Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Clin Immunol ; 44(6): 127, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38773005

RESUMEN

We described the diagnosis and treatment of a patient with autoinflammatory disease, named "Deficiency in ELF4, X-linked (DEX)". A novel ELF4 variant was discovered and its pathogenic mechanism was elucidated. The data about clinical, laboratory and endoscopic features, treatment, and follow-up of a patient with DEX were analyzed. Whole exome sequencing and Sanger sequencing were performed to identify potential pathogenic variants. The mRNA and protein levels of ELF4 were analyzed by qPCR and Western blotting, respectively. The association of ELF4 frameshift variant with nonsense-mediated mRNA decay (NMD) in the pathogenesis DEX was examined. Moreover, RNA-seq was performed to identify the key molecular events triggered by ELF4 variant. The relationship between ELF4 and IFN-ß activity was validated using a dual-luciferase reporter assay and a ChIP-qPCR assay. An 11-year-old boy presented with a Behçet's-like phenotype. The laboratory abnormality was the most obvious in elevated inflammatory indicators. Endoscopy revealed multiple ileocecal ulcers. Intestinal histopathology showed inflammatory cell infiltrations. The patient was treated with long-term immunosuppressant and TNF-α blocker (adalimumab), which reaped an excellent response over 16 months of follow-up. Genetic analysis identified a maternal hemizygote frameshift variant (c.1022del, p.Q341Rfs*30) in ELF4 gene in the proband. The novel variant decreased the mRNA level of ELF4 via the NMD pathway. Mechanistically, insufficient expression of ELF4 disturbed the immune system, leading to immunological disorders and pathogen susceptibility, and disrupted ELF4-activating IFN-ß responses. This analysis detailed the clinical characteristics of a Chinese patient with DEX who harbored a novel ELF4 frameshift variant. For the first time, we used patient-derived cells and carried out transcriptomic analysis to delve into the mechanism of ELF4 variant in DEX.


Asunto(s)
Mutación del Sistema de Lectura , Perfilación de la Expresión Génica , Niño , Humanos , Masculino , Secuenciación del Exoma , Predisposición Genética a la Enfermedad , Degradación de ARNm Mediada por Codón sin Sentido , Linaje , Proteínas Proto-Oncogénicas c-ets/genética , Factores de Transcripción/genética , Transcriptoma
2.
J Clin Immunol ; 44(2): 44, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38231408

RESUMEN

Defining monogenic drivers of autoinflammatory syndromes elucidates mechanisms of disease in patients with these inborn errors of immunity and can facilitate targeted therapeutic interventions. Here, we describe a cohort of patients with a Behçet's- and inflammatory bowel disease (IBD)-like disorder termed "deficiency in ELF4, X-linked" (DEX) affecting males with loss-of-function variants in the ELF4 transcription factor gene located on the X chromosome. An international cohort of fourteen DEX patients was assessed to identify unifying clinical manifestations and diagnostic criteria as well as collate findings informing therapeutic responses. DEX patients exhibit a heterogeneous clinical phenotype including weight loss, oral and gastrointestinal aphthous ulcers, fevers, skin inflammation, gastrointestinal symptoms, arthritis, arthralgia, and myalgia, with findings of increased inflammatory markers, anemia, neutrophilic leukocytosis, thrombocytosis, intermittently low natural killer and class-switched memory B cells, and increased inflammatory cytokines in the serum. Patients have been predominantly treated with anti-inflammatory agents, with the majority of DEX patients treated with biologics targeting TNFα.


Asunto(s)
Artritis , Síndrome de Behçet , Productos Biológicos , Enfermedades Inflamatorias del Intestino , Masculino , Humanos , Síndrome de Behçet/diagnóstico , Síndrome de Behçet/genética , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/genética , Artralgia , Proteínas de Unión al ADN , Factores de Transcripción/genética
3.
Acta Biochim Biophys Sin (Shanghai) ; 56(1): 129-139, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-37674363

RESUMEN

Esophageal squamous cell carcinoma (ESCC) commonly has aggressive properties and a poor prognosis. Investigating the molecular mechanisms underlying the progression of ESCC is crucial for developing effective therapeutic strategies. Here, by performing transcriptome sequencing in ESCC and adjacent normal tissues, we find that E74-like transcription factor 4 (ELF4) is the main upregulated transcription factor in ESCC. The results of the immunohistochemistry show that ELF4 is overexpressed in ESCC tissues and is significantly correlated with cancer staging and prognosis. Furthermore, we demonstrate that ELF4 could promote cancer cell proliferation, migration, invasion, and stemness by in vivo assays. Through RNA-seq and ChIP assays, we find that the stemness-related gene fucosyltransferase 9 ( FUT9) is transcriptionally activated by ELF4. Meanwhile, ELF4 is verified to affect ESCC cancer stemness by regulating FUT9 expression. Overall, we first discover that the transcription factor ELF4 is overexpressed in ESCC and can promote ESCC progression by transcriptionally upregulating the stemness-related gene FUT9.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Humanos , Carcinoma de Células Escamosas de Esófago/metabolismo , Neoplasias Esofágicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo
4.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 756-761, 2024 May 20.
Artículo en Zh | MEDLINE | ID: mdl-38948265

RESUMEN

The patient 1, a 13-year-old boy, was admitted due to "recurrent oral ulcers for 3 years, abdominal pain for 8 months, and perianal ulcers for 10 days"; The patient 2, a 3-year-old boy, was admitted due to "recurrent abdominal pain, diarrhea, and fever for over 3 months". Genetic testing of both patients revealed "deficiency in ELF4, X-linked" (DEX), and the patients were diagnosed with Behcet's disease-like syndrome due to deficiency in ELF4, accordingly. The patient 1 was successively given intravenous methylprednisolone pulses and oral prednisone and mesalazine for symptomatic treatment. The patient 2 was successively treated with corticosteroids combined with enteral nutrition, as well as oral mercaptopurine. Subsequently, both patients showed improvements in symptoms and were discharged.


Asunto(s)
Síndrome de Behçet , Humanos , Masculino , Síndrome de Behçet/genética , Síndrome de Behçet/complicaciones , Preescolar , Adolescente
5.
J Clin Immunol ; 43(5): 933-939, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36823308

RESUMEN

Patients with DEX (deficiency in ELF4, X-linked) were recently reported by our team and others, and cases are very limited worldwide. Our knowledge of this new disease is currently preliminary. In this study, we described 5 more cases presenting mainly with oral ulcer, inflammatory bowel disease-like symptoms, fever of unknown origin, anemia, or systemic lupus erythematosus. Whole exome sequencing identified potential pathogenic ELF4 variants in all cases. The pathogenicity of these variants was confirmed by the detection of ELF4 expression in peripheral blood mononuclear cells from patients and utilizing a simple IFN-b luciferase reporter assay, as previously reported. Our findings significantly contribute to the current understanding of DEX.


Asunto(s)
Enfermedades del Sistema Inmune , Lupus Eritematoso Sistémico , Humanos , Leucocitos Mononucleares , China , Estudios de Cohortes , Proteínas de Unión al ADN , Factores de Transcripción
6.
Cell Biol Toxicol ; 39(2): 519-536, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35674868

RESUMEN

It has been manifested that tumor-derived exosomes (Exos) can deliver long noncoding RNAs to participate in gastric cancer (GC) progression. In this research, we intended to dissect out whether tumor-derived Exos carried LINC01091 to afflict the growth and metastasis of GC. GC tissues and human GC cells were attained for RNA and protein quantification. Accordingly, LINC01091, ELF4, and CDX2 were abundant but microRNA (miR)-128-3p was underexpressed in GC tissues and cells. Exos were isolated from LINC01091-silenced GC cells (Exo-sh-LINC01091). GC cells were co-cultured with Exo-sh-LINC01091 or manipulated with miR mimic, inhibitor, or overexpressing or silencing plasmids. Exo-sh-LINC01091, LINC01091, ELF4 or CDX2 silencing, or miR-128-3p upregulation augmented GC cell proliferative, migrating, and invasive properties. In addition, luciferase, RNA pull-down, and ChIP assays offered evidence supporting the mechanism that LINC01091 bound to miR-128-3p that inversely targeted ELF4, and ELF4 transcriptionally activated CDX2 by binding to its promoter in GC cells. Moreover, Exo-sh-LINC01091 modulated the miR-128-3p/ELF4/CDX2 axis and restrained the tumorigenesis and metastasis in vivo. Conclusively, LINC01091 shuttled by tumor-derived Exos might expedite GC development by activating the ELF4/CDX2 axis via miR-128-3p downregulation.


Asunto(s)
Exosomas , MicroARNs , Neoplasias Gástricas , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Gástricas/metabolismo , Exosomas/genética , Exosomas/metabolismo , Proliferación Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Transcripción CDX2/genética , Factor de Transcripción CDX2/metabolismo
7.
J Clin Immunol ; 42(4): 798-810, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35266071

RESUMEN

Monogenic autoinflammatory diseases (mAIDs) are a heterogeneous group of diseases affecting primarily innate immunity, with various genetic causes. Genetic diagnosis of mAIDs can assist in the patient's management and therapy. However, a large number of sporadic and familial cases remain genetically uncharacterized. Deficiency in ELF4, X-linked (DEX) is recently identified as a novel mAID. Here, we described a pediatric patient suffering from recurrent viral and bacterial respiratory infection, refractory oral ulcer, constipation, and arthritis. Whole-exome sequencing found a hemizygous variant in ELF4 (chrX:129205133 A > G, c.691 T > C, p.W231R). Using cells from patient and point mutation mice, we showed mutant cells failed to restrict viral replication effectively and produced more pro-inflammatory cytokines. RNA-seq identified several potential critical antiviral and anti-inflammation genes with decreased expression, and ChIP-qPCR assay suggested mutant ELF4 failed to bind to the promoters of these genes. Thus, we presented the second report of DEX.


Asunto(s)
Enfermedades Autoinflamatorias Hereditarias , Síndromes de Inmunodeficiencia , Síndrome de Inmunodeficiencia Adquirida del Murino , Animales , Niño , Proteínas de Unión al ADN/genética , Enfermedades Autoinflamatorias Hereditarias/diagnóstico , Enfermedades Autoinflamatorias Hereditarias/genética , Humanos , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/genética , Mutación con Pérdida de Función , Ratones , Mutación/genética , Factores de Transcripción/genética , Secuenciación del Exoma
8.
Int J Mol Sci ; 23(9)2022 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-35563234

RESUMEN

Alcohol liver disease (ALD) is characterized by intestinal barrier disruption and gut dysbiosis. Dysfunction of E74-like ETS transcription factor 4 (ELF4) leads to colitis. We aimed to test the hypothesis that intestinal ELF4 plays a critical role in maintaining the normal function of intestinal barrier and gut homeostasis in a mouse model of ALD. Intestinal ELF4 deficiency resulted in dysfunction of the intestinal barrier. Elf4-/- mice exhibited gut microbiota (GM) dysbiosis with the characteristic of a larger proportion of Proteobacteria. The LPS increased in Elf4-/- mice and was the most important differential metabolite between Elf4-/- mice and WT mice. Alcohol exposure increased liver-to-body weight ratio, and hepatic inflammation response and steatosis in WT mice. These deleterious effects were exaggerated in Elf4-/- mice. Alcohol exposure significantly increased serum levels of TG, ALT, and AST in Elf4-/- mice but not in WT mice. In addition, alcohol exposure resulted in enriched expression of genes associated with cholesterol metabolism and lipid metabolism in livers from Elf4-/- mice. 16S rRNA sequencing showed a decrease abundance of Akkermansia and Bilophila in Elf4-/- mice. In conclusion, intestinal ELF4 is an important host protective factor in maintaining gut homeostasis and alleviating alcohol exposure-induced hepatic steatosis and injury.


Asunto(s)
Hígado Graso Alcohólico , Hepatopatías Alcohólicas , Animales , Disbiosis/metabolismo , Etanol/metabolismo , Etanol/toxicidad , Hígado Graso Alcohólico/metabolismo , Homeostasis , Hígado/metabolismo , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética
9.
J Integr Plant Biol ; 64(7): 1310-1324, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35603836

RESUMEN

The circadian clock maintains the daily rhythms of plant growth and anticipates predictable ambient temperature cycles. The evening complex (EC), comprising EARLY FLOWERING 3 (ELF3), ELF4, and LUX ARRHYTHMO, plays an essential role in suppressing thermoresponsive hypocotyl growth by negatively regulating PHYTOCHROME INTERACTING FACTOR 4 (PIF4) activity and its downstream targets in Arabidopsis thaliana. However, how EC activity is attenuated by warm temperatures remains unclear. Here, we demonstrate that warm temperature-induced REVEILLE 7 (RVE7) fine-tunes thermoresponsive growth in Arabidopsis by repressing ELF4 expression. RVE7 transcript and RVE7 protein levels increased in response to warm temperatures. Under warm temperature conditions, an rve7 loss-of-function mutant had shorter hypocotyls, while overexpressing RVE7 promoted hypocotyl elongation. PIF4 accumulation and downstream transcriptional effects were reduced in the rve7 mutant but enhanced in RVE7 overexpression plants under warm conditions. RVE7 associates with the Evening Element in the ELF4 promoter and directly represses its transcription. ELF4 is epistatic to RVE7, and overexpressing ELF4 suppressed the phenotype of the RVE7 overexpression line under warm temperature conditions. Together, our results identify RVE7 as an important regulator of thermoresponsive growth that functions (in part) by controlling ELF4 transcription, highlighting the importance of ELF4 for thermomorphogenesis in plants.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Relojes Circadianos , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Relojes Circadianos/genética , Ritmo Circadiano/genética , Regulación de la Expresión Génica de las Plantas/genética , Hipocótilo/metabolismo , Temperatura , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Biochem Biophys Res Commun ; 554: 41-48, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33774278

RESUMEN

Fbxo7 is a key player in the differentiation and function of numerous blood cell types, and in neurons, oligodendrocytes and spermatocytes. In an effort to gain insight into the physiological and pathological settings where Fbxo7 is likely to play a key role, we sought to define the transcription factors which direct FBXO7 expression. Using sequence alignments across 28 species, we defined the human FBXO7 promoter and found that it contains two conserved regions enriched for multiple transcription factor binding sites. Many of these have roles in either neuronal or haematopoietic development. Using various FBXO7 promoter reporters, we found ELF4, Pax5 and c-Myb have functional binding sites that activate transcription. We find endogenous Pax5 is bound to the FBXO7 promoter in pre-B cells, and that the exogenous expression of Pax5 represses Fbxo7 transcription in early pro-B cells.


Asunto(s)
Linfocitos B/metabolismo , Proteínas F-Box/genética , Factor de Transcripción PAX5/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Linfocitos B/citología , Secuencia de Bases , Sitios de Unión , Diferenciación Celular/fisiología , Células Cultivadas , Secuencia Conservada , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas F-Box/metabolismo , Humanos , Factor de Transcripción PAX5/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myb/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
J Biol Chem ; 294(19): 7787-7796, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30898878

RESUMEN

Platelet factor 4 (PF4) is an anti-Plasmodium component of platelets. It is expressed in megakaryocytes and released from platelets following infection with Plasmodium Innate immunity is crucial for the host anti-Plasmodium response, in which type I interferon plays an important role. Whether there is cross-talk between innate immune signaling and the production of anti-Plasmodium defense peptides is unknown. Here we demonstrate that E74, like ETS transcription factor 4 (ELF4), a type I interferon activator, can help protect the host from Plasmodium yoelii infection. Mechanically, ELF4 binds to the promoter of genes of two C-X-C chemokines, Pf4 and pro-platelet basic protein (Ppbp), initiating the transcription of these two genes, thereby enhancing PF4-mediated killing of parasites from infected erythrocytes. Elf4-/- mice are much more susceptible to Plasmodium infection than WT littermates. The expression level of Pf4 and Ppbp in megakaryocytes from Elf4-/- mice is much lower than in those from control animals, resulting in increased parasitemia. In conclusion, our study uncovered a distinct role of ELF4, an innate immune molecule, in host defense against malaria.


Asunto(s)
Quimiocinas CXC/inmunología , Proteínas de Unión al ADN/inmunología , Malaria/inmunología , Plasmodium yoelii/inmunología , Factor Plaquetario 4/inmunología , Factores de Transcripción/inmunología , Transcripción Genética/inmunología , Animales , Quimiocinas CXC/genética , Proteínas de Unión al ADN/genética , Malaria/genética , Malaria/patología , Ratones , Ratones Noqueados , Factor Plaquetario 4/genética , Factores de Transcripción/genética , Transcripción Genética/genética
12.
RNA ; 22(9): 1349-59, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27365210

RESUMEN

Meiosis is a highly regulated process by which genetic information is transmitted through sexual reproduction. It encompasses unique mechanisms that do not occur in vegetative cells, producing a distinct, well-regulated meiotic transcriptome. During vegetative growth, many meiotic genes are constitutively transcribed, but most of the resulting mRNAs are rapidly eliminated by the Mmi1-MTREC (Mtl1-Red1 core) complex. While Mmi1-MTREC targets premature meiotic RNAs for degradation by the nuclear 3'-5' exoribonuclease exosome during mitotic growth, its role in meiotic gene expression during meiosis is not known. Here, we report that Red5, an essential MTREC component, interacts with pFal1, an ortholog of eukaryotic translation initiation factor eIF4aIII in the fission yeast Schizosaccharomyces pombe In mammals, together with MAGO (Mnh1), Rnps1, and Y14, elF4AIII (pFal1) forms the core of the exon junction complex (EJC), which is essential for transcriptional surveillance and localization of mature mRNAs. In fission yeast, two EJC orthologs, pFal1 and Mnh1, are functionally connected with MTREC, specifically in the process of meiotic gene expression during meiosis. Although pFal1 interacts with Mnh1, Y14, and Rnps1, its association with Mnh1 is not disrupted upon loss of Y14 or Rnps1. Mutations of Red1, Red5, pFal1, or Mnh1 produce severe meiotic defects; the abundance of meiotic transcripts during meiosis decreases; and mRNA maturation processes such as splicing are impaired. Since studying meiosis in mammalian germline cells is difficult, our findings in fission yeast may help to define the general mechanisms involved in accurate meiotic gene expression in higher eukaryotes.


Asunto(s)
Exosomas/metabolismo , Meiosis , Empalme del ARN , ARN Mensajero/genética , Schizosaccharomyces/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Exones , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Exosomas/genética , Intrones , ARN Mensajero/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Factores de Escisión y Poliadenilación de ARNm/genética , Factores de Escisión y Poliadenilación de ARNm/metabolismo
13.
J Biol Chem ; 289(38): 26143-26154, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25081543

RESUMEN

Myeloid Elf-1-like factor (MEF) or Elf4 is an ETS transcription factor that activates innate immunity-associated genes such as lysozyme (LYZ), human ß-defensin 2 (HßD2), and interleukin-8 (IL-8) in epithelial cells and is also known to influence cell cycle progression. MEF is transcriptionally activated by E2F1, but the E2F1-mediated transcriptional activation is inhibited by p53 through E2F1-p53 protein interaction. Although the transcriptional activation of MEF has been investigated in depth, its post-translational regulation is not well explored. By overexpressing MEF cDNA in human cell lines, here we show that MEF protein expression is suppressed by p53. By screening a number of E3 ligases regulated by p53, we found that MDM2 is involved in the effect of p53 on MEF. MDM2 is transcriptionally activated by p53 and interacts with MEF protein to enhance MEF degradation. MDM2 reduces MEF protein expression, as well as stability and function of MEF as transcriptional activator. Furthermore, MDM2 was able to down-regulate MEF in the absence of p53, indicating a p53-independent effect on MEF. Notably, MEF transcriptionally activates MDM2, which was previously demonstrated to be the mechanism by which MEF suppresses the p53 protein. These results reveal that in addition to the potential of MEF to down-regulate p53 by transcriptionally activating E3 ligase MDM2, MEF participates with MDM2 in a novel autoregulatory feedback loop to regulate itself. Taken together with the findings on the effect of p53 on MEF, these data provide evidence that the p53-MDM2-MEF axis is a feedback mechanism that exquisitely controls the balance of these transcriptional regulators.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Núcleo Celular/metabolismo , Regulación hacia Abajo , Retroalimentación Fisiológica , Expresión Génica , Células HCT116 , Humanos , Estabilidad Proteica , Proteolisis , Activación Transcripcional , Ubiquitinación
14.
Eur J Immunol ; 44(3): 715-27, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24338897

RESUMEN

Most differentiated CD8(+) T cells die off at the end of an infection, revealing two main subsets of memory T cells - central and effector memory - which can be found in lymphoid tissues or circulating through nonlymphoid organs, respectively. The cell intrinsic regulation of the differentiation of CD8(+) T cells to effector and central memory remains poorly studied. Herein, we describe a novel role of the ETS transcription factor ELF4 in the development and function of memory CD8(+) T cells following infection with Listeria monocytogenes. Adoptively transferred Elf4(-/-) naïve CD8(+) T cells produced lower numbers of effector memory CD8(+) T cells despite a normal pool of central memory. This was caused by suboptimal priming and decreased survival of CD8(+) T cells at the peak of response while enhanced Notch1 signaling and upregulation of eomesodermin correlated with "normal" development of Elf4(-/-) central memory. Finally, loss of ELF4 impaired the expansion of both central and effector memory CD8(+) T cells in a recall response by also activating Notch1 signaling. Altogether, ELF4 emerges as a novel transcriptional regulator of CD8(+) T-cell differentiation in response to infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas de Unión al ADN/genética , Memoria Inmunológica/genética , Listeria monocytogenes/inmunología , Listeriosis/genética , Listeriosis/inmunología , Factores de Transcripción/genética , Animales , Supervivencia Celular/inmunología , Proteínas de Unión al ADN/deficiencia , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Listeriosis/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Ovalbúmina/inmunología , Receptor Notch1/metabolismo , Transducción de Señal , Factores de Transcripción/deficiencia
15.
Plant Cell Rep ; 34(12): 2027-41, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26205509

RESUMEN

KEY MESSAGE: DhEFL2, 3 and 4 regulate the flowering of Doritaenopsis . These genes could rescue elf4-1 phenotype in Arabidopsis while its overexpression delayed flowering. Phalaenopsis are popular floral plants, and studies on orchid flowering genes could help develop off-season cultivars. Early flowering 4 (ELF4) of A. thaliana has been shown to be involved in photoperiod perception and circadian regulation. We isolated two members of the ELF4 family from Doritaenopsis hybrid (Doritaenopsis 'Tinny Tender' (Doritaenopsis Happy Smile × Happy Valentine)), namely, DhEFL2 and DhEFL3 (DhEFL4 has been previously cloned). Multiple alignment analysis of the deduced amino acid sequences of the three DhEFL homologs showed that DhEFL4 and DhEFL2 are similar with 72% identical amino acids, whereas DhEFL3 is divergent with 72% similarity with DhEFL2 and 68% similarity with DhEFL4. DhEFL3 forms a separate phylogenetic subgroup and is far away from DhEFL2 and DhEFL4. The diurnal expression patterns of DhEFL2, 3, and 4 are similar in the long-day photoperiod conditions; however, in the short-day conditions, DhEFL3 is different from DhEFL2 and 4. For the DhEFL2, 3, and 4 genes, the strongest audience expression organs are the stem, petal and bud, respectively. The ectopic expression of DhEFL2, 3, or 4 in transgenic A. thaliana plants (Ws-2 ecotype) showed novel phenotypes by late flowering and more rosette leaves. The ectopic expression of DhEFL2, 3, or 4 could complement the elf4-1 flowering time and hypocotyl length defects in transgenic A. thaliana elf4-1 mutant plants. These results strongly suggest that DhEFL2, 3, and 4 may be involved in regulation of flower formation and floral induction in Doritaenopsis.


Asunto(s)
Regulación de la Expresión Génica de las Plantas , Orchidaceae/genética , Proteínas de Plantas/metabolismo , Secuencia de Aminoácidos , Arabidopsis/genética , Arabidopsis/fisiología , Arabidopsis/efectos de la radiación , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Quimera , Flores/genética , Flores/fisiología , Flores/efectos de la radiación , Hipocótilo/genética , Hipocótilo/fisiología , Hipocótilo/efectos de la radiación , Datos de Secuencia Molecular , Mutación , Orchidaceae/fisiología , Orchidaceae/efectos de la radiación , Fenotipo , Fotoperiodo , Filogenia , Hojas de la Planta/genética , Hojas de la Planta/fisiología , Hojas de la Planta/efectos de la radiación , Proteínas de Plantas/genética , Plantas Modificadas Genéticamente , Plantones/genética , Plantones/fisiología , Plantones/efectos de la radiación , Alineación de Secuencia , Análisis de Secuencia de ADN , Factores de Tiempo
16.
World J Pediatr ; 20(5): 444-450, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38733460

RESUMEN

BACKGROUND: ELF4 deficiency has been recently recognized as a novel disorder within the spectrum of inborn errors of immunity (IEIs), specifically categorized as a "disease of immune dysregulation." Cases of this condition, reported by our team and others, are very limited worldwide. As such, our current knowledge of this new disease remains preliminary. This review aims to provide a brief overview of the clinical manifestations, pathogenesis, and treatment strategies for this novel IEI. DATA SOURCES: A comprehensive review was conducted after an extensive literature search in the PubMed/Medline database and websites concerning transcriptional factor ELF4 and reports concerning patients with ELF4 deficiency. Our search strategy was "ELF4 OR ETS-related transcription factor Elf-4 OR EL4-like factor 4 OR myeloid Elf-1-like factor" as of the time of manuscript submission. RESULTS: The current signature manifestations of ELF4 deficiency disorder are recurrent and prolonged oral ulcer, abdominal pain, and diarrhea in pediatric males. In some cases, immunodeficiency and autoimmunity can also be prominent. Targeted Sanger sequencing or whole exome sequencing can be used to detect variation in ELF4 gene. Western blotting for ELF4 expression of the patient's cells can confirm the pathogenic effect of the variant. To fully confirm the pathogenicity of the variant, further functional test is strongly advised. Glucocorticoid and biologics are the mainstream management of ELF4 deficiency disorder. CONCLUSIONS: Pediatric males presenting with recurring ulcerations in digestive tract epithelium with or without recurrent fever should be suspected of DEX. When atypical presentations are prominent, variations in ELF4 gene should be carefully evaluated functionally due to the complex nature of ELF4 function. Experience of treating DEX includes use of glucocorticoid and biologics and more precise treatment needs more patients to identify and further mechanistic study.


Asunto(s)
Proteínas de Unión al ADN , Factores de Transcripción , Humanos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas Proto-Oncogénicas c-ets/genética , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Enfermedades del Sistema Inmune/genética
17.
BMC Mol Cell Biol ; 24(1): 22, 2023 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-37474923

RESUMEN

BACKGROUND: Renal ischemia/reperfusion (I/R) injury is a major cause of acute kidney injury (AKI). Dysfunction of E74-like ETS transcription factor 4 (ELF4) leads to inflammation. This research intended to look into the function and mechanisms of ELF4 in I/R and oxygen-glucose deprivation/reperfusion (OGD/R) model. RESULTS: In I/R and OGD/R model, ELF4 expression was downregulated. ELF4 knockout aggravated I/R-induced kidney injury, oxidative stress (OS), endoplasmic reticulum stress (ERS), apoptosis, inflammation, and pyroptosis in mice. In HK-2 cells treated with OGD/R, suppression of ELF4 expression inhibited cell proliferation and promoted cell apoptosis, OS, ERS, inflammation, and pyroptosis. Moreover, ELF4 overexpression led to the opposite results. CONCLUSION: ELF4 deficiency aggravated I/R induced AKI, which was involved in apoptosis, OS, ERS, inflammation, and pyroptosis. Targeting ELF4 may be a promising new therapeutic strategy for preventing inflammation after IR-AKI.


Asunto(s)
Lesión Renal Aguda , Daño por Reperfusión , Ratones , Animales , Piroptosis , Riñón , Estrés Oxidativo , Daño por Reperfusión/genética , Daño por Reperfusión/tratamiento farmacológico , Isquemia/complicaciones , Isquemia/metabolismo , Lesión Renal Aguda/genética , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/metabolismo , Reperfusión/efectos adversos , Estrés del Retículo Endoplásmico
18.
Neuro Oncol ; 25(3): 459-470, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-35862252

RESUMEN

BACKGROUND: The loss of neurogenic tumor suppressor microRNAs miR-124, miR-128, and miR-137 is associated with glioblastoma's undifferentiated state. Most of their impact comes via the repression of a network of oncogenic transcription factors. We conducted a high-throughput functional siRNA screen in glioblastoma cells and identify E74 like ETS transcription factor 4 (ELF4) as the leading contributor to oncogenic phenotypes. METHODS: In vitro and in vivo assays were used to assess ELF4 impact on cancer phenotypes. We characterized ELF4's mechanism of action via genomic and lipidomic analyses. A MAPK reporter assay verified ELF4's impact on MAPK signaling, and qRT-PCR and western blotting were used to corroborate ELF4 regulatory role on most relevant target genes. RESULTS: ELF4 knockdown resulted in significant proliferation delay and apoptosis in GBM cells and long-term growth delay and morphological changes in glioma stem cells (GSCs). Transcriptomic analyses revealed that ELF4 controls two interlinked pathways: 1) Receptor tyrosine kinase signaling and 2) Lipid dynamics. ELF4 modulation directly affected receptor tyrosine kinase (RTK) signaling, as mitogen-activated protein kinase (MAPK) activity was dependent upon ELF4 levels. Furthermore, shotgun lipidomics revealed that ELF4 depletion disrupted several phospholipid classes, highlighting ELF4's importance in lipid homeostasis. CONCLUSIONS: We found that ELF4 is critical for the GBM cell identity by controlling genes of two dependent pathways: RTK signaling (SRC, PTK2B, and TNK2) and lipid dynamics (LRP1, APOE, ABCA7, PLA2G6, and PITPNM2). Our data suggest that targeting these two pathways simultaneously may be therapeutically beneficial to GBM patients.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , MicroARNs , Humanos , Factores de Transcripción/genética , Glioblastoma/patología , MicroARNs/genética , Proteínas Tirosina Quinasas Receptoras/genética , Regulación Neoplásica de la Expresión Génica , Lípidos , Proliferación Celular , Línea Celular Tumoral , Neoplasias Encefálicas/patología , Proteínas de Unión al ADN/genética , Proteínas Tirosina Quinasas/metabolismo
19.
Front Immunol ; 14: 1270411, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38022496

RESUMEN

Background: Inflammatory bowel disease (IBD) is a chronic immune-mediated disorder affecting millions worldwide. Due to the complexity of its pathogenesis, the treatment options for IBD are limited. This study focuses on ELF4, a member of the ETS transcription factor family, as a target to elucidate its role in IBD and investigate its mechanism of action in alleviating IBD symptoms by activating IL1RN transcription to suppress the activity of inflammatory TH17 cells. Methods: Using the GEO database, this study examined LPS-induced intestinal inflammatory genes and their regulation mechanisms. We examined the colon length of LPS-treated mice and derived the Disease Activity Index (DAI). H&E staining, ELISA, and flow cytometry were used to detect mice colon tissue damage, inflammatory factor levels in mouse serum, mouse macrophage types and inflammatory TH17 cell activity. RT-qPCR and Western blot detected ELF4, IL1RN, M1, and M2 polarization markers. In Vitro, using dual-luciferase and ChIP assays, we tested mouse bone marrow-derived macrophages (BMDMs) and mouse intestinal epithelial cells for IL1RN promoter activity and ELF4 enrichment. Results: Bioinformatics showed that LPS-induced colitis animals have reduced ELF4 expression in their colon tissue. In vivo tests confirmed reduced ELF4 expression in mice with LPS-induced colitis. ELF4 overexpression reduced mouse intestinal inflammation. ELF4 activated IL1RN transcription in bioinformatics and in vitro tests. ELF4 promoted IL1RN transcription and macrophage M2 polarization to limit intestinal epithelial cell death and inflammation and reduce mouse intestinal inflammation in vitro. ELF4 also reduced the Th17/Treg ratio by increasing IL1RN transcription. Conclusion: ELF4 activates IL1RN transcription, suppresses inflammatory TH17 cells, and induces macrophage M2 polarization to treat IBD.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Animales , Ratones , Diferenciación Celular/genética , Colitis/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Lipopolisacáridos/efectos adversos , Macrófagos/metabolismo , Células Th17 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Oncol Lett ; 26(3): 395, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37600328

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most prominent subtype of renal cancer and E47-like factors (ELFs) are important in tumorigenesis; however, the specific role of key ELFs in ccRCC remains unclear. The present study comprehensively analyzed RNA sequencing and clinical data from multiple databases, and identified differentially expressed ELFs (ELF3-5) in ccRCC. The DNA promoter methylation, genetic variation and clinical significance of ELF3-5 in ccRCC were analyzed using the cBioPortal and UALCAN databases. The association between ELF3-5 and multiple immune cell infiltration was analyzed using Tumor Immune Estimation Resource. Subsequently, ELF4 was selected and its association with biological functions was assessed. Cell counting kit-8 (CCK-8), colony formation, Transwell, macrophage chemotaxis and polarization assays were conducted to validate the functions of ELF4. Notably, the mRNA expression levels of ELF4 were significantly upregulated in ccRCC, whereas ELF3 and ELF5 mRNA expression levels were significantly downregulated. Clinical significance analysis revealed that ELF4 showed a high clinical significance with tumor grade, clear cell type A and B subtypes, and incidence rates of amplification in genetic variation. Further analyses indicated that ELF4 may be involved in multiple immune cell differentiation. Additionally, cell experiments revealed that ELF4 inhibition downregulated 769-P and 786-O proliferation, migration and invasion. Knockdown of ELF4 in cancer cells also inhibited M2 macrophage polarization and chemotaxis towards 769-P and 786-O cells. Conclusively, the present findings indicated the clinical significance of ELF4 in ccRCC, and verified its key role in driving cell proliferation, migration and invasion, and promoting M2 macrophage polarization and chemotaxis in ccRCC.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda