Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cell ; 186(13): 2748-2764.e22, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37267948

RESUMEN

Ferroptosis, a cell death process driven by iron-dependent phospholipid peroxidation, has been implicated in various diseases. There are two major surveillance mechanisms to suppress ferroptosis: one mediated by glutathione peroxidase 4 (GPX4) that catalyzes the reduction of phospholipid peroxides and the other mediated by enzymes, such as FSP1, that produce metabolites with free radical-trapping antioxidant activity. In this study, through a whole-genome CRISPR activation screen, followed by mechanistic investigation, we identified phospholipid-modifying enzymes MBOAT1 and MBOAT2 as ferroptosis suppressors. MBOAT1/2 inhibit ferroptosis by remodeling the cellular phospholipid profile, and strikingly, their ferroptosis surveillance function is independent of GPX4 or FSP1. MBOAT1 and MBOAT2 are transcriptionally upregulated by sex hormone receptors, i.e., estrogen receptor (ER) and androgen receptor (AR), respectively. A combination of ER or AR antagonist with ferroptosis induction significantly inhibited the growth of ER+ breast cancer and AR+ prostate cancer, even when tumors were resistant to single-agent hormonal therapies.


Asunto(s)
Ferroptosis , Masculino , Humanos , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Peroxidación de Lípido , Peróxidos , Fosfolípidos
2.
Mol Cell ; 82(12): 2215-2227, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35390277

RESUMEN

Ferroptosis, a newly emerged form of regulated necrotic cell death, has been demonstrated to play an important role in multiple diseases including cancer, neurodegeneration, and ischemic organ injury. Mounting evidence also suggests its potential physiological function in tumor suppression and immunity. The execution of ferroptosis is driven by iron-dependent phospholipid peroxidation. As such, the metabolism of biological lipids regulates ferroptosis via controlling phospholipid peroxidation, as well as various other cellular processes relevant to phospholipid peroxidation. In this review, we provide a comprehensive analysis by focusing on how lipid metabolism impacts the initiation, propagation, and termination of phospholipid peroxidation; how multiple signal transduction pathways communicate with ferroptosis via modulating lipid metabolism; and how such intimate cross talk of ferroptosis with lipid metabolism and related signaling pathways can be exploited for the development of rational therapeutic strategies.


Asunto(s)
Ferroptosis , Ferroptosis/genética , Hierro/metabolismo , Metabolismo de los Lípidos , Peroxidación de Lípido , Fosfolípidos
3.
Nat Immunol ; 18(8): 877-888, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28650480

RESUMEN

The origin and specification of human dendritic cells (DCs) have not been investigated at the clonal level. Through the use of clonal assays, combined with statistical computation, to quantify the yield of granulocytes, monocytes, lymphocytes and three subsets of DCs from single human CD34+ progenitor cells, we found that specification to the DC lineage occurred in parallel with specification of hematopoietic stem cells (HSCs) to the myeloid and lymphoid lineages. This started as a lineage bias defined by specific transcriptional programs that correlated with the combinatorial 'dose' of the transcription factors IRF8 and PU.1, which was transmitted to most progeny cells and was reinforced by upregulation of IRF8 expression driven by the hematopoietic cytokine FLT3L during cell division. We propose a model in which specification to the DC lineage is driven by parallel and inheritable transcriptional programs in HSCs and is reinforced over cell division by recursive interactions between transcriptional programs and extrinsic signals.


Asunto(s)
Linaje de la Célula , Células Dendríticas/citología , Células Madre Hematopoyéticas/citología , Factores Reguladores del Interferón/metabolismo , Leucopoyesis , Células Madre Multipotentes/citología , Animales , Diferenciación Celular , Sangre Fetal , Citometría de Flujo , Humanos , Factores Reguladores del Interferón/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Análisis de Componente Principal , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Regulación hacia Arriba
4.
Gastroenterology ; 167(2): 231-249, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38431204

RESUMEN

Ferroptosis is a form of nonapoptotic cell death that involves iron-dependent phospholipid peroxidation induced by accumulation of reactive oxygen species, and results in plasma membrane damage and the release of damage-associated molecular patterns. Ferroptosis has been implicated in aging and immunity, as well as disease states including intestinal and liver conditions and cancer. To date, several ferroptosis-associated genes and pathways have been implicated in liver disease. Although ferroptotic cell death is associated with dysfunction of the intestinal epithelium, the underlying molecular basis is poorly understood. As the mechanisms regulating ferroptosis become further elucidated, there is clear potential to use ferroptosis to achieve therapeutic benefit.


Asunto(s)
Ferroptosis , Enfermedades Gastrointestinales , Especies Reactivas de Oxígeno , Humanos , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/patología , Enfermedades Gastrointestinales/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Animales , Hierro/metabolismo , Transducción de Señal , Peroxidación de Lípido
5.
PLoS Pathog ; 18(3): e1010390, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35286345

RESUMEN

[This corrects the article DOI: 10.1371/journal.ppat.1004253.].

7.
PLoS Pathog ; 15(2): e1007628, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30811506

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) latently infects host cells and establishes lifelong persistence as an extra-chromosomal episome in the nucleus. To persist in proliferating cells, the viral genome typically replicates once per cell cycle and is distributed into daughter cells. This process involves host machinery utilized by KSHV, however the underlying mechanisms are not fully elucidated. In present study, we found that N-Myc downstream regulated gene 1 (NDRG1), a cellular gene known to be non-detectable in primary B cells and endothelial cells which are the major cell types for KSHV infection in vivo, was highly upregulated by KSHV in these cells. We further demonstrated that the high expression of NDRG1 was regulated by latency-associated nuclear antigen (LANA), the major viral latent protein which tethers the viral genome to host chromosome and plays an essential role in viral genome maintenance. Surprisingly, knockdown of NDRG1 in KSHV latently infected cells resulted in a significant decrease of viral genome copy number in these cells. Interestingly, NDRG1 can directly interact with proliferating cell nuclear antigen (PCNA), a cellular protein which functions as a DNA polymerase clamp during DNA replication. Intriguingly, we found that NDRG1 forms a complex with LANA and PCNA and serves as a scaffold protein bridging these two proteins. We further demonstrated that NDRG1 is critical for mediating LANA to recruit PCNA onto terminal repeat (TR) of KSHV genome, and facilitates viral DNA replication and episome persistence. Taken together, our findings suggest that NDRG1 plays an important role in KSHV viral genome replication, and provide new clues for understanding of KSHV persistence.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Herpesvirus Humano 8/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Adulto , Antígenos Virales/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular , Núcleo Celular/metabolismo , Replicación del ADN , ADN Viral/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Genoma Viral , Células HEK293 , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Proteínas Nucleares/metabolismo , Plásmidos/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virología , Regulación hacia Arriba , Latencia del Virus , Replicación Viral
8.
PLoS Pathog ; 13(9): e1006580, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28957431

RESUMEN

There is increasing consensus that males are more vulnerable than females to infection by several pathogens. However, the underlying mechanism needs further investigation. Here, it was showed that knockdown of androgen receptor (AR) expression or pre-treatment with 5α-dihydrotestosterone, the AR agonist, led to a considerably dysregulated Kaposi's sarcoma-associated herpesvirus (KSHV) infection. In endothelial cells, membrane-localized AR promoted the endocytosis and nuclear trafficking of KSHV. The AR interacted with ephrin receptor A2 (EphA2) and increased its phosphorylation at residue Ser897, which was specifically upregulated upon KSHV infection. This phosphorylation resulted from the AR-mediated recruitment of Src, which resulted in the activation of p90 ribosomal S6 kinase 1 (RSK1), which directly phosphorylates EphA2 at Ser897. Finally, the EphA2-mediated entry of KSHV was abolished in a Ser897Asn EphA2 mutant. Taken together, membrane-localized AR was identified as a KSHV entry factor that cooperatively activates Src/RSK1/EphA2 signaling, which subsequently promotes KSHV infection of both endothelial and epithelial cells.


Asunto(s)
Andrógenos/farmacología , Endocitosis/efectos de los fármacos , Efrina-A2/metabolismo , Infecciones por Herpesviridae/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Sarcoma de Kaposi/metabolismo , Andrógenos/metabolismo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Pinocitosis , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/metabolismo , Sarcoma de Kaposi/tratamiento farmacológico , Proteínas Virales/metabolismo , Internalización del Virus/efectos de los fármacos
9.
J Virol ; 91(16)2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28592529

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is a typical gammaherpesvirus that establishes persistent lifelong infection in host cells. In order to establish successful infection, KSHV has evolved numerous immune evasion strategies to bypass or hijack the host immune system. However, host cells still produce immune cytokines abundantly during primary KSHV infection. Whether the immune effectors produced are able to inhibit viral infection and how KSHV successfully conquers these immune effectors remain largely unknown. The guanylate-binding protein 1 (GBP1) gene is an interferon-stimulated gene and exerts antiviral functions on several RNA viruses; however, its function in DNA virus infection is less well understood. In this study, we found that KSHV infection increases both the transcriptional and protein levels of GBP1 at the early stage of primary infection by activating the NF-κB pathway. The overexpression of GBP1 significantly inhibited KSHV infection, while the knockdown of GBP1 promoted KSHV infection. The GTPase activity and dimerization of GBP1 were demonstrated to be responsible for its anti-KSHV activity. Furthermore, we found that GBP1 inhibited the nuclear delivery of KSHV virions by disrupting the formation of actin filaments. Finally, we demonstrated that replication and transcription activator (RTA) promotes the degradation of GBP1 through a proteasome pathway. Taken together, these results provide a new understanding of the antiviral mechanism of GBP1, which possesses potent anti-KSHV activity, and suggest the critical role of RTA in the evasion of the innate immune response during primary infection by KSHV.IMPORTANCE GBP1 can be induced by various cytokines and exerts antiviral activities against several RNA viruses. Our study demonstrated that GBP1 can exert anti-KSHV function by inhibiting the nuclear delivery of KSHV virions via the disruption of actin filaments. Moreover, we found that KSHV RTA can promote the degradation of GBP1 through a proteasome-mediated pathway. Taken together, our results elucidate a novel mechanism of GBP1 anti-KSHV activity and emphasize the critical role of RTA in KSHV evasion of the host immune system during primary infection.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Unión al GTP/metabolismo , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Proteínas Inmediatas-Precoces/metabolismo , Evasión Inmune , Transactivadores/metabolismo , Virión/metabolismo , Transporte Biológico , Línea Celular , Herpesvirus Humano 8/inmunología , Humanos , Multimerización de Proteína
10.
PLoS Pathog ; 12(10): e1005900, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27760204

RESUMEN

Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) is an oncogenic pathogen that displays latent and lytic life cycles. In KS lesions, infiltrated immune cells, secreted viral and/or cellular cytokines, and hypoxia orchestrate a chronic pro-lytic microenvironment that can promote KSHV reactivation. However, only a small subset of viruses spontaneously undergoes lytic replication in this pro-lytic microenvironment while the majority remains in latency. Here, we show that the expression of the Notch ligand JAG1 is induced by KSHV-encoded replication and transcription activator (RTA) during reactivation. JAG1 up-regulation activates Notch signaling in neighboring cells and prevents viral lytic replication. The suppression of JAG1 and Notch1 with inhibitors or small interfering RNA promotes lytic replication in the presence of RTA induction or under conditions of hypoxia. The underlying mechanism involves the Notch downstream effector hairy and enhancer of split 1 (Hes1), which directly binds lytic gene promoters and attenuates viral lytic gene expression. RTA interacts with lymphoid enhancer-binding factor 1 (LEF1), disrupts LEF1/Groucho/TLE suppressive complexes and releases LEF1 to activate JAG1 expression. Taken together, our results suggest that cells with viral lytic replication can inhibit KSHV reactivation in neighboring cells through an RTA-JAG1-Notch pathway. These data provide insight into the mechanism by which the virus maintains the balance between lytic and latent infection in the pro-lytic tumor microenvironment.


Asunto(s)
Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Proteína Jagged-1/metabolismo , Receptores Notch/metabolismo , Transactivadores/metabolismo , Latencia del Virus/fisiología , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Técnicas de Cocultivo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Sarcoma de Kaposi/virología , Transducción de Señal/fisiología , Transfección , Activación Viral/fisiología
12.
J Virol ; 90(3): 1158-68, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26537687

RESUMEN

UNLABELLED: The oncogenic herpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV) is known to encode four viral interferon regulatory factors (vIRF1 to -4) to subvert the host antiviral immune response, but their detailed DNA-binding profiles as transcription factors in the host remain uncharacterized. Here, we first performed genome-wide vIRF2-binding site mapping in the human genome using chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq). vIRF2 was capable of binding to the promoter regions of 100 putative target genes. Importantly, we confirmed that vIRF2 can specifically interact with the promoters of the genes encoding PIK3C3, HMGCR, and HMGCL, which are associated with autophagosome formation or tumor progression and metastasis, and regulate their transcription in vivo. The crystal structure of the vIRF2 DNA-binding domain (DBD) (referred to here as vIRF2DBD) showed variable loop conformations and positive-charge distributions different from those of vIRF1 and cellular IRFs that are associated with DNA-binding specificities. Structure-based mutagenesis revealed that Arg82 and Arg85 are required for the in vitro DNA-binding activity of vIRF2DBD and can abolish the transcription regulation function of vIRF2 on the promoter reporter activity of PIK3C3, HMGCR, and HMGCL. Collectively, our study provided unique insights into the DNA-binding potency of vIRF2 and suggested that vIRF2 could act as a transcription factor of its target genes in the host antiviral immune response. IMPORTANCE: The oncogenic herpesvirus KSHV is the etiological agent of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. KSHV has developed a unique mechanism to subvert the host antiviral immune responses by encoding four homologues of cellular interferon regulatory factors (vIRF1 to -4). However, none of their DNA-binding profiles in the human genome have been characterized until now, and the structural basis for their diverse DNA-binding properties remain poorly understood. In this study, we performed the first genome-wide vIRF2-binding site mapping in the human genome and found vIRF2 can bind to the promoter regions of 100 target cellular genes. X-ray structure analysis and functional studies provided unique insights into its DNA-binding potency and regulation of target gene expression. Our study suggested that vIRF2 could act as a transcription factor of its target genes and contribute to KSHV infection and pathogenesis through versatile functions.


Asunto(s)
ADN Viral/metabolismo , Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Factores Reguladores del Interferón/metabolismo , Factores de Transcripción/metabolismo , Proteínas Virales/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Análisis Mutacional de ADN , ADN Viral/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Herpesvirus Humano 8/genética , Humanos , Evasión Inmune , Factores Reguladores del Interferón/genética , Modelos Moleculares , Unión Proteica , Conformación Proteica , Factores de Transcripción/química , Factores de Transcripción/genética , Proteínas Virales/genética
13.
J Virol ; 89(1): 415-27, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25320320

RESUMEN

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) is a human gammaherpesvirus with latent and lytic reactivation cycles. The mechanism by which KSHV evades the innate immune system to establish latency has not yet been precisely elucidated. Toll-like receptors (TLRs) are the first line of defense against viral infections. Myeloid differentiation factor 88 (MyD88) is a key adaptor that interacts with all TLRs except TLR3 to produce inflammatory factors and type I interferons (IFNs), which are central components of innate immunity against microbial infection. Here, we found that KSHV replication and transcription activator (RTA), which is an immediate-early master switch protein of viral cycles, downregulates MyD88 expression at the protein level by degrading MyD88 through the ubiquitin (Ub)-proteasome pathway. We identified the interaction between RTA and MyD88 in vitro and in vivo and demonstrated that RTA functions as an E3 ligase to ubiquitinate MyD88. MyD88 also was repressed at the early stage of de novo infection as well as in lytic reactivation. We also found that RTA inhibited lipopolysaccharide (LPS)-triggered activation of the TLR4 pathway by reducing IFN production and NF-κB activity. Finally, we showed that MyD88 promoted the production of IFNs and inhibited KSHV LANA-1 gene transcription. Taken together, our results suggest that KSHV RTA facilitates the virus to evade innate immunity through the degradation of MyD88, which might be critical for viral latency control. IMPORTANCE: MyD88 is an adaptor for all TLRs other than TLR3, and it mediates inflammatory factors and IFN production. Our study demonstrated that the KSHV RTA protein functions as an E3 ligase to degrade MyD88 through the ubiquitin-proteasome pathway and block the transmission of TLRs signals. Moreover, we found that KSHV inhibited MyD88 expression during the early stage of de novo infection as well as in lytic reactivation. These results provide a potential mechanism for the virus to evade innate immunity.


Asunto(s)
Herpesvirus Humano 8/inmunología , Interacciones Huésped-Patógeno , Evasión Inmune , Factor 88 de Diferenciación Mieloide/metabolismo , Transactivadores/metabolismo , Línea Celular , Humanos , Mapeo de Interacción de Proteínas , Proteolisis , Ubiquitina/metabolismo
14.
PLoS Pathog ; 10(7): e1004253, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25010525

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma (KS), a malignancy commonly found in AIDS patients. Whether KS is a true neoplasm or hyperplasia has been a subject of intensive debate until recently when KSHV is unequivocally shown to efficiently infect, immortalize and transform rat primary mesenchymal precursor cells (MM). Moreover, KSHV-transformed MM cells (KMM) efficiently induce tumors with hallmark features of KS when inoculated into nude mice. Here, we showed Smad1 as a novel binding protein of KSHV latency-associated nuclear antigen (LANA). LANA interacted with and sustained BMP-activated p-Smad1 in the nucleus and enhanced its loading on the Id promoters. As a result, Ids were significantly up-regulated in KMM cells and abundantly expressed in human KS lesions. Strikingly, genetic and chemical inhibition of the BMP-Smad1-Id pathway blocked the oncogenic phenotype of KSHV-transformed cells in vitro and in vivo. These findings illustrate a novel mechanism by which a tumor virus hijacks and converts a developmental pathway into an indispensable oncogenic pathway for tumorigenesis. Importantly, our results demonstrate the efficacy of targeting the BMP-Smad1-Id pathway for inhibiting the growth of KSHV-induced tumors, and therefore identify the BMP pathway as a promising therapeutic target for KS.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Transformación Celular Viral , Herpesvirus Humano 8/metabolismo , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Transducción de Señal , Proteína Smad1/metabolismo , Animales , Antígenos Virales , Línea Celular Transformada , Femenino , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Nucleares , Ratas , Sarcoma de Kaposi/metabolismo
15.
J Virol ; 88(13): 7331-44, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24741090

RESUMEN

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) typically displays two different phases in its life cycle, the default latent phase and the lytic phase. There is a short period of lytic gene expression in the early stage of KSHV primary infection. The factors involved in the shutdown process of lytic gene expression are poorly identified. It has been shown that the latency-associated nuclear antigen (LANA) encoded by KSHV plays an important role in the establishment of viral latency. In screening, we identified a host protein, Krüppel-associated box domain-associated protein 1 (KAP1), that bound to LANA. We validated the interaction between LANA and KAP1 in vivo and in vitro, as well as their colocalization in the nucleus. We mapped out that LANA interacted with both the N- and C-terminal domains of KAP1. Based on the interface of LANA-KAP1 interaction determined, we proved that LANA recruited KAP1 to the RTA promoter region of the KSHV genome. We revealed that KAP1 was involved in transcriptional repression by LANA. We found multiple cooccupation sites of LANA and KAP1 on the whole KSHV genome by chromatin immunoprecipitation for sequencing (ChIP-seq) and demonstrated that LANA-recruited KAP1 played a critical role in the shutdown of lytic gene expression during the early stage of KSHV primary infection. Taken together, our data suggest that LANA interacts with KAP1 and represses lytic gene expression to facilitate the establishment of KSHV latency. IMPORTANCE: Our study revealed the mechanism of transcriptional repression by LANA during KSHV primary infection, providing new insights into the process of KSHV latency establishment.


Asunto(s)
Antígenos Virales/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/fisiología , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Sarcoma de Kaposi/virología , Latencia del Virus/fisiología , Antígenos Virales/genética , Western Blotting , Inmunoprecipitación de Cromatina , Técnica del Anticuerpo Fluorescente , Genoma Viral , Humanos , Inmunoprecipitación , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/metabolismo , Transactivadores , Proteína 28 que Contiene Motivos Tripartito , Células Tumorales Cultivadas , Activación Viral/genética
16.
Cell Chem Biol ; 31(2): 187-189, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38364774

RESUMEN

In this issue of Cell Chemical Biology, Rodencal et al.1 report that cell-cycle arrest by p53 stabilizers or CDK4/6 inhibitors (CDK4/6i) can lead to phospholipid remodeling and hence sensitize cancer cells to GPX4 inhibitor (GPX4i)-triggered ferroptosis. This study suggests a novel cancer therapeutic strategy combining CDK4/6i with GPX4i.


Asunto(s)
Ferroptosis , Hierro , Fosfolípidos , Inhibidores de Proteínas Quinasas
17.
Redox Biol ; 75: 103211, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38908072

RESUMEN

Ferroptosis is a pervasive non-apoptotic form of cell death highly relevant in various degenerative diseases and malignancies. The hallmark of ferroptosis is uncontrolled and overwhelming peroxidation of polyunsaturated fatty acids contained in membrane phospholipids, which eventually leads to rupture of the plasma membrane. Ferroptosis is unique in that it is essentially a spontaneous, uncatalyzed chemical process based on perturbed iron and redox homeostasis contributing to the cell death process, but that it is nonetheless modulated by many metabolic nodes that impinge on the cells' susceptibility to ferroptosis. Among the various nodes affecting ferroptosis sensitivity, several have emerged as promising candidates for pharmacological intervention, rendering ferroptosis-related proteins attractive targets for the treatment of numerous currently incurable diseases. Herein, the current members of a Germany-wide research consortium focusing on ferroptosis research, as well as key external experts in ferroptosis who have made seminal contributions to this rapidly growing and exciting field of research, have gathered to provide a comprehensive, state-of-the-art review on ferroptosis. Specific topics include: basic mechanisms, in vivo relevance, specialized methodologies, chemical and pharmacological tools, and the potential contribution of ferroptosis to disease etiopathology and progression. We hope that this article will not only provide established scientists and newcomers to the field with an overview of the multiple facets of ferroptosis, but also encourage additional efforts to characterize further molecular pathways modulating ferroptosis, with the ultimate goal to develop novel pharmacotherapies to tackle the various diseases associated with - or caused by - ferroptosis.

18.
J Virol ; 86(23): 12717-30, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22973026

RESUMEN

It is widely held that any given virus uses only one type of nucleic acid for genetic information storage. However, this consensus has been challenged slightly by several recent studies showing that many RNA species are present within a range of DNA viruses that include Kaposi's sarcoma-associated herpesvirus (KSHV). RNAs extracted from purified DNA virus particles exhibit great diversity in terms of length, abundance, temporal expression, cellular localization, and coding capacity during viral infection. In addition to known RNA species, the current study showed that small regulatory RNAs were present in KSHV virions. A large number of viral and cellular microRNAs (miRNAs), as well as unusual small RNAs (usRNAs), were detected in KSHV virions by using deep sequencing. Both viral and host miRNAs detected in small RNAs extracted from KSHV virions were further shown to colocalize with KSHV virions directly by in situ hybridization (ISH)-electron microscopy (EM) (ISH-EM). Some of these miRNAs were differentially present in the host cells and KSHV virions, suggesting that they are not randomly present in KSHV virions. The virional miRNAs could be transported into host cells, and they are biologically functional during de novo viral infection. Our study revealed miRNAs and usRNAs as a novel group of components in KSHV virions.


Asunto(s)
Herpesvirus Humano 8/genética , MicroARNs/genética , ARN Pequeño no Traducido/genética , Secuencias Reguladoras de Ácido Ribonucleico/genética , Virión/genética , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Cartilla de ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hibridación in Situ , Microscopía Electrónica , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Virol ; 86(3): 1372-81, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22013049

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) encodes 12 pre-microRNAs (pre-miRNAs). Current studies have shown that these miRNAs are involved in regulation of viral and host gene expression, implicating a role in the maintenance of viral latency and suppression of antiviral innate immunity. However, the functions of these miRNAs remain largely unknown. On the basis of the sequence homology between oncogenic miR-155 and KSHV-encoded miR-K12-11, we hypothesized that miR-K12-11 could attenuate transforming growth factor ß (TGF-ß) signaling, facilitating viral infection and tumorigenesis. In the present study, we demonstrated that ectopic expression of miR-K12-11 in Ramos, a TGF-ß-sensitive cell line, downregulated TGF-ß signaling and facilitated cell proliferation upon TGF-ß treatment by directly targeting SMAD5, an important mediator in TGF-ß signaling. In addition, the downregulation of SMAD5 by miR-K12-11 was further confirmed in a de novo KSHV infection system or latently infected KSHV-positive B-lymphoma cell lines. More importantly, repression of miR-K12-11 by a specific sponge inhibitor restored the expression of SMAD5 in both de novo-infected and latently infected cells. Finally, we found that restoration of SMAD5, in addition to the TGF-ß type II receptor, which was epigenetically silenced by the latent viral protein latency-associated nuclear antigen, sensitized BC3 cells to the cytostatic effect of TGF-ß signaling. Taken together, our findings highlight a novel mechanism in which miR-K12-11 downregulates TGF-ß signaling and suggest that viral miRNAs and proteins may exert a dichotomy regulation in virus-induced oncogenesis by targeting the same signaling pathway.


Asunto(s)
Herpesvirus Humano 8/fisiología , MicroARNs/fisiología , Transducción de Señal/fisiología , Proteína Smad5/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Secuencia de Bases , Línea Celular , Cartilla de ADN , Herpesvirus Humano 8/genética , Humanos , Reacción en Cadena de la Polimerasa/métodos , Proteína Smad5/metabolismo
20.
J Virol ; 86(9): 4956-69, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22379075

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8, is closely associated with several malignancies, including Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. KSHV can establish lifelong latency in the host, but the mechanism is not fully understood. Previous studies have proposed a feedback model in which the viral replication and transcription activator (RTA) can induce the expression of the latency-associated nuclear antigen (LANA) during early infection. LANA, in turn, represses transcription and RTA function to establish and maintain KSHV latency. The interaction between LANA and the recombination signal sequence binding protein Jκ (RBP-Jκ, also called CSL), a major transcriptional repressor of the Notch signaling pathway, is essential for RTA repression. In the present study, we show that the LANA carboxyl-terminal amino acids 1052 to 1082 are responsible for the LANA interaction with RBP-Jκ. The secondary structure of the LANA carboxyl terminus resembles the RBP-Jκ-associated module (RAM) of Notch receptor. Furthermore, deletion of the region of LANA residues 1052 to 1082 resulted in aberrant expression of RTA, leading to elevated viral lytic replication. For the first time, we dissected a conserved RBP-Jκ binding domain in LANA and demonstrated that this domain was indispensable for LANA-mediated repression of KSHV lytic genes, thus helping the virus maintain latency and control viral reactivation.


Asunto(s)
Antígenos Virales/química , Antígenos Virales/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Replicación Viral/fisiología , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Antígenos Virales/genética , Línea Celular , Secuencia de Consenso , Genes Virales , Genoma Viral , Humanos , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Unión Proteica , Estructura Secundaria de Proteína , Eliminación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA