Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 18(3): e1010354, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35255110

RESUMEN

Latency is the main obstacle towards an HIV cure, with cure strategies aiming to either elicit or prevent viral reactivation. While these strategies have shown promise, they have only succeeded in modulating latency in a fraction of the latent HIV reservoir, suggesting that the mechanisms controlling HIV latency are not completely understood, and that comprehensive latency modulation will require targeting of multiple latency maintenance pathways. We show here that the transcriptional co-activator and the central mediator of canonical Wnt signaling, ß-catenin, inhibits HIV transcription in CD4+ T cells via TCF-4 LTR binding sites. Further, we show that inhibiting the ß-catenin pathway reactivates HIV in a primary TCM cell model of HIV latency, primary cells from cART-controlled HIV donors, and in CD4+ latent cell lines. ß-catenin inhibition or activation also enhanced or inhibited the activity of several classes of HIV latency reversing agents, respectively, in these models, with significant synergy of ß-catenin and each LRA class tested. In sum, we identify ß-catenin as a novel regulator of HIV latency in vitro and ex vivo, adding new therapeutic targets that may be combined for comprehensive HIV latency modulation in HIV cure efforts.


Asunto(s)
Infecciones por VIH , VIH-1 , beta Catenina , Linfocitos T CD4-Positivos/metabolismo , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Humanos , Activación Viral , Latencia del Virus , beta Catenina/genética , beta Catenina/metabolismo
2.
J Virol ; 96(15): e0080422, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35852355

RESUMEN

CD4dim CD8bright T cells are a mature population of CD8+ T cells that upon activation upregulate CD4 dimly on their surface. Expression of CD4 on these cells suggests that they can be an additional source of HIV neuroinvasion and persistence in the brain. We used HIV-infected NOD/SCID/IL-2rcγ-/- (NSG) humanized mice to track CD4dim CD8bright T cell homing to the brain and define their role in HIV dissemination into the brain. We report here that CD4dim CD8bright T cells are found in the brain at a median frequency of 2.6% and in the spleen at median frequency of 7.6% of CD3+ T cells. In the brain, 10 to 20% of CD4dim CD8bright T cells contain integrated provirus, which is infectious as demonstrated by viral outgrowth assay. CD4dim CD8bright T cells in the brain exhibited significantly higher expression of the brain homing receptors CX3CR1 and CXCR3 in comparison to their single-positive CD8+ T cell counterpart. Blocking lymphocyte trafficking into the brain of humanized mice via anti-VLA4 and anti-LFA1 antibodies reduced CD4dim CD8bright T cell trafficking into the brain by 60% and diminished brain HIV proviral DNA by 72%. Collectively, our findings demonstrate that CD4dim CD8bright T cells can home to the brain and support productive HIV replication. These studies also reveal for the first time that CD4dim CD8bright T cells are capable of HIV neuroinvasion and are a reservoir for HIV. IMPORTANCE We report here a seminal finding of a novel population of T cells, termed CD4dim CD8bright T cells, that plays a role in HIV neuroinvasion and is a reservoir for HIV in the brain.


Asunto(s)
Encéfalo , Antígenos CD4 , Antígenos CD8 , Linfocitos T CD8-positivos , Movimiento Celular , Infecciones por VIH , VIH-1 , Tropismo Viral , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/virología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptor 1 de Quimiocinas CX3C/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/patogenicidad , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Ratones , Ratones Endogámicos NOD , Ratones SCID , Provirus/genética , Provirus/aislamiento & purificación , Receptores CXCR3/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo
3.
Antimicrob Agents Chemother ; 66(4): e0194121, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35293780

RESUMEN

Combination antiretroviral therapy (cART) dramatically changed the face of the HIV/AIDS pandemic, making it one of the most prominent medical breakthroughs of the past 3 decades. However, as the life span of persons living with HIV (PLWH) continues to approach that of the general population, the same cannot be said regarding their quality of life. PLWH are affected by comorbid conditions such as high blood pressure, diabetes, and neurocognitive impairment at a higher rate and increased severity than their age-matched counterparts. PLWH also have higher levels of inflammation, the drivers of which are not entirely clear. As cART treatment is lifelong, we assessed here the effects of cART, independent of HIV, on primary human monocyte-derived macrophages (MDMs). MDMs were unskewed or skewed to an alternative phenotype and treated with Atripla or Triumeq, two first-line cART treatments. We report that Triumeq skewed alternative MDMs toward an inflammatory nonsenescent phenotype. Both Atripla and Triumeq caused mitochondrial dysfunction, specifically efavirenz and abacavir. Additionally, transcriptome sequencing (RNA-seq) demonstrated that both Atripla and Triumeq caused differential regulation of genes involved in immune regulation and cell cycle and DNA repair. Collectively, our data demonstrate that cART, independent of HIV, alters the MDM phenotype. This suggests that cART may contribute to cell dysregulation in PLWH that subsequently results in increased susceptibility to comorbidities.


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , VIH-1 , Fármacos Anti-VIH/uso terapéutico , Combinación Efavirenz, Emtricitabina y Fumarato de Tenofovir Disoproxil/metabolismo , Combinación Efavirenz, Emtricitabina y Fumarato de Tenofovir Disoproxil/farmacología , Combinación Efavirenz, Emtricitabina y Fumarato de Tenofovir Disoproxil/uso terapéutico , Humanos , Macrófagos , Mitocondrias , Calidad de Vida
4.
J Virol ; 95(17): e0070521, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34260264

RESUMEN

The latest outbreak of Zika virus (ZIKV) in the Americas was associated with significant neurologic complications, including microcephaly of newborns. We evaluated mechanisms that regulate ZIKV entry into human fetal astrocytes (HFAs). Astrocytes are key players in maintaining brain homeostasis. We show that the central mediator of canonical Wnt signaling, ß-catenin, regulates Axl, a receptor for ZIKV infection of HFAs, at the transcriptional level. In turn, ZIKV inhibited ß-catenin, potentially as a mechanism to overcome its restriction of ZIKV internalization through regulation of Axl. This was evident with three ZIKV strains tested but not with a laboratory-adapted strain which has a large deletion in its envelope gene. Finally, we show that ß-catenin-mediated Axl-dependent internalization of ZIKV may be of increased importance for brain cells, as it regulated ZIKV infection of astrocytes and human brain microvascular cells but not kidney epithelial (Vero) cells. Collectively, our studies reveal a role for ß-catenin in ZIKV infection and highlight a dynamic interplay between ZIKV and ß-catenin to modulate ZIKV entry into susceptible target cells. IMPORTANCE ZIKV is an emerging pathogen with sporadic outbreaks throughout the world. The most recent outbreak in North America was associated with small brains (microcephaly) in newborns. We studied the mechanism(s) that may regulate ZIKV entry into astrocytes. Astrocytes are a critical resident brain cell population with diverse functions that maintain brain homeostasis, including neurogenesis and neuronal survival. We show that three ZIKV strains (and not a heavily laboratory-adapted strain with a large deletion in its envelope gene) require Axl for internalization. Most importantly, we show that ß-catenin, the central mediator of canonical Wnt signaling, negatively regulates Axl at the transcriptional level to prevent ZIKV internalization into human fetal astrocytes. To overcome this restriction, ZIKV downregulates ß-catenin to facilitate Axl expression. This highlights a dynamic host-virus interaction whereby ZIKV inhibits ß-catenin to promote its internalization into human fetal astrocytes through the induction of Axl.


Asunto(s)
Astrocitos/virología , Encéfalo/virología , Feto/virología , Replicación Viral , Infección por el Virus Zika/prevención & control , Virus Zika/fisiología , beta Catenina/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Chlorocebus aethiops , Feto/metabolismo , Humanos , Riñón/metabolismo , Riñón/virología , Células Vero , Internalización del Virus , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología , beta Catenina/genética
5.
PLoS Pathog ; 16(6): e1008381, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32525948

RESUMEN

HIV invades the brain during acute infection. Yet, it is unknown whether long-lived infected brain cells release productive virus that can egress from the brain to re-seed peripheral organs. This understanding has significant implication for the brain as a reservoir for HIV and most importantly HIV interplay between the brain and peripheral organs. Given the sheer number of astrocytes in the human brain and their controversial role in HIV infection, we evaluated their infection in vivo and whether HIV infected astrocytes can support HIV egress to peripheral organs. We developed two novel models of chimeric human astrocyte/human peripheral blood mononuclear cells: NOD/scid-IL-2Rgc null (NSG) mice (huAstro/HuPBMCs) whereby we transplanted HIV (non-pseudotyped or VSVg-pseudotyped) infected or uninfected primary human fetal astrocytes (NHAs) or an astrocytoma cell line (U138MG) into the brain of neonate or adult NSG mice and reconstituted the animals with human peripheral blood mononuclear cells (PBMCs). We also transplanted uninfected astrocytes into the brain of NSG mice and reconstituted with infected PBMCs to mimic a biological infection course. As expected, the xenotransplanted astrocytes did not escape/migrate out of the brain and the blood brain barrier (BBB) was intact in this model. We demonstrate that astrocytes support HIV infection in vivo and egress to peripheral organs, at least in part, through trafficking of infected CD4+ T cells out of the brain. Astrocyte-derived HIV egress persists, albeit at low levels, under combination antiretroviral therapy (cART). Egressed HIV evolved with a pattern and rate typical of acute peripheral infection. Lastly, analysis of human cortical or hippocampal brain regions of donors under cART revealed that astrocytes harbor between 0.4-5.2% integrated HIV gag DNA and 2-7% are HIV gag mRNA positive. These studies establish a paradigm shift in the dynamic interaction between the brain and peripheral organs which can inform eradication of HIV reservoirs.


Asunto(s)
Astrocitos , Barrera Hematoencefálica , Infecciones por VIH , VIH-1/metabolismo , Hipocampo , Liberación del Virus , Animales , Antirretrovirales/farmacología , Astrocitos/metabolismo , Astrocitos/patología , Astrocitos/virología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/virología , Línea Celular Tumoral , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , VIH-1/genética , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/virología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID
6.
J Immunol ; 205(8): 2046-2055, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32887752

RESUMEN

CD8+ T cells do not rely solely on cytotoxic functions for significant HIV control. Moreover, the noncytotoxic CD8+ T cell antiviral response is a primary mediator of natural HIV control such as that seen in HIV elite controllers and long-term nonprogressors that does not require combined antiretroviral therapy. In this study, we investigated the biological factors contributing to the noncytotoxic control of HIV replication mediated by primary human CD8+ T cells. We report that canonical Wnt signaling inhibits HIV transcription in an MHC-independent, noncytotoxic manner and that mediators of this pathway correlate with HIV controller clinical status. We show that CD8+ T cells express all 19 Wnts and CD8+ T cell-conditioned medium (CM) induced canonical Wnt signaling in infected recipient cells while simultaneously inhibiting HIV transcription. Antagonizing canonical Wnt activity in CD8+ T cell CM resulted in increased HIV transcription in infected cells. Further, Wnt2b expression was upregulated in HIV controllers versus viremic patients, and in vitro depletion of Wnt2b and/or Wnt9b from CD8+ CM reversed HIV inhibitory activity. Finally, plasma concentration of Dkk-1, an antagonist of canonical Wnt signaling, was higher in viremic patients with lower CD4 counts. This study demonstrates that canonical Wnt signaling inhibits HIV and significantly correlates with HIV controller status.


Asunto(s)
Linfocitos T CD8-positivos , Regulación de la Expresión Génica/inmunología , Glicoproteínas , Infecciones por VIH , VIH-1 , Inmunidad Celular , Proteínas Wnt , Vía de Señalización Wnt/inmunología , Adulto , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Femenino , Glicoproteínas/sangre , Glicoproteínas/inmunología , Infecciones por VIH/sangre , Infecciones por VIH/inmunología , Infecciones por VIH/patología , VIH-1/inmunología , VIH-1/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/inmunología , Masculino , Proteínas Wnt/sangre , Proteínas Wnt/inmunología
7.
Circ Res ; 124(4): e6-e19, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30595089

RESUMEN

RATIONALE: Atherosclerosis is, in part, caused by immune and inflammatory cell infiltration into the vascular wall, leading to enhanced inflammation and lipid accumulation in the aortic endothelium. Understanding the molecular mechanisms underlying this disease is critical for the development of new therapies. Our recent studies demonstrate that epsins, a family of ubiquitin-binding endocytic adaptors, are critical regulators of atherogenicity. Given the fundamental contribution lesion macrophages make to fuel atherosclerosis, whether and how myeloid-specific epsins promote atherogenesis is an open and significant question. OBJECTIVE: We will determine the role of myeloid-specific epsins in regulating lesion macrophage function during atherosclerosis. METHODS AND RESULTS: We engineered myeloid cell-specific epsins double knockout mice (LysM-DKO) on an ApoE-/- background. On Western diet, these mice exhibited marked decrease in atherosclerotic lesion formation, diminished immune and inflammatory cell content in aortas, and reduced necrotic core content but increased smooth muscle cell content in aortic root sections. Epsins deficiency hindered foam cell formation and suppressed proinflammatory macrophage phenotype but increased efferocytosis and anti-inflammatory macrophage phenotype in primary macrophages. Mechanistically, we show that epsin loss specifically increased total and surface levels of LRP-1 (LDLR [low-density lipoprotein receptor]-related protein 1), an efferocytosis receptor with antiatherosclerotic properties. We further show that epsin and LRP-1 interact via epsin's ubiquitin-interacting motif domain. ox-LDL (oxidized LDL) treatment increased LRP-1 ubiquitination, subsequent binding to epsin, and its internalization from the cell surface, suggesting that epsins promote the ubiquitin-dependent internalization and downregulation of LRP-1. Crossing ApoE-/-/LysM-DKO mice onto an LRP-1 heterozygous background restored, in part, atherosclerosis, suggesting that epsin-mediated LRP-1 downregulation in macrophages plays a pivotal role in propelling atherogenesis. CONCLUSIONS: Myeloid epsins promote atherogenesis by facilitating proinflammatory macrophage recruitment and inhibiting efferocytosis in part by downregulating LRP-1, implicating that targeting epsins in macrophages may serve as a novel therapeutic strategy to treat atherosclerosis.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Aterosclerosis/metabolismo , Regulación hacia Abajo , Receptores de LDL/genética , Proteínas Supresoras de Tumor/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Apolipoproteínas E/genética , Aterosclerosis/genética , Células Cultivadas , Eliminación de Gen , Células HEK293 , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Macrófagos/metabolismo , Ratones , Células Mieloides/metabolismo , Células RAW 264.7 , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación
8.
Cytokine ; 136: 155252, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32818703

RESUMEN

Expression of cytokines/chemokines is tightly regulated at the transcription level. This is crucial in the central nervous system to maintain neuroimmune homeostasis. IL-8 a chemoattractant, which recruits neutrophils, T cells, and basophils into the brain in response to inflammation and/or injury is secreted predominantly by neurons, microglia, and astrocytes. Here, we investigated the mechanism by which astrocytes regulate IL-8 expression. We demonstrate that while ß-catenin negatively regulated IL-8 transcription, its canonical transcriptional partners, members of the TCF/LEF transcription factors (TCF1, TCF3, TCF4 and LEF1) and Activating transcription factor 2 (ATF2) positively regulated IL-8 transcription. We further identified a putative TCF/LEF binding site at -175nt close to the minimal transcription region on the IL-8 promoter, mutation of which caused a significant reduction in IL-8 promoter activity. Chromatin immunoprecipitation demonstrated binding of TCF1, TCF4, LEF1 and ATF2 on the IL-8 promoter suggesting that TCFs/LEF partner with ATF2 to induce IL-8 transcription. These findings demonstrate a novel role for ß-catenin in suppression of IL-8 expression and for TCFs/LEF/ATF2 in inducing IL-8. These findings reveal a unique mechanism by which astrocytes tightly regulate IL-8 expression.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Astrocitos/metabolismo , Regulación de la Expresión Génica , Interleucina-8/biosíntesis , Factores de Transcripción TCF/metabolismo , beta Catenina/metabolismo , Línea Celular Tumoral , Humanos
9.
Cell Commun Signal ; 18(1): 93, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32546183

RESUMEN

BACKGROUND: The Wnt/ß-catenin signaling pathway is a prolific regulator of cell-to-cell communication and gene expression. Canonical Wnt/ß-catenin signaling involves partnering of ß-catenin with members of the TCF/LEF family of transcription factors (TCF1, TCF3, TCF4, LEF1) to regulate gene expression. IL-6 is a key cytokine involved in inflammation and is particularly a hallmark of inflammation in the brain. Astrocytes, specialized glial cells in the brain, secrete IL-6. How astrocytes regulate IL-6 expression is not entirely clear, although in other cells NFκB and C/EBP pathways play a role. We evaluated here the interface between ß-catenin, TCFs/LEF and C/EBP and NF-κB in relation to IL-6 gene regulation in astrocytes. METHODS: We performed molecular loss and/or gain of function studies of ß-catenin, TCF/LEF, NFκB, and C/EBP to assess IL-6 regulation in human astrocytes. Specifically, siRNA mediated target gene knockdown, cDNA over expression of target gene, and pharmacological agents for regulation of target proteins were used. IL-6 levels was evaluated by real time quantitative PCR and ELISA. We also cloned the IL-6 promoter under a firefly luciferase reporter and used bioinformatics, site directed mutagenesis, and chromatin immunoprecipitation to probe the interaction between ß-catenin/TCFs/LEFs and IL-6 promoter activity. RESULTS: ß-catenin binds to TCF/LEF to inhibits IL-6 while TCFs/LEF induce IL-6 transcription through interaction with ATF-2/SMADs. ß-catenin independent of TCFs/LEF positively regulates C/EBP and NF-κB, which in turn activate IL-6 expression. The IL-6 promoter has two putative regions for TCFs/LEF binding, a proximal site located at -91 nt and a distal site at -948 nt from the transcription start site, both required for TCF/LEF induction of IL-6 independent of ß-catenin. CONCLUSION: IL-6 regulation in human astrocytes engages a discordant interaction between ß-catenin and TCF/LEF. These findings are intriguing given that no role for ß-catenin nor TCFs/LEF to date is associated with IL-6 regulation and suggest that ß-catenin expression in astrocytes is a critical regulator of anti-inflammatory responses and its disruption can potentially mediate persistent neuroinflammation. Video Abstract.


Asunto(s)
Astrocitos/metabolismo , Factores Nucleares del Hepatocito/metabolismo , Interleucina-6/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Astrocitos/citología , Línea Celular , Humanos
10.
Nicotine Tob Res ; 21(1): 101-110, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30085294

RESUMEN

Introduction: Smokeless tobacco products such as snuff and snus are used worldwide. However, little is known about the systemic and cardiovascular toxicity of smokeless tobacco exposure. Methods: Biomarkers of endothelial activation and injury, immune functions, platelet activation and insulin resistance were measured in 8-week old male C57BL/6 mice exposed to commercial snuff, CRP-2 reference snuff, commercial snus, CRP-1 reference snus, and nicotine in drinking water (100 µg/mL) for 4, 12, and 24 weeks. Results: Twenty-four weeks of exposure to smokeless tobacco products or nicotine significantly decreased the levels of circulating Flk+/Sca+ endothelial progenitor cells. Twelve and 24 weeks of exposure to all the smokeless tobacco products and nicotine significantly decreased the levels of circulating CD19+ B cells, CD4+ T cells, CD8+ T cells, and CD11b+ monocytes, whereas 4 weeks of exposure to Camel snus and Copenhagen snuff significantly depleted the levels of peripheral blood CD19+ B cells and CD11b+ monocytes. Twenty-four weeks of exposure to smokeless tobacco products or nicotine significantly decreased plasma IFNγ levels. However, plasma TNFα levels were significantly increased in mice exposed to Copenhagen snuff or nicotine for 24 weeks. This was accompanied by a five to sevenfold increase in the hepatic expression of TNFα. Neither smokeless products nor nicotine affected plasma lipoproteins, platelet activation, or systemic insulin sensitivity. Conclusions: Chronic exposure to snuff and snus suppresses circulating levels of EPCs, endothelial microparticles and immune cells, but increases plasma TNF-α levels. These effects of smokeless tobacco products are attributable, at least in part, to nicotine. Implications: Exposure to smokeless tobacco products results in the depletion of endothelial progenitor cells, which may impair the endothelium repair. Suppression of the circulating levels of immune cells upon exposure to smokeless tobacco products may increase the susceptibility to secondary infection. Increased formation of proinflammatory cytokines such as TNFα by nicotine or Copenhagen snuff may lead to vascular inflammation and thereby exacerbate atherogenesis.


Asunto(s)
Biomarcadores/análisis , Endotelio Vascular/patología , Inmunidad Celular/efectos de los fármacos , Resistencia a la Insulina , Activación Plaquetaria/efectos de los fármacos , Trombosis/patología , Tabaco sin Humo/toxicidad , Animales , Endotelio Vascular/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Trombosis/inducido químicamente
11.
J Immunol ; 196(1): 317-27, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26582945

RESUMEN

The role of CD8(+) T cells in HIV control in the brain and the consequences of such control are unclear. Approximately 3% of peripheral CD8(+) T cells dimly express CD4 on their surface. This population is known as CD4(dim)CD8(bright) T cells. We evaluated the role of CD4(dim)CD8(bright) and CD8 single positive T cells in HIV-infected brain using NOD/SCID/IL-2rcγ(-/-) mice reconstituted with human PBMCs (NSG-huPBMC). All three T cell populations (CD4 single positive, CD8 single positive, and CD4(dim)CD8(bright)) were found in NSG-huPBMC mouse brain within 2 wk of infection. Wnts secreted from astrocytes induced CD4(dim)CD8(bright) T cells by 2-fold in vitro. Injection of highly purified CD8 single positive T cells into mouse brain induced CD4(dim)CD8(bright) T cells by 10-fold, which were proliferative and exhibited a terminally differentiated effector memory phenotype. Brain CD4(dim)CD8(bright) T cells from HIV-infected mice exhibited anti-HIV-specific responses, as demonstrated by induction of CD107ab post exposure to HIV peptide-loaded targets. Further, higher frequency of CD4(dim)CD8(bright) T cells (R = -0.62; p ≤ 0.001), but not CD8 single positive T cells (R = -0.24; p ≤ 0.27), negatively correlated with HIV gag mRNA transcripts in HIV-infected NSG-huPBMC brain. Together, these studies indicate that single positive CD8(+) T cells entering the CNS during HIV infection can give rise to CD4(dim)CD8(bright) T cells, likely through a Wnt signaling-dependent manner, and that these cells are associated with potent anti-HIV control in the CNS. Thus, CD4(dim)CD8(bright) T cells are capable of HIV control in the CNS and may offer protection against HIV-associated neurocognitive disorders.


Asunto(s)
Encéfalo/inmunología , Antígenos CD4/metabolismo , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Vía de Señalización Wnt/inmunología , Animales , Astrocitos/metabolismo , Encéfalo/citología , Encéfalo/virología , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/trasplante , Movimiento Celular/inmunología , Células Cultivadas , Infecciones por VIH/virología , Xenoinjertos , Subunidad gamma Común de Receptores de Interleucina/genética , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/trasplante , Activación de Linfocitos/inmunología , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Bazo/citología
12.
Rapid Commun Mass Spectrom ; 31(1): 121-128, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27788280

RESUMEN

RATIONALE: 1,2,5-Thiadiazoles are an important class of compounds mostly used in synthetic chemistry, and as herbicides, insecticides, drugs, organic conductors, etc. Recently, they have been used as a source for the generation and study of nitrile N-sulfides, RCNS, and its isomers. In this study, we monitor the fragmentation pattern of ionic halogenated 1,2,5-thiadiazoles, namely, 3,4-dichloro-1,2,5-thiadiazole, which generates the nitrile sulfides, to establish its various dissociation mechanisms. METHODS: The molecular cation of 3,4-dichloro-1,2,5-thiadiazole was prepared using multiphoton excitation using a laser at 235 nm. Various product ions upon fragmentation of the molecular ion were mass analyzed using time-of-flight mass spectrometry. Laser power dependence studies were conducted for various product ions to arrive at the dissociation mechanism. Theoretical calculations were performed for the estimation of the ΔH values for various reactions to support the experimental data. RESULTS: The most abundant product ion was observed to be the NS+ radical cation followed by the S+ ion and the SCl+ radical cation. The other product ions such as the CNS+ radical cation and the ClCNS+ and ClCN+ cations were also observed to a lesser extent in the fragmentation pattern of the parent molecular ion. Various dissociation channels were identified and supported with ab initio calculations. CONCLUSIONS: In conclusion, we have studied the fragmentation pattern of the molecular cation of 3,4-dichloro-1,2,5-thiadiazole and the formation mechanisms of various product ions have been assigned. It has been also observed that most of the product ions are nitrile N-sulfides. Finally, it is inferred that there are two primary paths for the fragmentation of the parent molecular cation, namely, (1) Cl atom migration and subsequent ring opening by N-S bond cleavage and (2) direct ring opening by N-S bond cleavage. The ionization energies were accurately predicted for various species using ab initio calculations. Copyright © 2016 John Wiley & Sons, Ltd.

13.
Europace ; 19(10): 1664-1669, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28204456

RESUMEN

AIM: During ablation of the posterior wall (PW), luminal oesophageal temperature elevation (OTE) prompts attenuation of radiofrequency (RF) energy delivery to minimize oesophageal injury. This strategy on lesion efficacy is unknown. The goal of this study was to analyse the relationship between OTE and pulmonary vein reconnection (PVR). METHODS AND RESULTS: During the index antral pulmonary vein (PV) isolation procedure with an irrigated RF ablation catheter, OTE was detected with a multisensor oesophageal temperature probe. Posterior wall ablation did not exceed 25 W and was terminated when the temperature was ≥38.5°C. Patients undergoing redo procedures (n = 142) were studied for PW sites of PVR along 4 segments: left and right superior, and left and right inferior. Pulmonary vein reconnections had occurred in 51 of the 142 patients (36%), in 58 of 284 PV pairs (20%). Among these 58 reconnected pairs, 83% (n = 48) were along the PW. Oesophageal temperature elevation had occurred in 30 patients (59%). No difference in characteristics was seen between the patients with OTE (n = 30) and those without (n = 21). For superior segments, there was no interaction between the presence or absence of OTE and PVR. For inferior segments, there were more PVRs in the group with OTE: for the right-inferior segment, the PVR rate was 72% for OTE cases vs. 42% without (P = 0.04), and for the left-inferior segment, the PVR rate was 44% for OTE cases vs. 22.9% without (P = 0.12). CONCLUSION: Pulmonary vein reconnections are predominantly posteriorly located. Along the right- and left-inferior PW segments, there was an association with elevated oesophageal temperature during the index procedure.


Asunto(s)
Fibrilación Atrial/cirugía , Regulación de la Temperatura Corporal , Ablación por Catéter , Esófago/fisiopatología , Atrios Cardíacos/cirugía , Venas Pulmonares/cirugía , Potenciales de Acción , Adulto , Anciano , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/fisiopatología , Catéteres Cardíacos , Ablación por Catéter/efectos adversos , Ablación por Catéter/instrumentación , Diseño de Equipo , Esófago/lesiones , Femenino , Atrios Cardíacos/fisiopatología , Frecuencia Cardíaca , Humanos , Masculino , Persona de Mediana Edad , Monitoreo Intraoperatorio/métodos , Venas Pulmonares/fisiopatología , Recurrencia , Reoperación , Factores de Riesgo , Irrigación Terapéutica/efectos adversos , Irrigación Terapéutica/instrumentación , Termometría , Factores de Tiempo , Resultado del Tratamiento
14.
J Neuroinflammation ; 13(1): 242, 2016 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-27612942

RESUMEN

BACKGROUND: Neurological disorders have been linked to abnormal excitatory neurotransmission. Perturbations in glutamate cycling can have profound impacts on normal activity, lead to excitotoxicity and neuroinflammation, and induce and/or exacerbate impairments in these diseases. Astrocytes play a key role in excitatory signaling as they both clear glutamate from the synaptic cleft and house enzymes responsible for glutamate conversion to glutamine. However, mechanisms responsible for the regulation of glutamate cycling, including the main astrocytic glutamate transporter excitatory amino acid transporter 2 (EAAT2 or GLT-1 in rodents) and glutamine synthetase (GS) which catalyzes the ATP-dependent reaction of glutamate and ammonia into glutamine, remain largely undefined. METHODS: Gain and loss of function for ß-catenin in human progenitor-derived astrocyte (PDAs) was used to assess EAAT2 and GS levels by PCR, western blot, luciferase reporter assays, and chromatin immunoprecipitation (ChIP). Further, morpholinos were stereotaxically injected into C57BL/6 mice and western blots measured the protein levels of ß-catenin, GLT-1, and GS. RESULTS: ß-Catenin, a transcriptional co-activator and the central mediator of Wnt/ß-catenin signaling pathway, positively regulates EAAT2 and GS at the transcriptional level in PDAs by partnering with T cell factor 1 (TCF-1) and TCF-3, respectively. This pathway is conserved in vivo as the knockdown of ß-catenin in the prefrontal cortex results in reduced GLT-1 and GS expression. CONCLUSIONS: These studies confirm that ß-catenin regulates key proteins responsible for excitatory glutamate neurotransmission in vitro and in vivo and reveal the therapeutic potential of ß-catenin modulation in treating diseases with abnormal glutamatergic neurotransmission and excitotoxicity.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Ácido Glutámico/metabolismo , beta Catenina/metabolismo , Animales , Encéfalo/efectos de los fármacos , Células Cultivadas , Inmunoprecipitación de Cromatina , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Ratones , Morfolinos/farmacología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Transfección , beta Catenina/genética
15.
Glia ; 63(3): 441-51, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25331637

RESUMEN

HIV-mediated neuropathogenesis is a multifaceted process involving several players, including resident brain cells (neurons, astrocytes, and microglia) and infiltrating cells [peripheral blood mononuclear cells (PBMCs)]. We evaluated the dynamic interaction between astrocytes and infiltrating PBMCs as it impacts HIV in the CNS. We demonstrate that human primary-derived astrocytes (PDAs) predominantly secrete Wnt 1, 2b, 3, 5b, and 10b. Wnts are small secreted glycoproteins that initiate either ß-catenin-dependent or independent signal transduction. The Wnt pathway plays a vital role in the regulation of CNS activities including neurogenesis, neurotransmitter release, synaptic plasticity, and memory consolidation. We show that HIV infection of PDAs altered astrocyte Wnt profile by elevating Wnts 2b and 10b. Astrocyte conditioned media (ACM) inhibited HIV replication in PBMCs by 50%. Removal of Wnts from ACM abrogated its ability to suppress HIV replication in PBMCs. Inversely, PBMCs supernatant activated PDAs, as demonstrated by a 10-fold increase in HLA-DR and a 5-fold increase in IFNγ expression, and enhanced astrocyte susceptibility to HIV by 2-fold, which was mediated by IFNγ in a Stat-3-dependent manner. Collectively, these data demonstrate a dynamic interaction between astrocytes and PBMCs, whereby astrocyte-secreted Wnts exert an anti-HIV effect on infected PBMCs and PBMCs, in turn, secrete IFNγ that enhance astrocyte susceptibility to productive HIV infection and mediate their activation.


Asunto(s)
Complejo SIDA Demencia/fisiopatología , Astrocitos/fisiología , Leucocitos Mononucleares/fisiología , Complejo SIDA Demencia/virología , Astrocitos/virología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados , VIH/fisiología , Antígenos HLA-DR/metabolismo , Humanos , Interferón gamma/metabolismo , Leucocitos Mononucleares/virología , Factor de Transcripción STAT3/metabolismo , Proteínas Wnt/metabolismo
16.
J Virol ; 88(5): 3031-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24352441

RESUMEN

HIV infiltrates the brain at early times postinfection and remains latent within astrocytes and macrophages. Because astrocytes are the most abundant cell type in the brain, we evaluated epigenetic regulation of HIV latency in astrocytes. We have shown that class I histone deacetylases (HDACs) and a lysine-specific histone methyltransferase, SU(VAR)3-9, play a significant role in silencing of HIV transcription in astrocytes. Our studies add to a growing body of evidence demonstrating that astrocytes are a reservoir for HIV.


Asunto(s)
Astrocitos/virología , Epigénesis Genética , Regulación Viral de la Expresión Génica , VIH-1/fisiología , Latencia del Virus/genética , Astrocitos/metabolismo , Línea Celular , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica/efectos de los fármacos , Infecciones por VIH/virología , Duplicado del Terminal Largo de VIH , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Piperazinas/farmacología , Regiones Promotoras Genéticas , Transcripción Genética/efectos de los fármacos , Integración Viral
17.
Arterioscler Thromb Vasc Biol ; 33(6): 1162-70, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23559625

RESUMEN

OBJECTIVE: Atherosclerotic lesions are associated with the accumulation of reactive aldehydes derived from oxidized lipids. Although inhibition of aldehyde metabolism has been shown to exacerbate atherosclerosis and enhance the accumulation of aldehyde-modified proteins in atherosclerotic plaques, no therapeutic interventions have been devised to prevent aldehyde accumulation in atherosclerotic lesions. APPROACH AND RESULTS: We examined the efficacy of carnosine, a naturally occurring ß-alanyl-histidine dipeptide, in preventing aldehyde toxicity and atherogenesis in apolipoprotein E-null mice. In vitro, carnosine reacted rapidly with lipid peroxidation-derived unsaturated aldehydes. Gas chromatography mass-spectrometry analysis showed that carnosine inhibits the formation of free aldehydes 4-hydroxynonenal and malonaldialdehyde in Cu(2+)-oxidized low-density lipoprotein. Preloading bone marrow-derived macrophages with cell-permeable carnosine analogs reduced 4-hydroxynonenal-induced apoptosis. Oral supplementation with octyl-D-carnosine decreased atherosclerotic lesion formation in aortic valves of apolipoprotein E-null mice and attenuated the accumulation of protein-acrolein, protein-4-hydroxyhexenal, and protein-4-hydroxynonenal adducts in atherosclerotic lesions, whereas urinary excretion of aldehydes as carnosine conjugates was increased. CONCLUSIONS: The results of this study suggest that carnosine inhibits atherogenesis by facilitating aldehyde removal from atherosclerotic lesions. Endogenous levels of carnosine may be important determinants of atherosclerotic lesion formation, and treatment with carnosine or related peptides could be a useful therapy for the prevention or the treatment of atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Carnosina/farmacología , Suplementos Dietéticos , Peroxidación de Lípido/fisiología , Lipoproteínas LDL/metabolismo , Aldehídos/metabolismo , Animales , Apolipoproteínas E/deficiencia , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Distribución Aleatoria , Valores de Referencia
18.
Sci Rep ; 14(1): 6521, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38499637

RESUMEN

Grid computing emerged as a powerful computing domain for running large-scale parallel applications. Scheduling computationally intensive parallel applications such as scientific, commercial etc., computational grids is a NP-complete problem. Many researchers have proposed several task scheduling algorithms on grids based on formulating and solving it as an optimization problem with different objective functions such as makespan, cost, energy etc. Further to address the requirements/demands/needs of the users (lesser cost, lower latency etc.) and grid service providers (high utilization and high profitability), a task scheduler needs to be designed based on solving a multi-objective optimization problem due to several trade-offs among the objective functions. In this direction, we propose an efficient multi-objective task scheduling framework to schedule computationally intensive tasks on heterogeneous grid networks. This framework minimizes turnaround time, communication, and execution costs while maximizing grid utilization. We evaluated the performance of our proposed algorithm through experiments conducted on standard, random, and scientific task graphs using the GridSim simulator.

19.
JBMR Plus ; 8(5): ziae037, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38590756

RESUMEN

Bone mineral density (BMD) loss in people living with HIV occurs with the initiation of combined antiretroviral therapy (cART), particularly with tenofovir disoproxil fumarate (TDF) containing cART. Switching from TDF to abacavir (ABC) or dolutegravir (DTG) leads to increased BMD. Whether BMD gains are due to cessation of TDF or anabolic effects of ABC or DTG is unclear. We investigated the effects of ABC and DTG on osteoblast lineage cells in vitro and in vivo. Primary human osteoblasts and male C57BL/6 mice were treated with individual antiretrovirals (ARVs) or a combination of ABC/DTG/lamivudine (3TC). Nearly all ARVs and cART inhibited osteogenic activity in vitro. Due to the importance of Wnt/ß-catenin in bone formation, we further investigated ARV effects on the Wnt/ß-catenin pathway. ABC, alone and as part of ABC/DTG/3TC, increased osteoblastic ß-catenin activity as indicated by increased TOPFlash activity, hypo-phosphorylated (active) ß-catenin staining, and ß-catenin targeted gene expression. Mice treated with TDF had decreased lumbar spine BMD and trabecular connectivity density in the vertebrae, while those treated with ABC/DTG/3TC reduced cortical area and thickness in the femur. Mice treated with ABC alone had no bone structural changes, increased circulating levels of the bone formation marker, P1NP, and elevated expression of the Wnt/ß-catenin target gene, Lef1, in osteocyte enriched samples. Further, bones from ARV-treated mice were isolated to evaluate ARV distribution. All ARVs were detected in the bone tissue, which was inclusive of bone marrow, but when bone marrow was removed, only TDF, ABC, and DTG were detected at ~0.1% of the circulating levels. Overall, our findings demonstrate that ABC activates Wnt/ß-catenin signaling, but whether this leads to increased bone formation requires further study. Assessing the impact of ARVs on bone is critical to informing ARV selection and/or discovery of regimens that do not negatively impact the skeleton.

20.
bioRxiv ; 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38915632

RESUMEN

Cerebral organoids (COs) are a valuable tool to study the intricate interplay between glial cells and neurons in brain development and disease, including HIV-associated neuroinflammation. We developed a novel approach to generate microglia containing COs (CO-iMs) by co-culturing hematopoietic progenitors and induced pluripotent stem cells. This approach allowed for the differentiation of microglia within the organoids concomitantly to the neuronal progenitors. CO- iMs exhibited higher efficiency in generation of CD45 + /CD11b + /Iba-1 + microglia cells compared to conventional COs with physiologically relevant proportion of microglia (∼7%). CO-iMs exhibited substantially higher expression of microglial homeostatic and sensome markers as well as markers for the complement cascade. CO-iMs showed susceptibility to HIV infection resulting in a significant increase in several pro-inflammatory cytokines/chemokines and compromised neuronal function, which were abrogated by addition of antiretrovirals. Thus, CO-iM is a robust model to decipher neuropathogenesis, neurological disorders, and viral infections of brain cells in a 3D culture system.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA