Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell Immunol ; 401-402: 104837, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38810592

RESUMEN

The activation of macrophages, essential for the innate defense against invading pathogens, revolves around Toll-like receptors (TLRs). Nevertheless, a comprehensive understanding of the molecular mechanisms governing TLR signaling in the course of macrophage activation remains to be fully clarified. Although Zc3h12c was originally identified as being enriched in organs associated with macrophages, its precise function remains elusive. In this study, we observed a significant induction of Zc3h12c in macrophages following stimulation with TLR agonists and pathogens. Overexpression of Zc3h12c significantly mitigated the release of TNF-α and IL-6 triggered by lipopolysaccharide (LPS), whereas depletion of Zc3h12c increased the production of the cytokines mentioned above. Notably, the expression of IFN-ß was not influenced by Zc3h12c. Luciferase reporter assays revealed that Zc3h12c could suppress the TNF-α promoter activity. Moreover, Zc3h12c exerted a notable inhibitory effect on JNK, ERK, p38, and NF-κB signaling induced by LPS. In summary, the findings of our study suggest that Zc3h12c functions as a robust suppressor of innate immunity, potentially playing a role in the pathogenesis of infectious diseases.


Asunto(s)
Inmunidad Innata , Lipopolisacáridos , Activación de Macrófagos , Macrófagos , Transducción de Señal , Factor de Necrosis Tumoral alfa , Inmunidad Innata/inmunología , Animales , Activación de Macrófagos/inmunología , Ratones , Lipopolisacáridos/farmacología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Transducción de Señal/inmunología , FN-kappa B/metabolismo , Receptores Toll-Like/metabolismo , Receptores Toll-Like/inmunología , Humanos , Interleucina-6/metabolismo , Interleucina-6/inmunología
2.
Proc Natl Acad Sci U S A ; 120(28): e2217301120, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37399423

RESUMEN

A common event upon receptor-ligand engagement is the formation of receptor clusters on the cell surface, in which signaling molecules are specifically recruited or excluded to form signaling hubs to regulate cellular events. These clusters are often transient and can be disassembled to terminate signaling. Despite the general relevance of dynamic receptor clustering in cell signaling, the regulatory mechanism underlying the dynamics is still poorly understood. As a major antigen receptor in the immune system, T cell receptors (TCR) form spatiotemporally dynamic clusters to mediate robust yet temporal signaling to induce adaptive immune responses. Here we identify a phase separation mechanism controlling dynamic TCR clustering and signaling. The TCR signaling component CD3ε chain can condensate with Lck kinase through phase separation to form TCR signalosomes for active antigen signaling. Lck-mediated CD3ε phosphorylation, however, switched its binding preference to Csk, a functional suppressor of Lck, to cause the dissolvement of TCR signalosomes. Modulating TCR/Lck condensation by targeting CD3ε interactions with Lck or Csk directly affects T cell activation and function, highlighting the importance of the phase separation mechanism. The self-programmed condensation and dissolvement is thus a built-in mechanism of TCR signaling and might be relevant to other receptors.


Asunto(s)
Proteína Tirosina Quinasa p56(lck) Específica de Linfocito , Receptores de Antígenos de Linfocitos T , Transducción de Señal/fisiología , Fosforilación , Antígenos/metabolismo
5.
Biomolecules ; 11(6)2021 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-34067359

RESUMEN

DNMT3A mutations are frequently identified in acute myeloid leukemia (AML) and indicate poor prognosis. Previously, we found that the hotspot mutation DNMT3A R882H could upregulate CDK1 and induce AML in conditional knock-in mice. However, the mechanism by which CDK1 is involved in leukemogenesis of DNMT3A mutation-related AML, and whether CDK1 could be a therapeutic target, remains unclear. In this study, using fluorescence resonance energy transfer and immunoprecipitation analysis, we discovered that increased CDK1 could compete with EZH2 to bind to the PHD-like motif of DNMT3A, which may disturb the protein interaction between EZH2 and DNMT3A. Knockdown of CDK1 in OCI-AML3 cells with DNMT3A mutation markedly inhibited proliferation and induced apoptosis. CDK1 selective inhibitor CGP74514A (CGP) and the pan-CDK inhibitor flavopiridol (FLA) arrested OCI-AML3 cells in the G2/M phase, and induced cell apoptosis. CGP significantly increased CD163-positive cells. Moreover, the combined application of CDK1 inhibitor and traditional chemotherapy drugs synergistically inhibited proliferation and induced apoptosis of OCI-AML3 cells. In conclusion, this study highlights CDK1 overexpression as a pathogenic factor and a potential therapeutic target for DNMT3A mutation-related AML.


Asunto(s)
Proteína Quinasa CDC2/biosíntesis , Carcinogénesis/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/metabolismo , Mutación , Proteínas de Neoplasias/metabolismo , Animales , Proteína Quinasa CDC2/genética , Carcinogénesis/genética , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Proteína Potenciadora del Homólogo Zeste 2/genética , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Células 3T3 NIH , Proteínas de Neoplasias/genética
6.
J Immunol ; 206(10): 2353-2365, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33941656

RESUMEN

IL-17A plays an essential role in the pathogenesis of many autoimmune diseases, including psoriasis and multiple sclerosis. Act1 is a critical adaptor in the IL-17A signaling pathway. In this study, we report that an anti-sense long noncoding RNA, TRAF3IP2-AS1, regulates Act1 expression and IL-17A signaling by recruiting SRSF10, which downregulates the expression of IRF1, a transcriptional factor of Act1. Interestingly, we found that a psoriasis-susceptible variant of TRAF3IP2-AS1 A4165G (rs13210247) is a gain-of-function mutant. Furthermore, we identified a mouse gene E130307A14-Rik that is homologous to TRAF3IP2-AS1 and has a similar ability to regulate Act1 expression and IL-17A signaling. Importantly, treatment with lentiviruses expressing E130307A14-Rik or SRSF10 yielded therapeutic effects in mouse models of psoriasis and experimental autoimmune encephalomyelitis. These findings suggest that TRAF3IP2-AS1 and/or SRSF10 may represent attractive therapeutic targets in the treatment of IL-17-related autoimmune diseases, such as psoriasis and multiple sclerosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Interleucina-17/metabolismo , Psoriasis/metabolismo , ARN Largo no Codificante/metabolismo , ARN/metabolismo , Proteínas Represoras/metabolismo , Factores de Empalme Serina-Arginina/metabolismo , Transducción de Señal/genética , Animales , Proteínas de Ciclo Celular/genética , Técnicas de Inactivación de Genes , Células HaCaT , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN/genética , ARN Largo no Codificante/genética , Proteínas Represoras/genética , Factores de Empalme Serina-Arginina/genética , Transfección
7.
Proc Natl Acad Sci U S A ; 117(25): 14395-14404, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32513696

RESUMEN

Retinoic acid-inducible gene I (RIG-I) is up-regulated during granulocytic differentiation of acute promyelocytic leukemia (APL) cells induced by all-trans retinoic acid (ATRA). It has been reported that RIG-I recognizes virus-specific 5'-ppp-double-stranded RNA (dsRNA) and activates the type I interferons signaling pathways in innate immunity. However, the functions of RIG-I in hematopoiesis remain unclear, especially regarding its possible interaction with endogenous RNAs and the associated pathways that could contribute to the cellular differentiation and maturation. Herein, we identified a number of RIG-I-binding endogenous RNAs in APL cells following ATRA treatment, including the tripartite motif-containing protein 25 (TRIM25) messenger RNA (mRNA). TRIM25 encodes the protein known as an E3 ligase for ubiquitin/interferon (IFN)-induced 15-kDa protein (ISG15) that is involved in RIG-I-mediated antiviral signaling. We show that RIG-I could bind TRIM25 mRNA via its helicase domain and C-terminal regulatory domain, enhancing the stability of TRIM25 transcripts. RIG-I could increase the transcriptional expression of TRIM25 by caspase recruitment domain (CARD) domain through an IFN-stimulated response element. In addition, RIG-I activated other key genes in the ISGylation pathway by activating signal transducer and activator of transcription 1 (STAT1), including the modifier ISG15 and several enzymes responsible for the conjugation of ISG15 to protein substrates. RIG-I cooperated with STAT1/2 and interferon regulatory factor 1 (IRF1) to promote the activation of the ISGylation pathway. The integrity of ISGylation in ATRA or RIG-I-induced cell differentiation was essential given that knockdown of TRIM25 or ISG15 resulted in significant inhibition of this process. Our results provide insight into the role of the RIG-I-TRIM25-ISGylation axis in myeloid differentiation.


Asunto(s)
Diferenciación Celular , Citocinas/metabolismo , Proteína 58 DEAD Box/metabolismo , Granulocitos/fisiología , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/metabolismo , Línea Celular Tumoral , Citocinas/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Estabilidad del ARN , ARN Mensajero/metabolismo , Receptores Inmunológicos , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinas/genética , Regulación hacia Arriba
8.
BMC Cancer ; 19(1): 1072, 2019 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-31703632

RESUMEN

BACKGROUND: DNMT3A R882H, a frequent mutation in acute myeloid leukemia (AML), plays a critical role in malignant hematopoiesis. Recent findings suggest that DNMT3A mutant acts as a founder mutation and requires additional genetic events to induce full-blown AML. Here, we investigated the cooperation of mutant DNMT3A and NRAS in leukemogenesis by generating a double knock-in (DKI) mouse model harboring both Dnmt3a R878H and Nras G12D mutations. METHODS: DKI mice with both Dnmt3a R878H and Nras G12D mutations were generated by crossing Dnmt3a R878H knock-in (KI) mice and Nras G12D KI mice. Routine blood test, flow cytometry analysis and morphological analysis were performed to determine disease phenotype. RNA-sequencing (RNA-seq), RT-PCR and Western blot were carried out to reveal the molecular mechanism. RESULTS: The DKI mice developed a more aggressive AML with a significantly shortened lifespan and higher percentage of blast cells compared with KI mice expressing Dnmt3a or Nras mutation alone. RNA-seq analysis showed that Dnmt3a and Nras mutations collaboratively caused abnormal expression of a series of genes related to differentiation arrest and growth advantage. Myc transcription factor and its target genes related to proliferation and apoptosis were up-regulated, thus contributing to promote the process of leukemogenesis. CONCLUSION: This study showed that cooperation of DNMT3A mutation and NRAS mutation could promote the onset of AML by synergistically disturbing the transcriptional profiling with Myc pathway involvement in DKI mice.


Asunto(s)
Carcinogénesis/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Técnicas de Sustitución del Gen , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas de Unión al GTP Monoméricas/genética , Animales , Apoptosis/genética , Diferenciación Celular/genética , ADN Metiltransferasa 3A , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Longevidad/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Fenotipo , Proyectos Piloto , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , RNA-Seq , Transcripción Genética
9.
Proc Natl Acad Sci U S A ; 116(6): 2220-2225, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30659143

RESUMEN

Homoharringtonine (HHT), a known protein synthesis inhibitor, has an anti-myeloid leukemia effect and potentiates the therapeutic efficacy of anthracycline/cytarabine induction regimens for acute myelogenous leukemia (AML) with favorable and intermediate prognoses, especially in the t(8;21) subtype. Here we provide evidence showing that HHT inhibits the activity of leukemia-initiating cells (Lin-/Sca-1-/c-kit+; LICs) in a t(8;21) murine leukemia model and exerts a down-regulating effect on MYC pathway genes in human t(8;21) leukemia cells (Kasumi-1). We discovered that NF-κB repressing factor (NKRF) is bound directly by HHT via the second double-strand RNA-binding motif (DSRM2) domain, which is the nuclear localization signal of NKRF. A series of deletion and mutagenesis experiments mapped HHT direct binding sites to K479 and C480 amino acids in the DSRM2 domain. HHT treatment shifts NKRF from the nucleus (including nucleoli) to the cytoplasm by occupying the DSRM2 domain, strengthens the p65-NKRF interaction, and interferes with p65-p50 complex formation, thereby attenuating the transactivation activity of p65 on the MYC gene. Moreover, HHT significantly decreases the expression of KIT, a frequently mutated and/or highly expressed gene in t(8;21) AML, in concert with MYC down-regulation. Our work thus identifies a mechanism of action of HHT that is different from, but acts in concert with, the known mode of action of this compound. These results justify further clinical testing of HHT in AML.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Genes myc , Homoharringtonina/farmacología , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Biomarcadores de Tumor , Línea Celular Tumoral , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Homoharringtonina/química , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Represoras/química , Factor de Transcripción ReIA/metabolismo , Transcripción Genética , Translocación Genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Proc Natl Acad Sci U S A ; 114(20): 5237-5242, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28461508

RESUMEN

DNMT3A is frequently mutated in acute myeloid leukemia (AML). To explore the features of human AML with the hotspot DNMT3A R882H mutation, we generated Dnmt3a R878H conditional knockin mice, which developed AML with enlarged Lin-Sca1+cKit+ cell compartments. The transcriptome and DNA methylation profiling of bulk leukemic cells and the single-cell RNA sequencing of leukemic stem/progenitor cells revealed significant changes in gene expression and epigenetic regulatory patterns that cause differentiation arrest and growth advantage. Consistent with leukemic cell accumulation in G2/M phase, CDK1 was up-regulated due to mTOR activation associated with DNA hypomethylation. Overexpressed CDK1-mediated EZH2 phosphorylation resulted in an abnormal trimethylation of H3K27 profile. The mTOR inhibitor rapamycin elicited a significant therapeutic response in Dnmt3aR878H/WT mice.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Leucemia Mieloide Aguda/genética , Animales , Secuencia de Bases , Diferenciación Celular , Metilación de ADN , ADN Metiltransferasa 3A , Metilasas de Modificación del ADN/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica/métodos , Técnicas de Sustitución del Gen/métodos , Leucemia Mieloide Aguda/metabolismo , Ratones , Mutación , Serina-Treonina Quinasas TOR/metabolismo , Transcriptoma
11.
J Virol ; 90(22): 10414-10422, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27630226

RESUMEN

Herpes simplex virus 1 (HSV-1) remodels nuclear membranes during virus egress. Although the UL31 and UL34 proteins control nucleocapsid transit in infected cells, the molecular interactions required for their function are unclear. Here we report that the γ134.5 gene product of HSV-1 facilitates nucleocapsid release to the cytoplasm through bridging the UL31/UL34 complex, cellular p32, and protein kinase C. Unlike wild-type virus, an HSV mutant devoid of γ134.5 or its amino terminus is crippled for viral growth and release. This is attributable to a defect in virus nuclear egress. In infected cells, wild-type virus recruits protein kinase C to the nuclear membrane and triggers its activation, whereas the γ134.5 mutants fail to exert such an effect. Accordingly, the γ134.5 mutants are unable to induce phosphorylation and reorganization of lamin A/C. When expressed in host cells γ134.5 targets p32 and protein kinase C. Meanwhile, it communicates with the UL31/UL34 complex through UL31. Deletion of the amino terminus from γ134.5 disrupts its activity. These results suggest that disintegration of the nuclear lamina mediated by γ134.5 promotes HSV replication. IMPORTANCE: HSV nuclear egress is a key step that determines the outcome of viral infection. While the nuclear egress complex mediates capsid transit across the nuclear membrane, the regulatory components are not clearly defined in virus-infected cells. We report that the γ134.5 gene product, a virulence factor of HSV-1, facilitates nuclear egress cooperatively with cellular p32, protein kinase C, and the nuclear egress complex. This work highlights a viral mechanism that may contribute to the pathogenesis of HSV infection.


Asunto(s)
Herpesvirus Humano 1/metabolismo , Lamina Tipo A/metabolismo , Fosforilación/fisiología , Proteínas Virales/metabolismo , Liberación del Virus/fisiología , Animales , Cápside/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/virología , Chlorocebus aethiops , Citoplasma/metabolismo , Citoplasma/virología , Células HeLa , Humanos , Membrana Nuclear/metabolismo , Membrana Nuclear/virología , Lámina Nuclear/metabolismo , Lámina Nuclear/virología , Proteínas Nucleares/metabolismo , Nucleocápside/metabolismo , Proteína Quinasa C/metabolismo , Células Vero , Ensamble de Virus/fisiología
12.
Proc Natl Acad Sci U S A ; 112(49): 15084-9, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26598702

RESUMEN

Arsenic is highly effective for treating acute promyelocytic leukemia (APL) and has shown significant promise against many other tumors. However, although its mechanistic effects in APL are established, its broader anticancer mode of action is not understood. In this study, using a human proteome microarray, we identified 360 proteins that specifically bind arsenic. Among the most highly enriched proteins in this set are those in the glycolysis pathway, including the rate-limiting enzyme in glycolysis, hexokinase-1. Detailed biochemical and metabolomics analyses of the highly homologous hexokinase-2 (HK2), which is overexpressed in many cancers, revealed significant inhibition by arsenic. Furthermore, overexpression of HK2 rescued cells from arsenic-induced apoptosis. Our results thus strongly implicate glycolysis, and HK2 in particular, as a key target of arsenic. Moreover, the arsenic-binding proteins identified in this work are expected to serve as a valuable resource for the development of synergistic antitumor therapeutic strategies.


Asunto(s)
Arsénico/farmacología , Proteínas Portadoras/análisis , Hexoquinasa/antagonistas & inhibidores , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Arsénico/metabolismo , Trióxido de Arsénico , Arsenicales/farmacología , Proteínas Portadoras/metabolismo , Biología Computacional , Glucólisis , Humanos , Metabolómica , Datos de Secuencia Molecular , Óxidos/farmacología , Proteoma
13.
J Biol Chem ; 290(25): 15670-15678, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-25907557

RESUMEN

Herpes simplex virus 1 (HSV-1) is the most prevalent human virus and causes global morbidity because the virus is able to infect multiple cell types. Remarkably, HSV infection switches between lytic and latent cycles, where T cells play a critical role. However, the precise way of virus-host interactions is incompletely understood. Here we report that HSV-1 productively infected Jurkat T-cells and inhibited antigen-induced T cell receptor activation. We discovered that HSV-1-encoded Us3 protein interrupted TCR signaling and interleukin-2 production by inactivation of the linker for activation of T cells. This study unveils a mechanism by which HSV-1 intrudes into early events of TCR-mediated cell signaling and may provide novel insights into HSV infection, during which the virus escapes from host immune surveillance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Proteínas de la Membrana/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología , Proteínas Virales/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Herpes Simple/genética , Herpes Simple/patología , Herpesvirus Humano 1/genética , Humanos , Evasión Inmune/genética , Interleucina-2/genética , Interleucina-2/inmunología , Células Jurkat , Proteínas de la Membrana/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores de Antígenos de Linfocitos T , Transducción de Señal/genética , Linfocitos T/patología , Linfocitos T/virología , Factor 6 Asociado a Receptor de TNF/genética , Proteínas Virales/genética
14.
J Biol Chem ; 289(52): 35795-805, 2014 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-25355318

RESUMEN

As a large double-stranded DNA virus, herpes simplex virus type 1 (HSV-1) assembles capsids in the nucleus where the viral particles exit by budding through the inner nuclear membrane. Although a number of viral and host proteins are involved, the machinery of viral egress is not well understood. In a search for host interacting proteins of ICP34.5, which is a virulence factor of HSV-1, we identified a cellular protein, p32 (gC1qR/HABP1), by mass spectrophotometer analysis. When expressed, ICP34.5 associated with p32 in mammalian cells. Upon HSV-1 infection, p32 was recruited to the inner nuclear membrane by ICP34.5, which paralleled the phosphorylation and rearrangement of nuclear lamina. Knockdown of p32 in HSV-1-infected cells significantly reduced the production of cell-free viruses, suggesting that p32 is a mediator of HSV-1 nuclear egress. These observations suggest that the interaction between HSV-1 ICP34.5 and p32 leads to the disintegration of nuclear lamina and facilitates the nuclear egress of HSV-1 particles.


Asunto(s)
Proteínas Portadoras/metabolismo , Núcleo Celular/virología , Herpesvirus Humano 1/fisiología , Proteínas Mitocondriales/metabolismo , Proteínas Virales/metabolismo , Animales , Chlorocebus aethiops , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Receptores Citoplasmáticos y Nucleares/metabolismo , Células Vero , Liberación del Virus , Receptor de Lamina B
15.
Proc Natl Acad Sci U S A ; 110(42): 17017-22, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24082129

RESUMEN

The 2-hydroxyglutarate (2-HG) has been reported to result from mutations of isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) genes and to function as an "oncometabolite." To evaluate the clinical significance of serum 2-HG levels in hematologic malignancies, acute myeloid leukemia (AML) in particular, we analyzed this metabolite in distinct types of human leukemia and lymphoma and established the range of serum 2-HG in appropriate normal control individuals by using gas chromatograph-time-of-flight mass spectrometry. Aberrant serum 2-HG pattern was detected in the multicenter group of AML, with 62 of 367 (17%) patients having 2-HG levels above the cutoff value (2.01, log2-transformed from 4.03 µg/mL). IDH1/2 mutations occurred in 27 of 31 (87%) AML cases with very high 2-HG, but were observed only in 9 of 31 (29%) patients with moderately high 2-HG, suggesting other genetic or biochemical events may exist in causing 2-HG elevation. Indeed, glutamine-related metabolites exhibited a pattern in favor of 2-HG synthesis in the high 2-HG group. In AML patients with cytogenetically normal AML (n = 234), high 2-HG represented a negative prognostic factor in both overall survival and event-free survival. Univariate and multivariate analyses confirmed high serum 2-HG as a strong prognostic predictor independent of other clinical and molecular features. We also demonstrated distinct gene-expression/DNA methylation profiles in AML blasts with high 2-HG compared with those with normal ones, supporting a role that 2-HG plays in leukemogenesis.


Asunto(s)
Glutaratos/sangre , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/mortalidad , China/epidemiología , Metilación de ADN/genética , Supervivencia sin Enfermedad , Femenino , Regulación Leucémica de la Expresión Génica/genética , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Mutación , Tasa de Supervivencia
16.
J Proteome Res ; 12(10): 4393-401, 2013 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-23998518

RESUMEN

Acute myeloid leukemia (AML) is a life-threatening hematological disease. Novel diagnostic and prognostic markers will be essential for new therapeutics and for significantly improving the disease prognosis. To characterize the metabolic features associated with AML and search for potential diagnostic and prognostic methods, here we analyzed the phenotypic characteristics of serum metabolite composition (metabonome) in a cohort of 183 patients with de novo acute myeloid leukemia together with 232 age- and gender-matched healthy controls using (1)H NMR spectroscopy in conjunction with multivariate data analysis. We observed significant serum metabonomic differences between AML patients and healthy controls and between AML patients with favorable and intermediate cytogenetic risks. Such differences were highlighted by systems differentiations in multiple metabolic pathways including glycolysis/gluconeogenesis, TCA cycle, biosynthesis of proteins and lipoproteins, and metabolism of fatty acids and cell membrane components, especially choline and its phosphorylated derivatives. This demonstrated the NMR-based metabonomics as a rapid and less invasive method for potential AML diagnosis and prognosis. The serum metabolic phenotypes observed here indicated that integration of metabonomics with other techniques will be useful for better understanding the biochemistry of pathogenesis and progression of leukemia.


Asunto(s)
Biomarcadores de Tumor/sangre , Leucemia Mieloide Aguda/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Humanos , Leucemia Mieloide Aguda/diagnóstico , Masculino , Redes y Vías Metabólicas , Metaboloma , Metabolómica , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Adulto Joven
17.
Acta Biochim Biophys Sin (Shanghai) ; 45(1): 27-35, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23212076

RESUMEN

Cancer cells have distinct metabolism that highly depends on glycolysis instead of mitochondrial oxidative phosphorylation alone, known as aerobic glycolysis. Pyruvate kinase (PK), which catalyzes the final step of glycolysis, has emerged as a potential regulator of this metabolic phenotype. Expression of PK type M2 (PKM2) is increased and facilitates lactate production in cancer cells, which determines whether the glucose carbons are degraded to pyruvate and lactate or are channeled into synthetic processes. Modulation of PKM2 catalytic activity also regulates the synthesis of DNA and lipids that are required for cell proliferation. However, the mechanisms by which PKM2 coordinates high-energy requirements with high anabolic activities to support cancer cell proliferation are still not completely understood. This review summarizes the biological characteristics of PKM2 and discusses the dual role in cancer metabolism as well as the potential therapeutic applications. Given its pleiotropic effects on cancer biology, PKM2 represents an attractive target for cancer therapy.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de la Membrana/fisiología , Neoplasias/metabolismo , Hormonas Tiroideas/fisiología , Humanos , Fosforilación , Proteínas de Unión a Hormona Tiroide
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA