without tocilizumab therapy, decreasing in the former and increasing in the latter during the follow-up period. Thus, CSS in COVID-19, characterized by a correlated release of multiple cytokines, does not fully correspond to the standard parameters of disease severity. Analysis of the cytokine signature, including the IL-1RA level in addition to standard clinical and laboratory parameters may be useful to define the onset of a cytokine storm in COVID-19 as well as the indications for anti-cytokine therapy.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Anticuerpos Monoclonales Humanizados , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Citocinas , Humanos , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-6 , SARS-CoV-2
11.
Clin Infect Dis ; 71(10): 2655-2662, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31768525

RESUMEN

BACKGROUND: Seminal human immunodeficiency virus (HIV) transmission from men to their partners remains the main driver of HIV epidemics worldwide. Semen is not merely a carrier of the virus, but also provides an immunological milieu that affects HIV transmission. METHODS: We collected blood and semen from people with HIV whose epidemiologically linked sexual partners either did or did not acquire HIV. Viral transmission was confirmed by phylogenetic linkage (HIV pol). We measured the concentration of 34 cytokines/chemokines by Luminex in the blood and semen of 21 source partners who transmitted HIV (transmitters) and 22 who did not transmit HIV (nontransmitters) to their sexual partners. Differences between cytokine profiles in transmitters versus nontransmitters were analyzed using the multivariate statistical technique of partial least square discriminant analysis. RESULTS: The cytokine profile in seminal fluid, but not in peripheral blood, was significantly different between men who have sex with men (MSM) who transmitted HIV and those who did not transmit HIV to their sexual partners (E = 19.77; P < .01). This difference persisted after excluding people with undetectable HIV RNA levels in nontransmitters. CONCLUSIONS: Seminal cytokine profiles correlated with transmission or nontransmission of HIV from the infected MSM to their partners, independently from seminal viral load. Seminal cytokine spectra might be a contributing determinant of sexual HIV transmission, thus providing new directions for the development of strategies aimed at preventing HIV transmission.


Asunto(s)
Citocinas/inmunología , Infecciones por VIH/inmunología , Minorías Sexuales y de Género , Infecciones por VIH/transmisión , VIH-1 , Homosexualidad Masculina , Humanos , Masculino , Filogenia
12.
Proc Natl Acad Sci U S A ; 113(33): 9155-61, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27432966

RESUMEN

Extracellular vesicles (EVs) released by various cells are small phospholipid membrane-enclosed entities that can carry miRNA. They are now central to research in many fields of biology because they seem to constitute a new system of cell-cell communication. Physical and chemical characteristics of many EVs, as well as their biogenesis pathways, resemble those of retroviruses. Moreover, EVs generated by virus-infected cells can incorporate viral proteins and fragments of viral RNA, being thus indistinguishable from defective (noninfectious) retroviruses. EVs, depending on the proteins and genetic material incorporated in them, play a significant role in viral infection, both facilitating and suppressing it. Deciphering the mechanisms of EV-cell interactions may facilitate the design of EVs that inhibit viral infection and can be used as vehicles for targeted drug delivery.


Asunto(s)
Vesículas Extracelulares/fisiología , Virosis/etiología , Desaminasa APOBEC-3G/fisiología , Animales , Exosomas/fisiología , Humanos , MicroARNs/fisiología , Virión/fisiología , Virosis/terapia
13.
J Virol ; 90(21): 9833-9840, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27558419

RESUMEN

Unprotected sexual intercourse with HIV-infected men is the major cause of new infections. HIV virions are released into semen by various cells of the male genital tract, as well as by infected monocytes and lymphocytes present in semen. Some of these virions may attach to the surfaces of cells, infected or uninfected. We investigated whether cells carrying attached HIV on their surfaces can transmit infection. We addressed this question in a model system of human tissue exposed ex vivo to monocytes and lymphocytes carrying HIV on their surfaces. We gamma irradiated the cells to prevent their productive infection. In spite of comparable amounts of HIV attached to monocytes and lymphocytes, only monocytes were capable of transmitting infection and triggering productive infection in tissue. This HIV-1 transmission was mediated by cell-cell contacts. Our experiments suggest that in vivo, HIV attached to infected or uninfected monocytes, which far outnumber lymphocytes in HIV-infected semen, may contribute to sexual transmission of HIV from men to their partners. IMPORTANCE: The vast majority of new HIV infections occur through sexual transmission, in which HIV is transferred from the semen of an infected male to an uninfected partner. In semen, HIV-1 particles may exist as free-floating virions; inside infected cells; or attached to the surfaces of cells, whether they are infected or not. Here, we investigated whether HIV attached to the surfaces of monocytes or lymphocytes could transmit infection to human tissue. Incubation of human tissue with monocyte-attached HIV resulted in productive tissue infection. In contrast, there was no infection of tissues when they were incubated with lymphocyte-attached HIV-1. Our results highlight the important role that seminal monocytes may play in HIV transmission in vivo, especially since monocytes far outnumber lymphocytes in the semen of HIV-infected individuals.


Asunto(s)
Infecciones por VIH/transmisión , Linfocitos/virología , Monocitos/virología , Línea Celular , Línea Celular Tumoral , Femenino , Infecciones por VIH/virología , VIH-1 , Humanos
14.
Platelets ; 28(2): 165-173, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27595614

RESUMEN

A diverse population of small extracellular vesicles (EVs) that are released by various cells has been characterized predominantly in bulk, a procedure whereby the individual characteristics of EVs are lost. Here, we used a new nanotechnology-based flow cytometric analysis to characterize the antigenic composition of individual EVs in patients with acute coronary syndrome (ACS). Plasma EVs were captured with 15-nm magnetic nanoparticles coupled to antibodies against CD31 (predominantly an endothelial marker), CD41a (a marker for platelets), and CD63 or MHC class I (common EV markers). The total amounts of EVs were higher in the ACS patients than in the controls, predominantly due to the contribution of patients with acute myocardial infarction. For all captured fractions, the differences in the EV amounts were restricted to CD41a+ EVs. The increase in the numbers of EVs in the ACS patients, predominantly of platelet origin, probably reflects platelet activation and may indicate disease progression.


Asunto(s)
Síndrome Coronario Agudo/metabolismo , Plaquetas/metabolismo , Vesículas Extracelulares/metabolismo , Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/diagnóstico , Anciano , Antígenos CD/metabolismo , Biomarcadores , Estudios de Casos y Controles , Electrocardiografía , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/sangre , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/metabolismo
15.
Clin Infect Dis ; 60(11): 1708-14, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25740794

RESUMEN

BACKGROUND: Acyclovir (ACV), a highly specific anti-herpetic drug, acts as a DNA chain terminator for several human herpesviruses (HHVs), including HHV-2 (HSV-2), a common human immunodeficiency virus (HIV)-1 co-pathogen. Several trials demonstrated that HSV-2 suppressive therapy using ACV or its prodrug valacyclovir (valACV) reduced plasma HIV-1 viral load (VL) in HIV-1/HSV-2 coinfected persons, and this was proposed to be due to a decrease in generalized immune activation. Recently, however, we found that ACV directly suppresses HIV-1 ex vivo in tissues free of HSV-2 but endogenously coinfected with other HHVs. Here, we asked whether valACV suppresses VL in HIV-1 infected HSV-2-seronegative persons. METHODS: Eighteen HIV-1 infected HSV-2-seronegative individuals were randomly assigned in a double blind placebo-controlled, crossover trial. Eligible participants had CD4 cell counts of ≥500 cells/µL and were not taking antiretroviral therapy. Subjects in group A received 12 weeks of valACV 500 mg given twice daily by mouth followed by 2 weeks of a no treatment washout and then 12 weeks of placebo; subjects in group B received 12 weeks of placebo followed by 2 weeks of no treatment washout and then 12 weeks of valACV 500 mg twice daily. RESULTS: HIV-1 VL in plasma of patients treated with valACV 500 mg twice daily for 12 weeks was reduced on average by 0.37 log10 copies/mL. CONCLUSIONS: These data indicate that the effects of valACV on HIV-1 replication are not related to the suppression of HSV-2-mediated inflammation and are consistent with a direct effect of ACV on HIV-1 replication.


Asunto(s)
Aciclovir/análogos & derivados , Fármacos Anti-VIH/administración & dosificación , Infecciones por VIH/virología , VIH-1/aislamiento & purificación , Plasma/virología , ARN Viral/sangre , Valina/análogos & derivados , Carga Viral , Aciclovir/administración & dosificación , Adulto , Estudios Cruzados , Femenino , Humanos , Masculino , Placebos/administración & dosificación , Valaciclovir , Valina/administración & dosificación , Replicación Viral/efectos de los fármacos , Adulto Joven
16.
PLoS Pathog ; 9(2): e1003148, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23408885

RESUMEN

The majority of HIV-1 infections in women occur through vaginal intercourse, in which virus-containing semen is deposited on the cervico-vaginal mucosa. Semen is more than a mere carrier of HIV-1, since it contains many biological factors, in particular cytokines, that may affect HIV-1 transmission. The concentration of interleukin (IL)-7, one of the most prominent cytokines in semen of healthy individuals, is further increased in semen of HIV-1-infected men. Here, we investigated the potential role of IL-7 in HIV-1 vaginal transmission in an ex vivo system of human cervico-vaginal tissue. We simulated an in vivo situation by depositing HIV-1 on cervico-vaginal tissue in combination with IL-7 at concentrations comparable with those measured in semen of HIV-1-infected individuals. We found that IL-7 significantly enhanced virus replication in ex vivo infected cervico-vaginal tissue. Similarly, we observed an enhancement of HIV-1 replication in lymphoid tissue explants. Analysis of T cells isolated from infected tissues showed that IL-7 reduced CD4⁺ T cell depletion preventing apoptosis, as shown by the decrease in the number of cells expressing the apoptotic marker APO2.7 and the increase in the expression of the anti-apoptotic protein B-cell lymphoma (Bcl)-2. Also, IL-7 increased the fraction of cycling CD4⁺ T cells, as evidenced by staining for the nuclear factor Ki-67. High levels of seminal IL-7 in vivo may be relevant to the survival of the founder pool of HIV-1-infected cells in the cervico-vaginal mucosa at the initial stage of infection, promoting local expansion and dissemination of HIV infection.


Asunto(s)
Cuello del Útero/virología , Infecciones por VIH/transmisión , VIH-1/fisiología , Interleucina-7/metabolismo , Vagina/virología , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Proliferación Celular , Femenino , Regulación Viral de la Expresión Génica , Infecciones por VIH/virología , Humanos , Antígeno Ki-67/metabolismo , Tejido Linfoide/virología , Masculino , Datos de Secuencia Molecular , Tonsila Palatina/virología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Recombinantes , Replicación Viral
17.
PLoS Pathog ; 9(7): e1003456, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935482

RESUMEN

Human immunodeficiency virus (HIV) infection is often accompanied by infection with other pathogens, in particular herpes simplex virus type 2 (HSV-2). The resulting coinfection is involved in a vicious circle of mutual facilitations. Therefore, an important task is to develop a compound that is highly potent against both viruses to suppress their transmission and replication. Here, we report on the discovery of such a compound, designated PMEO-DAPym. We compared its properties with those of the structurally related and clinically used acyclic nucleoside phosphonates (ANPs) tenofovir and adefovir. We demonstrated the potent anti-HIV and -HSV activity of this drug in a diverse set of clinically relevant in vitro, ex vivo, and in vivo systems including (i) CD4⁺ T-lymphocyte (CEM) cell cultures, (ii) embryonic lung (HEL) cell cultures, (iii) organotypic epithelial raft cultures of primary human keratinocytes (PHKs), (iv) primary human monocyte/macrophage (M/M) cell cultures, (v) human ex vivo lymphoid tissue, and (vi) athymic nude mice. Upon conversion to its diphosphate metabolite, PMEO-DAPym markedly inhibits both HIV-1 reverse transcriptase (RT) and HSV DNA polymerase. However, in striking contrast to tenofovir and adefovir, it also acts as an efficient immunomodulator, inducing ß-chemokines in PBMC cultures, in particular the CCR5 agonists MIP-1ß, MIP-1α and RANTES but not the CXCR4 agonist SDF-1, without the need to be intracellularly metabolized. Such specific ß-chemokine upregulation required new mRNA synthesis. The upregulation of ß-chemokines was shown to be associated with a pronounced downmodulation of the HIV-1 coreceptor CCR5 which may result in prevention of HIV entry. PMEO-DAPym belongs conceptually to a new class of efficient multitargeted antivirals for concomitant dual-viral (HSV/HIV) infection therapy through inhibition of virus-specific pathways (i.e. the viral polymerases) and HIV transmission prevention through interference with host pathways (i.e. CCR5 receptor down regulation).


Asunto(s)
Antivirales/farmacología , VIH/efectos de los fármacos , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Organofosfonatos/farmacología , Pirimidinas/farmacología , Animales , Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/uso terapéutico , Antivirales/uso terapéutico , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Femenino , VIH/enzimología , VIH/inmunología , Herpes Simple/tratamiento farmacológico , Herpes Simple/inmunología , Herpes Simple/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 2/enzimología , Herpesvirus Humano 2/inmunología , Humanos , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Tejido Linfoide/efectos de los fármacos , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Tejido Linfoide/virología , Ratones , Ratones Pelados , Ratones Desnudos , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Organofosfonatos/uso terapéutico , Profármacos/farmacología , Profármacos/uso terapéutico , Pirimidinas/uso terapéutico , Inhibidores de la Transcriptasa Inversa/farmacología , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Técnicas de Cultivo de Tejidos
18.
Am J Obstet Gynecol ; 213(6): 836.e1-836.e18, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26232508

RESUMEN

OBJECTIVE: Intraamniotic inflammation/infection is the only mechanism of disease with persuasive evidence of causality for spontaneous preterm labor/delivery. Previous studies about the behavior of cytokines in preterm labor have been largely based on the analysis of the behavior of each protein independently. Emerging evidence indicates that the study of biologic networks can provide insight into the pathobiology of disease and improve biomarker discovery. The goal of this study was to characterize the inflammatory-related protein network in the amniotic fluid of patients with preterm labor. STUDY DESIGN: A retrospective cohort study was conducted that included women with singleton pregnancies who had spontaneous preterm labor and intact membranes (n = 135). These patients were classified according to the results of amniotic fluid culture, broad-range polymerase chain reaction coupled with electrospray ionization mass spectrometry, and amniotic fluid concentration of interleukin (IL)-6 into the following groups: (1) those without intraamniotic inflammation (n = 85), (2) those with microbial-associated intraamniotic inflammation (n = 15), and (3) those with intraamniotic inflammation without detectable bacteria (n = 35). Amniotic fluid concentrations of 33 inflammatory-related proteins were determined with the use of a multiplex bead array assay. RESULTS: Patients with preterm labor and intact membranes who had microbial-associated intraamniotic inflammation had a higher amniotic fluid inflammatory-related protein concentration correlation than those without intraamniotic inflammation (113 perturbed correlations). IL-1ß, IL-6, macrophage inflammatory protein (MIP)-1α, and IL-1α were the most connected nodes (highest degree) in this differential correlation network (degrees of 20, 16, 12, and 12, respectively). Patients with sterile intraamniotic inflammation had correlation patterns of inflammatory-related proteins, both increased and decreased, when compared to those without intraamniotic inflammation (50 perturbed correlations). IL-1α, MIP-1α, and IL-1ß were the most connected nodes in this differential correlation network (degrees of 12, 10, and 7, respectively). There were more coordinated inflammatory-related protein concentrations in the amniotic fluid of women with microbial-associated intraamniotic inflammation than in those with sterile intraamniotic inflammation (60 perturbed correlations), with IL-4 and IL-33 having the largest number of perturbed correlations (degrees of 15 and 13, respectively). CONCLUSIONS: We report for the first time an analysis of the inflammatory-related protein network in spontaneous preterm labor. Patients with preterm labor and microbial-associated intraamniotic inflammation had more coordinated amniotic fluid inflammatory-related proteins than either those with sterile intraamniotic inflammation or those without intraamniotic inflammation. The correlations were also stronger in patients with sterile intraamniotic inflammation than in those without intraamniotic inflammation. The findings herein could be of value in the development of biomarkers of preterm labor.


Asunto(s)
Líquido Amniótico/metabolismo , Corioamnionitis/metabolismo , Citocinas/metabolismo , Trabajo de Parto Prematuro/metabolismo , Adulto , Líquido Amniótico/microbiología , Biomarcadores/metabolismo , Calgranulina A/metabolismo , Estudios de Cohortes , Femenino , Proteína HMGB1/metabolismo , Humanos , Embarazo , Estudios Retrospectivos , Adulto Joven
19.
Nanomedicine ; 11(3): 489-98, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25481806

RESUMEN

Extracellular vesicles (EVs) are important in normal physiology and are altered in various pathologies. EVs produced by different cells are antigenically different. Since the majority of EVs are too small for routine flow cytometry, EV composition is studied predominantly in bulk, thus not addressing their antigenic heterogeneity. Here, we describe a nanoparticle-based technique for analyzing antigens on single nano-sized EVs. The technique consists of immuno-capturing of EVs with 15-nm magnetic nanoparticles, staining captured EVs with antibodies against their antigens, and separating them from unbound EVs and free antibodies in a magnetic field, followed by flow analysis. This technique allows us to characterize EVs populations according to their antigenic distribution, including minor EV fractions. We demonstrated that the individual blood EVs carry different sets of antigens, none being ubiquitous, and quantified their distribution. The physiological significance of antigenically different EVs and their correlation with different pathologies can now be directly addressed. From the clinical editor: This study reports a nanoparticle-based technique for analyzing antigens on single nano-sized extracellular vehicles (EV). The technique consists of immuno-capturing of EVs with 15-nm magnetic nanoparticles, followed by staining the captured EVs with antibodies and separating them via a magnetic field, followed by flow analysis. This technique enables studies of antigenic properties of individual EVs that conventionally can only be studied in bulk.


Asunto(s)
Antígenos CD/metabolismo , Micropartículas Derivadas de Células/metabolismo , Citometría de Flujo/métodos , Humanos
20.
J Infect Dis ; 205(1): 97-105, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22107749

RESUMEN

BACKGROUND: Semen is the main carrier of sexually transmitted viruses, including human immunodeficiency virus type 1 (HIV-1). However, semen is not just a mere passive transporter of virions but also plays an active role in HIV-1 transmission through cytokines and other biological factors. METHODS: To study the relationship between viruses and the chemokine-cytokine network in the male genital tract, we measured the concentrations of 21 cytokines/chemokines and the loads of HIV-1 and of 6 herpesviruses in seminal and blood plasma from HIV-1-infected and HIV-uninfected men. RESULTS: We found that (1) semen is enriched in cytokines and chemokines that play key roles in HIV-1 infection or transmission; (2) HIV-1 infection changes the chemokine-cytokine network in semen, further enriching it in cytokines that modulate its replication; (3) HIV-1 infection is associated with Epstein-Barr virus (EBV) and cytomegalovirus (CMV) compartmentalized seminal reactivation; (4) CMV and EBV concomitant seminal shedding is associated with higher HIV-1 loads in blood and seminal plasma; and (5) CMV seminal reactivation increases the seminal levels of the CCR5 ligands RANTES and eotaxin, and of the CXCR3 ligand monokine induced by gamma interferon (MIG). CONCLUSIONS: HIV-1 infection results in an aberrant production of cytokines and reactivation of EBV and CMV that further changes the seminal cytokine network. The altered seminal milieu in HIV-1 infection may be a determinant of HIV-1 sexual transmission.


Asunto(s)
Quimiocinas/metabolismo , Citocinas/metabolismo , VIH-1/aislamiento & purificación , Herpesviridae/aislamiento & purificación , Semen/virología , Esparcimiento de Virus , Adulto , Coinfección/inmunología , Coinfección/metabolismo , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Infecciones por Herpesviridae/complicaciones , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Humanos , Masculino , Semen/inmunología , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA