Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Addict Biol ; 28(12): e13350, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38017645

RESUMEN

The aim of this study is to determine if extended-release, bioabsorbable, subcutaneous naltrexone (NTX) implants inhibit respiratory depression after an IV injection of fentanyl. Bioabsorbable implants fabricated from two different release-controlling polymers, poly-D-L-lactide (PDLLA) and polycaprolactone (PCL), alone (placebo) or containing NTX, were subcutaneously implanted in Sprague Dawley rats. After 3.5 months of implantation, the rodents were administered an IV bolus of fentanyl through the tail vein. The placebo implant rats received a dose of 4 micrograms (mcg) - (10 mcg/kg/dose), while the NTX implanted animals received a dose of 8 mcg (20 mcg/kg/dose). The minimum active dose of fentanyl that caused a > 50 ± 2% depression in the respiration rate in the placebo implanted rodents was 4 mcg. The respiration rate of the placebo implanted rats dropped from 208 ± 14 breaths/minute at predose, to 84 ± 12 breaths/minute (p = 0.0003) at 2 min. In contrast, all NTX implanted animals easily tolerated twice the dose of 8 mcg of fentanyl without any significant reduction in respiration rate. The mean respiration rate = increased from 164 ± 22 breaths/minute at predose to 178 ± 17 breaths/minute (p = 0.24) at 2 min. The mean plasma concentrations of NTX, 3.5 months after implantation, ranged from 7.4 (±1.1) ng/mL to 80.3 (±37.5) ng/mL. Bioabsorbable implants containing NTX effectively blocked fentanyl-induced respiratory depression in rodents as compared with placebo implants, 3.5 months after implantation.


Asunto(s)
Naltrexona , Insuficiencia Respiratoria , Animales , Ratas , Implantes Absorbibles , Fentanilo/toxicidad , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Ratas Sprague-Dawley , Roedores
2.
PLoS One ; 12(1): e0169052, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28046007

RESUMEN

SAMHD1 hydrolyzes 2'-deoxynucleoside-5'-triphosphates (dNTPs) into 2'-deoxynucleosides and inorganic triphosphate products. In this paper, we evaluated the impact of 2' sugar moiety substitution for different nucleotides on being substrates for SAMHD1 and mechanisms of actions for the results. We found that dNTPs ((2'R)-2'-H) are only permissive in the catalytic site of SAMHD1 due to L150 exclusion of (2'R)-2'-F and (2'R)-2'-OH nucleotides. However, arabinose ((2'S)-2'-OH) nucleoside-5'-triphosphates analogs are permissive to bind in the catalytic site and be hydrolyzed by SAMHD1. Moreover, when the (2'S)-2' sugar moiety is increased to a (2'S)-2'-methyl as with the SMDU-TP analog, we detect inhibition of SAMHD1's dNTPase activity. Our computational modeling suggests that (2'S)-2'-methyl sugar moiety clashing with the Y374 of SAMHD1. We speculate that SMDU-TP mechanism of action requires that the analog first docks in the catalytic pocket of SAMHD1 but prevents the A351-V378 helix conformational change from being completed, which is needed before hydrolysis can occur. Collectively we have identified stereoselective 2' substitutions that reveal nucleotide substrate specificity for SAMHD1, and a novel inhibitory mechanism for the dNTPase activity of SAMHD1. Importantly, our data is beneficial for understanding if FDA-approved antiviral and anticancer nucleosides are hydrolyzed by SAMHD1 in vivo.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/metabolismo , Animales , Trifosfato de Arabinofuranosil Citosina , Carbohidratos/química , Pollos , Humanos , Hidrólisis , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Modelos Moleculares , Monocitos/citología , Nucleótidos/metabolismo , Multimerización de Proteína/efectos de los fármacos , Proteína 1 que Contiene Dominios SAM y HD , Especificidad por Sustrato/efectos de los fármacos
3.
Chem Rev ; 116(23): 14379-14455, 2016 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-27960273

RESUMEN

Nucleoside, nucleotide, and base analogs have been in the clinic for decades to treat both viral pathogens and neoplasms. More than 20% of patients on anticancer chemotherapy have been treated with one or more of these analogs. This review focuses on the chemical synthesis and biology of anticancer nucleoside, nucleotide, and base analogs that are FDA-approved and in clinical development since 2000. We highlight the cellular biology and clinical biology of analogs, drug resistance mechanisms, and compound specificity towards different cancer types. Furthermore, we explore analog syntheses as well as improved and scale-up syntheses. We conclude with a discussion on what might lie ahead for medicinal chemists, biologists, and physicians as they try to improve analog efficacy through prodrug strategies and drug combinations.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Nucleósidos/química , Nucleótidos/química , Antineoplásicos/química , Línea Celular , Humanos
4.
Virology ; 495: 92-100, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27183329

RESUMEN

SAMHD1 limits HIV-1 infection in non-dividing myeloid cells by decreasing intracellular dNTP pools. HIV-1 restriction by SAMHD1 in these cells likely prevents activation of antiviral immune responses and modulates viral pathogenesis, thus highlighting a critical role of SAMHD1 in HIV-1 physiopathology. Here, we explored the function of SAMHD1 in regulating cell proliferation, cell cycle progression and apoptosis in monocytic THP-1 cells. Using the CRISPR/Cas9 technology, we generated THP-1 cells with stable SAMHD1 knockout. We found that silencing of SAMHD1 in cycling cells stimulates cell proliferation, redistributes cell cycle population in the G1/G0 phase and reduces apoptosis. These alterations correlated with increased dNTP levels and more efficient HIV-1 infection in dividing SAMHD1 knockout cells relative to control. Our results suggest that SAMHD1, through its dNTPase activity, affects cell proliferation, cell cycle distribution and apoptosis, and emphasize a key role of SAMHD1 in the interplay between cell cycle regulation and HIV-1 infection.


Asunto(s)
Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Monocitos/metabolismo , Monocitos/virología , Proteínas de Unión al GTP Monoméricas/metabolismo , Apoptosis/genética , Biomarcadores , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Ciclo Celular/genética , Línea Celular , Proliferación Celular , Técnicas de Inactivación de Genes , Silenciador del Gen , Infecciones por VIH/genética , Humanos , Proteínas de Unión al GTP Monoméricas/genética , Proteína 1 que Contiene Dominios SAM y HD
5.
Virology ; 491: 106-14, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26895248

RESUMEN

We evaluated cellular metabolism profiles of HIV-1 and HIV-2 infected primary human monocyte-derived macrophages (MDMs). First, HIV-2 GL-AN displays faster production kinetics and greater amounts of virus as compared to HIV-1s: YU-2, 89.6 and JR-CSF. Second, quantitative LC-MS/MS metabolomics analysis demonstrates very similar metabolic profiles in glycolysis and TCA cycle metabolic intermediates between HIV-1 and HIV-2 infected macrophages, with a few notable exceptions. The most striking metabolic change in MDMs infected with HIV-2 relative to HIV-1-infected MDMs was the increased levels of quinolinate, a metabolite in the tryptophan catabolism pathway that has been linked to HIV/AIDS pathogenesis. Third, both HIV-1 and HIV-2 infected MDMs showed elevated levels of ribose-5-phosphate, a key metabolic component in nucleotide biosynthesis. Finally, HIV-2 infected MDMs display increased dNTP concentrations as predicted by Vpx-mediated SAMHD1 degradation. Collectively, these data show differential metabolic changes during HIV-1 and HIV-2 infection of macrophages.


Asunto(s)
Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , VIH-2/fisiología , Macrófagos/metabolismo , Metaboloma , Monocitos/metabolismo , Células Cultivadas , Ciclo del Ácido Cítrico , Glucólisis , Infecciones por VIH/genética , Humanos , Macrófagos/virología , Monocitos/virología
6.
Methods Mol Biol ; 1354: 61-70, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26714705

RESUMEN

Deoxynucleoside triphosphates (dNTPs) are the building blocks of DNA and their biosynthesis is tightly regulated in the cell. HPLC-MS and enzyme-based methods are currently employed to determine dNTP concentrations from cellular extracts. Here, we describe a highly efficient, HIV-1 reverse transcriptase (RT)-based assay to quantitate dNTP concentrations. The assay is based on the ability of HIV-1 RT to function at very low dNTP concentrations, thus providing for the high sensitivity of detection.


Asunto(s)
Desoxirribonucleótidos/análisis , Transcriptasa Inversa del VIH/metabolismo , VIH-1/enzimología , Fosfatos/análisis , Desoxirribonucleótidos/metabolismo , Electroforesis en Gel de Poliacrilamida/métodos , Pruebas de Enzimas/métodos , Humanos , Fosfatos/metabolismo
7.
Virology ; 485: 313-21, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26319213

RESUMEN

Vpx encoded by HIV-2 and SIVsm enhances retroviral reverse transcription in macrophages in vitro by mediating the degradation of the host SAMHD1 protein that hydrolyzes dNTPs and by elevating cellular dNTP levels. Here we employed RT-SHIV constructs (SIV encoding HIV-1 RT) to investigate the contribution of Vpx to the potency of NRTIs, which compete against dNTPs, in monocyte-derived macrophages (MDMs) and activated CD4(+) T cells. Relative to HIV-1, both SIV and RT-SHIV exhibited reduced sensitivities to AZT, 3TC and TDF in MDMs but not in activated CD4(+) T cells. However, when SIV and RT-SHIV constructs not coding for Vpx were utilized, we observed greater sensitivities to all NRTIs tested using activated CD4(+) T cells relative to the Vpx-coding counterparts. This latter phenomenon was observed for AZT only when using MDMs. Our data suggest that Vpx in RT-SHIVs may underestimate the antiviral efficacy of NRTIs in a cell type dependent manner.


Asunto(s)
Fármacos Anti-VIH/farmacología , Linfocitos T CD4-Positivos/virología , VIH-1/fisiología , Macrófagos/virología , Inhibidores de la Transcriptasa Inversa/farmacología , Virus de la Inmunodeficiencia de los Simios/fisiología , Proteínas Reguladoras y Accesorias Virales/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Farmacorresistencia Viral , Expresión Génica , Genes Reporteros , Vectores Genéticos/genética , VIH-1/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Activación de Linfocitos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Pruebas de Sensibilidad Microbiana , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/inmunología
8.
Retrovirology ; 11: 63, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25158827

RESUMEN

BACKGROUND: SAMHD1 degrades deoxyribonucleotides (dNTPs), suppressing viral DNA synthesis in macrophages. Recently, viral protein X (Vpx) of HIV-2/SIVsm was shown to target SAMHD1 for proteosomal degradation and led to elevation of dNTP levels, which in turn accelerated proviral DNA synthesis of lentiviruses in macrophages. RESULTS: We investigated both time-dependent and quantitative interplays between SAMHD1 level and dNTP concentrations during multiple exposures of Vpx in macrophages. The following were observed. First, SAMHD1 level was rapidly reduced by Vpx + VLP to undetectable levels by Western blot analysis. Recovery of SAMHD1 was very slow with less than 3% of the normal macrophage level detected at day 6 post Vpx treatment and only ~30% recovered at day 14. Second, dGTP, dCTP and dTTP levels peaked at day 1 post Vpx treatment, whereas dATP peaked at day 2. However, all dNTPs rapidly decreased starting at day 3, while SAMHD1 level was below the level of detection. Third, when Vpx pretreated macrophages were re-exposed to a second Vpx treatment at day 7, we observed dNTP elevation that had faster kinetics than the first Vpx + VLP treatment. Moreover, we performed a short kinetic analysis of the second Vpx treatment to find that dATP and dGTP levels peaked at 8 hours post secondary VLP treatment. dGTP peak was consistently higher than the primary, whereas peak dATP concentration was basically equivalent to the first Vpx + VLP treatment. Lastly, HIV-1 replication kinetics were faster in macrophages treated after the secondary Vpx treatments when compared to the initial single Vpx treatment. CONCLUSION: This study reveals that a very low level of SAMHD1 sufficiently modulates the normally low dNTP levels in macrophages and proposes potential diverse mechanisms of Vpx-mediated dNTP regulation in macrophages.


Asunto(s)
Macrófagos/metabolismo , Monocitos/citología , Proteínas de Unión al GTP Monoméricas/análisis , Nucleótidos/análisis , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , VIH-1/genética , Humanos , Proteína 1 que Contiene Dominios SAM y HD , Espectrometría de Masas en Tándem , Proteínas Reguladoras y Accesorias Virales/farmacología , Virión/fisiología , Gemcitabina
9.
Nat Med ; 20(8): 936-41, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25038827

RESUMEN

The HIV-1 restriction factor SAM domain- and HD domain-containing protein 1 (SAMHD1) is proposed to inhibit HIV-1 replication by depleting the intracellular dNTP pool. However, phosphorylation of SAMHD1 regulates its ability to restrict HIV-1 without decreasing cellular dNTP levels, which is not consistent with a role for SAMHD1 dNTPase activity in HIV-1 restriction. Here, we show that SAMHD1 possesses RNase activity and that the RNase but not the dNTPase function is essential for HIV-1 restriction. By enzymatically characterizing Aicardi-Goutières syndrome (AGS)-associated SAMHD1 mutations and mutations in the allosteric dGTP-binding site of SAMHD1 for defects in RNase or dNTPase activity, we identify SAMHD1 point mutants that cause loss of one or both functions. The RNase-positive and dNTPase-negative SAMHD1D137N mutant is able to restrict HIV-1 infection, whereas the RNase-negative and dNTPase-positive SAMHD1Q548A mutant is defective for HIV-1 restriction. SAMHD1 associates with HIV-1 RNA and degrades it during the early phases of cell infection. SAMHD1 silencing in macrophages and CD4(+) T cells from healthy donors increases HIV-1 RNA stability, rendering the cells permissive for HIV-1 infection. Furthermore, phosphorylation of SAMHD1 at T592 negatively regulates its RNase activity in cells and impedes HIV-1 restriction. Our results reveal that the RNase activity of SAMHD1 is responsible for preventing HIV-1 infection by directly degrading the HIV-1 RNA.


Asunto(s)
Infecciones por VIH/virología , VIH-1/fisiología , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , ARN Viral/metabolismo , Replicación Viral , Enfermedades Autoinmunes del Sistema Nervioso/genética , Secuencia de Bases , Sitios de Unión/genética , Linfocitos T CD4-Positivos , Línea Celular Tumoral , Infecciones por VIH/genética , Células HeLa , Humanos , Macrófagos , Mutación , Malformaciones del Sistema Nervioso/genética , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Viral/genética , Ribonucleasas/metabolismo , Proteína 1 que Contiene Dominios SAM y HD , Análisis de Secuencia de ARN
10.
PLoS Pathog ; 9(6): e1003481, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825958

RESUMEN

SAMHD1 is a newly identified anti-HIV host factor that has a dNTP triphosphohydrolase activity and depletes intracellular dNTP pools in non-dividing myeloid cells. Since DNA viruses utilize cellular dNTPs, we investigated whether SAMHD1 limits the replication of DNA viruses in non-dividing myeloid target cells. Indeed, two double stranded DNA viruses, vaccinia and herpes simplex virus type 1, are subject to SAMHD1 restriction in non-dividing target cells in a dNTP dependent manner. Using a thymidine kinase deficient strain of vaccinia virus, we demonstrate a greater restriction of viral replication in non-dividing cells expressing SAMHD1. Therefore, this study suggests that SAMHD1 is a potential innate anti-viral player that suppresses the replication of a wide range of DNA viruses, as well as retroviruses, which infect non-dividing myeloid cells.


Asunto(s)
Herpesvirus Humano 1/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Células Mieloides/metabolismo , Células Mieloides/virología , Virus Vaccinia/fisiología , Replicación Viral/fisiología , Línea Celular , Femenino , Humanos , Masculino , Proteínas de Unión al GTP Monoméricas/genética , Células Mieloides/patología , Proteína 1 que Contiene Dominios SAM y HD
11.
Retrovirology ; 9: 105, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23231760

RESUMEN

BACKGROUND: SAMHD1 is an HIV-1 restriction factor in non-dividing monocytes, dendritic cells (DCs), macrophages, and resting CD4+ T-cells. Acting as a deoxynucleoside triphosphate (dNTP) triphosphohydrolase, SAMHD1 hydrolyzes dNTPs and restricts HIV-1 infection in macrophages and resting CD4+ T-cells by decreasing the intracellular dNTP pool. However, the intracellular dNTP pool in DCs and its regulation by SAMHD1 remain unclear. SAMHD1 has been reported as a type I interferon (IFN)-inducible protein, but whether type I IFNs upregulate SAMHD1 expression in primary DCs and CD4+ T-lymphocytes is unknown. RESULTS: Here, we report that SAMHD1 significantly blocked single-cycle and replication-competent HIV-1 infection of DCs by decreasing the intracellular dNTP pool and thereby limiting the accumulation of HIV-1 late reverse transcription products. Type I IFN treatment did not upregulate endogenous SAMHD1 expression in primary DCs or CD4+ T-lymphocytes, but did in HEK 293T and HeLa cell lines. When SAMHD1 was over-expressed in these two cell lines to achieve higher levels than that in DCs, no HIV-1 restriction was observed despite partially reducing the intracellular dNTP pool. CONCLUSIONS: Our results suggest that SAMHD1-mediated reduction of the intracellular dNTP pool in DCs is a common mechanism of HIV-1 restriction in myeloid cells. Endogenous expression of SAMHD1 in primary DCs or CD4+ T-lymphocytes is not upregulated by type I IFNs.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Células Dendríticas/metabolismo , Células Dendríticas/virología , VIH-1/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Interferones/farmacología , Proteínas de Unión al GTP Monoméricas/genética , Proteolisis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Reversa , Proteína 1 que Contiene Dominios SAM y HD , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación Viral
12.
PLoS One ; 7(11): e50424, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23189201

RESUMEN

While pharmacological inhibition of Akt kinase has been regarded as a promising anti-cancer strategy, most of the Akt inhibitors that have been developed are enzymatic inhibitors that target the kinase active site of Akt. Another key cellular regulatory event for Akt activation is the translocation of Akt kinase to the cell membrane from the cytoplasm, which is accomplished through the pleckstrin homology (PH) domain of Akt. However, compounds specifically interacting with the PH domain of Akt to inhibit Akt activation are currently limited. Here we identified a compound, lancemaside A (LAN-A), which specifically binds to the PH domain of Akt kinase. First, our mass spectra analysis of cellular Akt kinase isolated from cells treated with LAN-A revealed that LAN-A specifically binds to the PH domain of cellular Akt kinase. Second, we observed that LAN-A inhibits the translocation of Akt kinase to the membrane and thus Akt activation, as examined by the phosphorylation of various downstream targets of Akt such as GSK3ß, mTOR and BAD. Third, in a co-cultured cell model containing human lung epithelial cancer cells (A549) and normal human primary lung fibroblasts, LAN-A specifically restricts the growth of the A549 cells. LAN-A also displayed anti-proliferative effects on various human cancer cell lines. Finally, in the A549-luciferase mouse transplant model, LAN-A effectively inhibited A549 cell growth with little evident cytotoxicity. Indeed, the therapeutic index of LAN-A in this mouse model was >250, supporting that LAN-A is a potential lead compound for PH domain targeting as a safe anti-cancer Akt inhibitor.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/química , Saponinas/farmacología , Animales , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Línea Celular Tumoral , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/toxicidad , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Saponinas/metabolismo , Saponinas/toxicidad , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Biol Chem ; 287(26): 21570-4, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22589553

RESUMEN

Recently, SAMHD1 has come under intense focus as a host anti-HIV factor. SAMHD1 is a dNTP triphosphohydrolase, which leads to the regulation of DNA metabolism in host cells. HIV-2/SIV (simian immunodeficiency virus) viral protein x (Vpx) has been shown to promote the degradation of SAMHD1. In this study, we examine the kinetics of SAMHD1 degradation, the increase in the dNTP pool level, and the efficiency of proviral DNA synthesis in Vpx+ virus-like particle (VLP)-treated monocyte-derived macrophages (MDMs). Our results indicate a very close temporal link with a reduction in SAMHD1 detected within the first few hours of Vpx+ VLP treatment. This loss of SAMHD1 is followed by a significant increase in cellular dNTP levels by 8 h after Vpx+ VLP addition, ultimately leading to the enhancement of the HIV proviral DNA synthesis rate and HIV infection in MDMs. Finally, the pretreatment of MDMs with the Vpx+ VLPs, which is a widely used protocol, displayed identical proviral DNA synthesis as compared with MDMs co-treated with Vpx+ VLP and HIV vector. These findings further indicate that Vpx degradation of SAMHD1 is sufficiently rapid to enable appropriate progression of reverse transcription in MDMs, even when present at the time of infection. Overall, this study demonstrates a tight interplay between SAMHD1 level, dNTP levels, and HIV proviral DNA synthesis kinetics in MDMs.


Asunto(s)
ADN Viral/metabolismo , Fosfatos de Dinucleósidos/metabolismo , VIH-1/metabolismo , Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación del ADN , Fosfatos de Dinucleósidos/química , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Cinética , Receptores de Lipopolisacáridos/biosíntesis , Modelos Biológicos , Nucleótidos/química , Reacción en Cadena de la Polimerasa/métodos , Proteína 1 que Contiene Dominios SAM y HD , Factores de Tiempo
14.
PLoS Pathog ; 8(4): e1002635, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496656

RESUMEN

Leishmaniasis is a parasitic disease that is widely prevalent in many tropical and sub-tropical regions of the world. Infection with Leishmania has been recognized to induce a striking acceleration of Human Immunodeficiency Virus Type 1 (HIV-1) infection in coinfected individuals through as yet incompletely understood mechanisms. Cells of the monocyte/macrophage lineage are the predominant cell types coinfected by both pathogens. Monocytes and macrophages contain extremely low levels of deoxynucleoside triphosphates (dNTPs) due to their lack of cell cycling and S phase, where dNTP biosynthesis is specifically activated. Lentiviruses, such as HIV-1, are unique among retroviruses in their ability to replicate in these non-dividing cells due, at least in part, to their highly efficient reverse transcriptase (RT). Nonetheless, viral replication progresses more efficiently in the setting of higher intracellular dNTP concentrations related to enhanced enzyme kinetics of the viral RT. In the present study, in vitro infection of CD14+ peripheral blood-derived human monocytes with Leishmania major was found to induce differentiation, marked elevation of cellular p53R2 ribonucleotide reductase subunit and R2 subunit expression. The R2 subunit is restricted to the S phase of the cell cycle. Our dNTP assay demonstrated significant elevation of intracellular monocyte-derived macrophages (MDMs) dNTP concentrations in Leishmania-infected cell populations as compared to control cells. Infection of Leishmania-maturated MDMs with a pseudotyped GFP expressing HIV-1 resulted in increased numbers of GFP+ cells in the Leishmania-maturated MDMs as compared to control cells. Interestingly, a sub-population of Leishmania-maturated MDMs was found to have re-entered the cell cycle, as demonstrated by BrdU labeling. In conclusion, Leishmania infection of primary human monocytes promotes the induction of an S phase environment and elevated dNTP levels with notable elevation of HIV-1 expression in the setting of coinfection.


Asunto(s)
Desoxirribonucleótidos/metabolismo , Infecciones por VIH , VIH-1/metabolismo , Leishmania major/metabolismo , Leishmaniasis Cutánea , Macrófagos , Proteínas de Ciclo Celular/biosíntesis , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Infecciones por VIH/complicaciones , Infecciones por VIH/metabolismo , Transcriptasa Inversa del VIH/metabolismo , Humanos , Leishmaniasis Cutánea/complicaciones , Leishmaniasis Cutánea/metabolismo , Macrófagos/metabolismo , Macrófagos/parasitología , Macrófagos/virología , Masculino , Monocitos , Ribonucleótido Reductasas/biosíntesis , Fase S
15.
PLoS One ; 6(7): e21781, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21765914

RESUMEN

The PI3K/Akt pathway regulates various stress-related cellular responses such as cell survival, cell proliferation, metabolism and protein synthesis. Many cancer cell types display the activation of this pathway, and compounds inhibiting this cell survival pathway have been extensively evaluated as anti-cancer agents. In addition to cancers, several human viruses, such as HTLV, HPV, HCV and HIV-1, also modulate this pathway, presumably in order to extend the life span of the infected target cells for productive viral replication. The expression of HIV-1 Tat protein exhibited the cytoprotective effect in macrophages and a human microglial cell line by inhibiting the negative regulator of this pathway, PTEN. This cytoprotective effect of HIV-1 appears to contribute to the long-term survival and persistent HIV-1 production in human macrophage reservoirs. In this study we exploited the PI3K/Akt dependent cytoprotective effect of Tat-expressing CHME5 cells. We screened a collection of compounds known to modulate inflammation, and identified three novel compounds: Lancemaside A, Compound K and Arctigenin that abolished the cytoprotective phenotype of Tat-expressing CHME5 cells. All three compounds antagonized the kinase activity of Akt. Further detailed signaling studies revealed that each of these three compounds targeted different steps of the PI3K/Akt pathway. Arctigenin regulates the upstream PI3K enzyme from converting PIP2 to PIP3. Lancemaside A1 inhibited the movement of Akt to the plasma membrane, a critical step for Akt activation. Compound K inhibited Akt phosphorylation. This study supports that Tat-expressing CHME5 cells are an effective model system for screening novel PI3K/Akt inhibitors.


Asunto(s)
Citoprotección/efectos de los fármacos , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/farmacología , VIH-1/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , VIH-1/efectos de los fármacos , Humanos , Fosfohidrolasa PTEN/metabolismo , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteína Letal Asociada a bcl/metabolismo
16.
Virology ; 415(2): 153-9, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21565377

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) infects both activated CD4+ T cells and macrophages. We tested if liquid chromatography-tandem mass spectrometry (LC-MS/MS) technology can monitor metabolic alterations induced by HIV-1 in the infected cells. Here we monitored glucose uptake and conducted LC-MS/MS-based metabolomic analysis in HIV-1 infected primary human CD4+ T cells and a macrophage model system: differentiated U1 (HIV-1 producing) and differentiated U937 (control) cells. HIV-1 infected CD4+ T cells have higher glucose uptake and increases in several metabolite pool sizes, whereas HIV-1 producing macrophages had substantial reductions in glucose uptake and steady state glycolytic intermediates. This data suggests that the two HIV-1 target cell types exhibit very different metabolic outcomes during viral production. This study also validates the LC-MS/MS technology as an effective metabolomic approach to monitor various metabolic alterations made by HIV-1 infection.


Asunto(s)
Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/metabolismo , Cromatografía Líquida de Alta Presión/métodos , Infecciones por VIH/metabolismo , VIH/fisiología , Macrófagos/química , Macrófagos/metabolismo , Espectrometría de Masas en Tándem/métodos , Linfocitos T CD4-Positivos/virología , Línea Celular , Células Cultivadas , Infecciones por VIH/virología , Humanos , Macrófagos/virología , Replicación Viral
17.
J Virol ; 84(13): 6687-98, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20410284

RESUMEN

Seasonal and pandemic influenza A virus (IAV) continues to be a public health threat. However, we lack a detailed and quantitative understanding of the immune response kinetics to IAV infection and which biological parameters most strongly influence infection outcomes. To address these issues, we use modeling approaches combined with experimental data to quantitatively investigate the innate and adaptive immune responses to primary IAV infection. Mathematical models were developed to describe the dynamic interactions between target (epithelial) cells, influenza virus, cytotoxic T lymphocytes (CTLs), and virus-specific IgG and IgM. IAV and immune kinetic parameters were estimated by fitting models to a large data set obtained from primary H3N2 IAV infection of 340 mice. Prior to a detectable virus-specific immune response (before day 5), the estimated half-life of infected epithelial cells is approximately 1.2 days, and the half-life of free infectious IAV is approximately 4 h. During the adaptive immune response (after day 5), the average half-life of infected epithelial cells is approximately 0.5 days, and the average half-life of free infectious virus is approximately 1.8 min. During the adaptive phase, model fitting confirms that CD8(+) CTLs are crucial for limiting infected cells, while virus-specific IgM regulates free IAV levels. This may imply that CD4 T cells and class-switched IgG antibodies are more relevant for generating IAV-specific memory and preventing future infection via a more rapid secondary immune response. Also, simulation studies were performed to understand the relative contributions of biological parameters to IAV clearance. This study provides a basis to better understand and predict influenza virus immunity.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Anticuerpos Antivirales/inmunología , Células Epiteliales/fisiología , Células Epiteliales/virología , Semivida , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Teóricos , Linfocitos T Citotóxicos/inmunología , Factores de Tiempo
18.
Virology ; 400(2): 224-32, 2010 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-20185156

RESUMEN

Influenza virus leads to acute respiratory disease resulting in seasonal epidemics and periodic pandemics. Little is known about the signaling events that regulate host defense to influenza. One particular pathway, the c-Jun amino-terminal kinase (JNK) cascade is activated following influenza infection and blocking JNK leads to enhanced viral replication. We hypothesize that Mixed Lineage Kinase 3 (MLK3), an upstream regulator of JNK, is involved in the host response to influenza. To test this, wild-type and MLK3-/- mice were infected with pathogenic strain of influenza A virus, A/PR/8/34 (PR8). Although, cellular and humoral immune responses were similar between wild-type and MLK3-/- hosts, the viral load in the lungs was comparatively higher in MLK3-/- mice at day 8 post-infection. Consistent with this, MLK3-/- murine lung fibroblast and epithelial cells had prolonged survival and increased virion production following infection compared to wild-type. These findings support a role for MLK3 in viral production during influenza infection.


Asunto(s)
Efecto Citopatogénico Viral , Virus de la Influenza A/inmunología , Pulmón/virología , Quinasas Quinasa Quinasa PAM/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Animales , Anticuerpos Antivirales/inmunología , Supervivencia Celular , Células Epiteliales/virología , Fibroblastos/virología , Quinasas Quinasa Quinasa PAM/deficiencia , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/virología , Linfocitos T/inmunología , Carga Viral , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
19.
J Virol ; 83(14): 7151-65, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19439465

RESUMEN

The cellular immune response to primary influenza virus infection is complex, involving multiple cell types and anatomical compartments, and is difficult to measure directly. Here we develop a two-compartment model that quantifies the interplay between viral replication and adaptive immunity. The fidelity of the model is demonstrated by accurately confirming the role of CD4 help for antibody persistence and the consequences of immune depletion experiments. The model predicts that drugs to limit viral infection and/or production must be administered within 2 days of infection, with a benefit of combination therapy when administered early, and cytotoxic CD8 T cells in the lung are as effective for viral clearance as neutralizing antibodies when present at the time of challenge. The model can be used to investigate explicit biological scenarios and generate experimentally testable hypotheses. For example, when the adaptive response depends on cellular immune cell priming, regulation of antigen presentation has greater influence on the kinetics of viral clearance than the efficiency of virus neutralization or cellular cytotoxicity. These findings suggest that the modulation of antigen presentation or the number of lung resident cytotoxic cells and the combination drug intervention are strategies to combat highly virulent influenza viruses. We further compared alternative model structures, for example, B-cell activation directly by the virus versus that through professional antigen-presenting cells or dendritic cell licensing of CD8 T cells.


Asunto(s)
Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Animales , Antivirales/uso terapéutico , Simulación por Computador , Humanos , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/fisiología , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Modelos Teóricos , Valor Predictivo de las Pruebas
20.
Blood ; 113(17): 4078-85, 2009 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-19244161

RESUMEN

Integrin-mediated cell migration is central to many biologic and pathologic processes. During inflammation, tissue injury results from excessive infiltration and sequestration of activated leukocytes. Recombinant human activated protein C (rhAPC) has been shown to protect patients with severe sepsis, although the mechanism underlying this protective effect remains unclear. Here, we show that rhAPC directly binds to beta(1) and beta(3) integrins and inhibits neutrophil migration, both in vitro and in vivo. We found that human APC possesses an Arg-Gly-Asp (RGD) sequence, which is critical for the inhibition. Mutation of this sequence abolished both integrin binding and inhibition of neutrophil migration. In addition, treatment of septic mice with a RGD peptide recapitulated the beneficial effects of rhAPC on survival. Thus, we conclude that leukocyte integrins are novel cellular receptors for rhAPC and the interaction decreases neutrophil recruitment into tissues, providing a potential mechanism by which rhAPC may protect against sepsis.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Integrinas/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Proteína C/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...