Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(15): 7439-7448, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30910955

RESUMEN

Cellular metabolism and signaling pathways are key regulators to determine conventional T cell fate and function, but little is understood about the role of cell metabolism for natural killer T (NKT) cell survival, proliferation, and function. We found that NKT cells operate distinct metabolic programming from CD4 T cells. NKT cells are less efficient in glucose uptake than CD4 T cells with or without activation. Gene-expression data revealed that, in NKT cells, glucose is preferentially metabolized by the pentose phosphate pathway and mitochondria, as opposed to being converted into lactate. In fact, glucose is essential for the effector functions of NKT cells and a high lactate environment is detrimental for NKT cell survival and proliferation. Increased glucose uptake and IFN-γ expression in NKT cells is inversely correlated with bacterial loads in response to bacterial infection, further supporting the significance of glucose metabolism for NKT cell function. We also found that promyelocytic leukemia zinc finger seemed to play a role in regulating NKT cells' glucose metabolism. Overall, our study reveals that NKT cells use distinct arms of glucose metabolism for their survival and function.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Mitocondrias/metabolismo , Células T Asesinas Naturales/inmunología , Fosforilación Oxidativa , Vía de Pentosa Fosfato/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Glucosa/genética , Glucosa/inmunología , Ratones , Ratones Noqueados , Mitocondrias/genética , Células T Asesinas Naturales/citología , Vía de Pentosa Fosfato/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/inmunología
2.
PLoS Pathog ; 15(10): e1007903, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31584995

RESUMEN

HIV-1 gene expression is regulated by host and viral factors that interact with viral motifs and is influenced by proviral integration sites. Here, expression variation among integrants was followed for hundreds of individual proviral clones within polyclonal populations throughout successive rounds of virus and cultured cell replication, with limited findings using CD4+ cells from donor blood consistent with observations in immortalized cells. Tracking clonal behavior by proviral "zip codes" indicated that mutational inactivation during reverse transcription was rare, while clonal expansion and proviral expression states varied widely. By sorting for provirus expression using a GFP reporter in the nef open reading frame, distinct clone-specific variation in on/off proportions were observed that spanned three orders of magnitude. Tracking GFP phenotypes over time revealed that as cells divided, their progeny alternated between HIV transcriptional activity and non-activity. Despite these phenotypic oscillations, the overall GFP+ population within each clone was remarkably stable, with clones maintaining clone-specific equilibrium mixtures of GFP+ and GFP- cells. Integration sites were analyzed for correlations between genomic features and the epigenetic phenomena described here. Integrants inserted in the sense orientation of genes were more frequently found to be GFP negative than those in the antisense orientation, and clones with high GFP+ proportions were more distal to repressive H3K9me3 peaks than low GFP+ clones. Clones with low frequencies of GFP positivity appeared to expand more rapidly than clones for which most cells were GFP+, even though the tested proviruses were Vpr-. Thus, much of the increase in the GFP- population in these polyclonal pools over time reflected differential clonal expansion. Together, these results underscore the temporal and quantitative variability in HIV-1 gene expression among proviral clones that are conferred in the absence of metabolic or cell-type dependent variability, and shed light on cell-intrinsic layers of regulation that affect HIV-1 population dynamics.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Infecciones por VIH/virología , VIH-1/fisiología , Provirus/genética , Integración Viral/genética , Replicación Viral , Linfocitos T CD4-Positivos/metabolismo , Infecciones por VIH/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Jurkat , Transducción Genética
4.
Eur J Immunol ; 48(7): 1255-1257, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29572809

RESUMEN

We show the presence of lymphoid tissue-resident PLZF+ CD45RA+ RO+ CD4 T cells in humans. They express HLA-DR, granzyme B, and perforin and are low on CCR7 like terminally differentiated effector memory (Temra) cells and are likely generated from effector T cells (Te) or from central (Tcm) or effector (Tem) memory T (Tcm) cells during immune responses. Tn, Naïve T cells.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Genotipo , Tejido Linfoide/inmunología , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Subgrupos de Linfocitos T/fisiología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Células Cultivadas , Granzimas/metabolismo , Antígenos HLA-DR/metabolismo , Humanos , Inmunidad Celular , Memoria Inmunológica , Perforina/metabolismo
5.
J Immunol ; 199(10): 3478-3487, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29021374

RESUMEN

Reactive oxygen species (ROS) are byproducts of aerobic metabolism and contribute to both physiological and pathological conditions as second messengers. ROS are essential for activation of T cells, but how ROS influence NKT cells is unknown. In the present study, we investigated the role of ROS in NKT cell function. We found that NKT cells, but not CD4 or CD8 T cells, have dramatically high ROS in the spleen and liver of mice but not in the thymus or adipose tissues. Accordingly, ROS-high NKT cells exhibited increased susceptibility and apoptotic cell death with oxidative stress. High ROS in the peripheral NKT cells were primarily produced by NADPH oxidases and not mitochondria. We observed that sorted ROS-high NKT cells were enriched in NKT1 and NKT17 cells, whereas NKT2 cells were dominant in ROS-low cells. Furthermore, treatment of NKT cells with antioxidants led to reduced frequencies of IFN-γ- and IL-17-expressing cells, indicating that ROS play a role in regulating the inflammatory function of NKT cells. The transcription factor promyelocytic leukemia zinc finger (PLZF) seemed to control the ROS levels. NKT cells from adipose tissues that do not express PLZF and those from PLZF haplodeficient mice have low ROS. Conversely, ROS were highly elevated in CD4 T cells from mice ectopically expressing PLZF. Thus, our findings demonstrate that PLZF controls ROS levels, which in turn governs the inflammatory function of NKT cells.


Asunto(s)
Hígado/inmunología , Células T Asesinas Naturales/inmunología , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Bazo/inmunología , Animales , Apoptosis , Células Cultivadas , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética
6.
J Hepatol ; 67(1): 100-109, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28267623

RESUMEN

BACKGROUND & AIMS: The liver is an immunologically-privileged organ. Breakdown of liver immune privilege has been reported in chronic liver disease; however, the role of adaptive immunity in liver injury is poorly defined. Nuclear factor-κB-inducing kinase (NIK) is known to regulate immune tissue development, but its role in maintaining liver homeostasis remains unknown. This study aimed to assess the role of NIK, particularly thymic NIK, in regulating liver adaptive immunity. METHODS: NIK was deleted systemically or conditionally using the Cre/loxp system. Cluster of differentiation [CD]4+ or CD8+ T cells were depleted using anti-CD4 or anti-CD8 antibody. Donor bone marrows or thymi were transferred into recipient mice. Immune cells were assessed by immunohistochemistry and flow cytometry. RESULTS: Global, but not liver-specific or hematopoietic lineage cell-specific, deletion of NIK induced fatal liver injury, inflammation, and fibrosis. Likewise, adoptive transfer of NIK-null, but not wild-type, thymi into immune-deficient mice induced liver inflammation, injury, and fibrosis in recipients. Liver inflammation was characterized by a massive expansion of T cells, particularly the CD4+ T cell subpopulation. Depletion of CD4+, but not CD8+, T cells fully protected against liver injury, inflammation, and fibrosis in NIK-null mice. NIK deficiency also resulted in inflammation in the lung, kidney, and pancreas, but to a lesser degree relative to the liver. CONCLUSIONS: Thymic NIK suppresses development of autoreactive T cells against liver antigens, and NIK deficiency in the thymus results in CD4+ T cell-orchestrated autoimmune hepatitis and liver fibrosis. Thus, thymic NIK is essential for the maintenance of liver immune privilege and liver homeostasis. LAY SUMMARY: We found that global or thymus-specific ablation of the NIK gene results in fatal autoimmune liver disease in mice. NIK-deficient mice develop liver inflammation, injury, and fibrosis. Our findings indicate that thymic NIK is essential for the maintenance of liver integrity and homeostasis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Hepatitis Autoinmune/etiología , Cirrosis Hepática Experimental/etiología , Hígado/inmunología , Proteínas Serina-Treonina Quinasas/fisiología , Timo/fisiología , Inmunidad Adaptativa , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinasa de Factor Nuclear kappa B
7.
J Immunol ; 194(1): 223-30, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25404366

RESUMEN

The mammalian target of rapamycin (mTOR) senses and incorporates different environmental cues via the two signaling complexes mTOR complex 1 (mTORC1) and mTORC2. As a result, mTOR controls cell growth and survival, and also shapes different effector functions of the cells including immune cells such as T cells. We demonstrate in this article that invariant NKT (iNKT) cell development is controlled by mTORC2 in a cell-intrinsic manner. In mice deficient in mTORC2 signaling because of the conditional deletion of the Rictor gene, iNKT cell numbers were reduced in the thymus and periphery. This is caused by decreased proliferation of stage 1 iNKT cells and poor development through subsequent stages. Functionally, iNKT cells devoid of mTORC2 signaling showed reduced number of IL-4-expressing cells, which correlated with a decrease in the transcription factor GATA-3-expressing cells. However, promyelocytic leukemia zinc-finger (PLZF), a critical transcription factor for iNKT cell development, is expressed at a similar level in mTORC2-deficient iNKT cells compared with that in the wild type iNKT cells. Furthermore, cellular localization of PLZF was not altered in the absence of mTOR2 signaling. Thus, our study reveals the PLZF-independent mechanisms of the development and function of iNKT cells regulated by mTORC2.


Asunto(s)
Proteínas Portadoras/inmunología , Factor de Transcripción GATA3/biosíntesis , Factores de Transcripción de Tipo Kruppel/biosíntesis , Complejos Multiproteicos/inmunología , Células T Asesinas Naturales/citología , Serina-Treonina Quinasas TOR/inmunología , Animales , Proteínas Portadoras/genética , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Interleucina-4/biosíntesis , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/genética , Células T Asesinas Naturales/inmunología , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal/genética , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/genética
8.
BMC Immunol ; 16: 62, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26482437

RESUMEN

BACKGROUND: Invariant Natural Killer T (iNKT) cells have been implicated in lung inflammation in humans and also shown to be a key cell type in inducing allergic lung inflammation in mouse models. iNKT cells differentiate and acquire functional characteristics during development in the thymus. However, the correlation between development of iNKT cells in the thymus and role in lung inflammation remains unknown. In addition, transcriptional control of differentiation of iNKT cells into iNKT cell effector subsets in the thymus during development is also unclear. In this report we show that ß-catenin dependent mechanisms direct differentiation of iNKT2 and iNKT17 subsets but not iNKT1 cells. METHODS: To study the role for ß-catenin in lung inflammation we utilize mice with conditional deletion and enforced expression of ß-catenin in a well-established mouse model for IL-25-dependen lung inflammation. RESULTS: Specifically, we demonstrate that conditional deletion of ß-catenin permitted development of mature iNKT1 cells while impeding maturation of iNKT2 and 17 cells. A role for ß-catenin expression in promoting iNKT2 and iNKT17 subsets was confirmed when we noted that enforced transgenic expression of ß-catenin in iNKT cell precursors enhanced the frequency and number of iNKT2 and iNKT17 cells at the cost of iNKT1 cells. This effect of expression of ß-catenin in iNKT cell precursors was cell autonomous. Furthermore, iNKT2 cells acquired greater capability to produce type-2 cytokines when ß-catenin expression was enhanced. DISCUSSION: This report shows that ß-catenin deficiency resulted in a profound decrease in iNKT2 and iNKT17 subsets of iNKT cells whereas iNKT1 cells developed normally. By contrast, enforced expression of ß-catenin promoted the development of iNKT2 and iNKT17 cells. It was important to note that the majority of iNKT cells in the thymus of C57BL/6 mice were iNKT1 cells and enforced expression of ß-catenin altered the pattern to iNKT2 and iNKT17 cells suggesting that ß-catenin may be a major factor in the distinct pathways that critically direct differentiation of iNKT effector subsets. CONCLUSIONS: Thus, we demonstrate that ß-catenin expression in iNKT cell precursors promotes differentiation toward iNKT2 and iNKT17 effector subsets and supports enhanced capacity to produce type 2 and 17 cytokines which in turn augment lung inflammation in mice.


Asunto(s)
Diferenciación Celular , Interleucina-17/metabolismo , Células T Asesinas Naturales/inmunología , Neumonía/inmunología , Neumonía/patología , beta Catenina/metabolismo , Animales , Hiperreactividad Bronquial/complicaciones , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Neumonía/complicaciones
9.
J Immunol ; 189(3): 1431-9, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22732591

RESUMEN

Variola and vaccinia viruses, the two most important members of the family Poxviridae, are known to encode homologs of the human complement regulators named smallpox inhibitor of complement enzymes (SPICE) and vaccinia virus complement control protein (VCP), respectively, to subvert the host complement system. Intriguingly, consistent with the host tropism of these viruses, SPICE has been shown to be more human complement-specific than VCP, and in this study we show that VCP is more bovine complement-specific than SPICE. Based on mutagenesis and mechanistic studies, we suggest that the major determinant for the switch in species selectivity of SPICE and VCP is the presence of oppositely charged residues in the central complement control modules, which help enhance their interaction with factor I and C3b, the proteolytically cleaved form of C3. Thus, our results provide a molecular basis for the species selectivity in poxviral complement regulators.


Asunto(s)
Complemento C3b/antagonistas & inhibidores , Vía Alternativa del Complemento/inmunología , Virus Vaccinia/inmunología , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Sustitución de Aminoácidos/inmunología , Animales , Bovinos , Complemento C3b/genética , Complemento C3b/metabolismo , Factor I de Complemento/antagonistas & inhibidores , Factor I de Complemento/metabolismo , Vía Alternativa del Complemento/genética , Humanos , Evasión Inmune/genética , Datos de Secuencia Molecular , Especificidad de la Especie , Electricidad Estática , Virus Vaccinia/metabolismo , Proteínas Virales/genética
10.
bioRxiv ; 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38712097

RESUMEN

Upon antigenic stimulation, CD4 + T-cells undergo clonal expansion, elevating their bioenergetic demands and utilization of nutrients like glucose and glutamine. The nuclear factor erythroid 2-related factor 2 (Nrf2) is a well-known regulator of oxidative stress, but its involvement in modulating the metabolism of CD4 + T-cells remains unexplored. Here, we elucidate the role of Nrf2 beyond the traditional antioxidation, in modulating activation-driven expansion of CD4 + T-cells by influencing their nutrient metabolism. T-cell-specific activation of Nrf2 enhances early activation and IL-2 secretion, upregulates TCR-signaling, and increases activation-driven proliferation of CD4 + T-cells. Mechanistically, high Nrf2 inhibits glucose metabolism through glycolysis but promotes glutamine metabolism via glutaminolysis to support increased T-cell proliferation. Further, Nrf2 expression is temporally regulated in activated CD4 + T-cells with elevated expression during the early activation, but decreased expression thereafter. Overall, our findings uncover a novel role of Nrf2 as a metabolic modulator of CD4 + T-cells, thus providing a framework for improving Nrf2-targeting therapies and T-cell immunotherapies.

11.
Clin Dev Immunol ; 2013: 617809, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24416060

RESUMEN

Developmental endothelial locus-1 (Del-1) is an endothelial cell-secreted protein that limits the recruitment of neutrophils by antagonizing the interaction between the LFA-1 integrin on neutrophils and the intercellular adhesion molecule (ICAM)-1 on endothelial cells. Mice with genetic or age-associated Del-1 deficiency exhibit increased neutrophil infiltration in the periodontium resulting in inflammatory bone loss. Here we investigated additional novel mechanisms whereby Del-1 could interfere with neutrophil recruitment and inflammation. Treatment of human endothelial cells with Del-1 did not affect the expression of endothelial molecules involved in the leukocyte adhesion cascade (ICAM-1, VCAM-1, and E-selectin). Moreover, genetic or age-associated Del-1 deficiency did not significantly alter the expression of these adhesion molecules in the murine periodontium, further ruling out altered adhesion molecule expression as a mechanism whereby Del-1 regulates leukocyte recruitment. Strikingly, Del-1 inhibited ICAM-1-dependent chemokine release (CXCL2, CCL3) by neutrophils. Therefore, Del-1 could potentially suppress the amplification of inflammatory cell recruitment mediated through chemokine release by infiltrating neutrophils. Interestingly, Del-1 was itself regulated by inflammatory stimuli, which generally exerted opposite effects on adhesion molecule expression. The reciprocal regulation between Del-1 and inflammation may contribute to optimally balance the protective and the potentially harmful effects of inflammatory cell recruitment.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Periodoncio/metabolismo , Animales , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Quimiocinas/biosíntesis , Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Noqueados , Neutrófilos/metabolismo , Osteoporosis/genética , Osteoporosis/metabolismo , Periodontitis/genética , Periodontitis/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
Immunohorizons ; 7(4): 288-298, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-37099275

RESUMEN

Ubiquitously expressed in mammalian cells, the Kelch-like ECH-associated protein 1 (Keap1)-NF erythroid 2-related factor 2 (Nrf2) complex forms the evolutionarily conserved antioxidation system to tackle oxidative stress caused by reactive oxygen species. Reactive oxygen species, generated as byproducts of cellular metabolism, were identified as essential second messengers for T cell signaling, activation, and effector responses. Apart from its traditional role as an antioxidant, a growing body of evidence indicates that Nrf2, tightly regulated by Keap1, modulates immune responses and regulates cellular metabolism. Newer functions of Keap1 and Nrf2 in immune cell activation and function, as well as their role in inflammatory diseases such as sepsis, inflammatory bowel disease, and multiple sclerosis, are emerging. In this review, we highlight recent findings about the influence of Keap1 and Nrf2 in the development and effector functions of adaptive immune cells, that is, T cells and B cells, and discuss the knowledge gaps in our understanding. We also summarize the research potential and targetability of Nrf2 for treating immune pathologies.


Asunto(s)
Antioxidantes , Factor 2 Relacionado con NF-E2 , Animales , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Mamíferos/metabolismo
13.
J Immunol ; 185(10): 6128-37, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20956343

RESUMEN

Vaccinia virus encodes a structural and functional homolog of human complement regulators named vaccinia virus complement control protein (VCP). This four-complement control protein domain containing secretory protein is known to inhibit complement activation by supporting the factor I-mediated inactivation of complement proteins, proteolytically cleaved form of C3 (C3b) and proteolytically cleaved form of C4 (C4b) (termed cofactor activity), and by accelerating the irreversible decay of the classical and to a limited extent of the alternative pathway C3 convertases (termed decay-accelerating activity [DAA]). In this study, we have mapped the VCP domains important for its cofactor activity and DAA by swapping its individual domains with those of human decay-accelerating factor (CD55) and membrane cofactor protein (MCP; CD46). Our data indicate the following: 1) swapping of VCP domain 2 or 3, but not 1, with homologous domains of decay-accelerating factor results in loss in its C3b and C4b cofactor activities; 2) swapping of VCP domain 1, but not 2, 3, or 4 with corresponding domains of MCP results in abrogation in its classical pathway DAA; and 3) swapping of VCP domain 1, 2, or 3, but not 4, with homologous MCP domains have marked effect on its alternative pathway DAA. These functional data together with binding studies with C3b and C4b suggest that in VCP, domains 2 and 3 provide binding surface for factor I interaction, whereas domain 1 mediates dissociation of C2a and Bb from the classical and alternative pathway C3 convertases, respectively.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , Virus Vaccinia/química , Virus Vaccinia/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Antígenos CD55/química , Antígenos CD55/inmunología , Antígenos CD55/metabolismo , Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/inmunología , Electroforesis en Gel de Poliacrilamida , Humanos , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie , Virus Vaccinia/inmunología , Proteínas Virales/inmunología
14.
J Immunol ; 184(4): 1956-67, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20089702

RESUMEN

Kaposica, the complement regulator of Kaposi's sarcoma-associated herpesvirus, inhibits complement by supporting factor I-mediated inactivation of the proteolytically activated form of C3 (C3b) and C4 (C4b) (cofactor activity [CFA]) and by accelerating the decay of classical and alternative pathway C3-convertases (decay-accelerating activity [DAA]). Previous data suggested that electrostatic interactions play a critical role in the binding of viral complement regulators to their targets, C3b and C4b. We therefore investigated how electrostatic potential on Kaposica influences its activities. We built a homology structure of Kaposica and calculated the electrostatic potential of the molecule, using the Poisson-Boltzmann equation. Mutants were then designed to alter the overall positive potential of the molecule or of each of its domains and linkers by mutating Lys/Arg to Glu/Gln, and the functional activities of the expressed mutants were analyzed. Our data indicate that 1) positive potential at specific sites and not the overall positive potential on the molecule guides the CFAs and classical pathway DAA; 2) positive potential around the linkers between complement control protein domains (CCPs) 1-2 and 2-3 is more important for DAAs than for CFAs; 3) positive potential in CCP1 is crucial for binding to C3b and C4b, and thereby its functional activities; 4) conversion to negative or enhancement of negative potential for CCPs 2-4 has a marked effect on C3b-linked activities as opposed to C4b-linked activities; and 5) reversal of the electrostatic potential of CCP4 to negative has a differential effect on classical and alternative pathway DAAs. Together, our data provide functional relevance to conservation of positive potential in CCPs 1 and 4 and the linkers of viral complement regulators.


Asunto(s)
Proteínas Inactivadoras de Complemento/fisiología , Herpesvirus Humano 8/inmunología , Electricidad Estática , Proteínas Virales/fisiología , Proteínas Inactivadoras de Complemento/genética , Vía Alternativa del Complemento/genética , Vía Alternativa del Complemento/inmunología , Herpesvirus Humano 8/genética , Humanos , Mutagénesis Sitio-Dirigida , Proteínas Virales/genética
15.
Cell Rep ; 38(2): 110216, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021084

RESUMEN

ATRX, a chromatin remodeler protein, is recurrently mutated in H3F3A-mutant pediatric glioblastoma (GBM) and isocitrate dehydrogenase (IDH)-mutant grade 2/3 adult glioma. Previous work has shown that ATRX-deficient GBM cells show enhanced sensitivity to irradiation, but the etiology remains unclear. We find that ATRX binds the regulatory elements of cell-cycle phase transition genes in GBM cells, and there is a marked reduction in Checkpoint Kinase 1 (CHEK1) expression with ATRX loss, leading to the early release of G2/M entry after irradiation. ATRX-deficient cells exhibit enhanced activation of master cell-cycle regulator ATM with irradiation. Addition of the ATM inhibitor AZD0156 doubles median survival in mice intracranially implanted with ATRX-deficient GBM cells, which is not seen in ATRX-wild-type controls. This study demonstrates that ATRX-deficient high-grade gliomas (HGGs) display Chk1-mediated dysregulation of cell-cycle phase transitions, which opens a window for therapies targeting this phenotype.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Glioma/metabolismo , Proteína Nuclear Ligada al Cromosoma X/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Ciclo Celular/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/fisiología , Femenino , Histonas/metabolismo , Humanos , Isocitrato Deshidrogenasa/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Recurrencia Local de Neoplasia/metabolismo , Cultivo Primario de Células , Proteína Nuclear Ligada al Cromosoma X/genética
16.
J Virol ; 83(19): 10299-304, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19640995

RESUMEN

Herpesvirus saimiri encodes a functional homolog of human regulator-of-complement-activation proteins named CCPH that inactivates complement by accelerating the decay of C3 convertases and by serving as a cofactor in factor I-mediated inactivation of their subunits C3b and C4b. Here, we map the functional domains of CCPH. We demonstrate that short consensus repeat 2 (SCR2) is the minimum domain essential for classical/lectin pathway C3 convertase decay-accelerating activity as well as for factor I cofactor activity for C3b and C4b. Thus, CCPH is the first example wherein a single SCR domain has been shown to display complement regulatory functions.


Asunto(s)
Proteínas del Sistema Complemento/química , Herpesvirus Saimiriino 2/metabolismo , Biotinilación , Complemento C3b/química , Complemento C4b/química , Vía Alternativa del Complemento , Electroforesis en Gel de Poliacrilamida , Eliminación de Gen , Humanos , Cinética , Lectinas/química , Ligandos , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes/química
17.
J Virol ; 82(7): 3283-94, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18216095

RESUMEN

Variola virus, the causative agent of smallpox, encodes a soluble complement regulator named SPICE. Previously, SPICE has been shown to be much more potent in inactivating human complement than the vaccinia virus complement control protein (VCP), although they differ only in 11 amino acid residues. In the present study, we have expressed SPICE, VCP, and mutants of VCP by substituting each or more of the 11 non-variant VCP residues with the corresponding residue of SPICE to identify hot spots that impart functional advantage to SPICE over VCP. Our data indicate that (i) SPICE is approximately 90-fold more potent than VCP in inactivating human C3b, and the residues Y98, Y103, K108 and K120 are predominantly responsible for its enhanced activity; (ii) SPICE is 5.4-fold more potent in inactivating human C4b, and residues Y98, Y103, K108, K120 and L193 mainly dictate this increase; (iii) the classical pathway decay-accelerating activity of activity is only twofold higher than that of VCP, and the 11 mutations in SPICE do not significantly affect this activity; (iv) SPICE possesses significantly greater binding ability to human C3b compared to VCP, although its binding to human C4b is lower than that of VCP; (v) residue N144 is largely responsible for the increased binding of SPICE to human C3b; and (vi) the human specificity of SPICE is dictated primarily by residues Y98, Y103, K108, and K120 since these are enough to formulate VCP as potent as SPICE. Together, these results suggest that principally 4 of the 11 residues that differ between SPICE and VCP partake in its enhanced function against human complement.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Virus Vaccinia/inmunología , Virus de la Viruela/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Complemento C3b/antagonistas & inhibidores , Complemento C4b/antagonistas & inhibidores , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Alineación de Secuencia , Resonancia por Plasmón de Superficie , Proteínas Virales/metabolismo
18.
Cell Rep ; 27(3): 699-707.e4, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995469

RESUMEN

Kelch-like ECH-associated protein 1 (Keap1) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) proteins work in concert to regulate the levels of reactive oxygen species (ROS). The Keap1-Nrf2 antioxidant system also participates in T cell differentiation and inflammation, but its role in innate T cell development and functions remains unclear. We report that T cell-specific deletion of Keap1 results in defective development and reduced numbers of invariant natural killer T (NKT) cells in the thymus and the peripheral organs in a cell-intrinsic manner. The frequency of NKT2 and NKT17 cells increases while NKT1 decreases in these mice. Keap1-deficient NKT cells show increased rates of proliferation and apoptosis, as well as increased glucose uptake and mitochondrial function, but reduced ROS, CD122, and Bcl2 expression. In NKT cells deficient in Nrf2 and Keap1, all these phenotypic and metabolic defects are corrected. Thus, the Keap1-Nrf2 system contributes to NKT cell development and homeostasis by regulating cell metabolism.


Asunto(s)
Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Células T Asesinas Naturales/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Subunidad beta del Receptor de Interleucina-2/genética , Subunidad beta del Receptor de Interleucina-2/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/deficiencia , Proteína 1 Asociada A ECH Tipo Kelch/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Timo/metabolismo
19.
J Cancer Sci Ther ; 10(6)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30147849

RESUMEN

Type I or invariant natural killer T cells belong to a unique lineage of innate T cells, which express markers of both T lymphocytes and NK cells, namely T cell receptor (TCR) and NK1.1 (CD161C), respectively. Thus, apart from direct killing of target cells like NK cells, and they also produce a myriad of cytokines which modulate the adaptive immune responses. Unlike traditional T cells which carry a conventional αß TCR, NKT cells express semi-invariant TCR - Vα14-Jα18, coupled with Vß8, Vß7 and Vß2 in mice. In humans, the invariant TCR is composed of Vα24-Jα18, coupled with Vß11.

20.
Indian J Biochem Biophys ; 44(5): 331-43, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18341208

RESUMEN

The complement system is a principal bastion of innate immunity designed to combat a myriad of existing as well as newly emerging pathogens. Since viruses are obligatory intracellular parasites, they are continuously exposed to host complement assault and, therefore, have imbibed various strategies to subvert it. One of them is molecular mimicry of the host complement regulators. Large DNA viruses such as pox and herpesviruses encode proteins that are structurally and functionally similar to human regulators of complement activation (RCA), a family of proteins that regulate complement. In this review, we have presented the structural and functional aspects of virally encoded RCA homologs (vRCA), in particular two highly studied vRCAs, vaccinia virus complement control protein (VCP) and Kaposi's sarcoma-associated herpesvirus complement regulator (kaposica). Importance of these evasion molecules in viral pathogenesis and their role beyond complement regulation are also discussed.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Inmunidad Innata/inmunología , Modelos Moleculares , Imitación Molecular/inmunología , Proteínas Virales/inmunología , Virus/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA