Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 718
Filtrar
1.
Mol Hum Reprod ; 30(9)2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39178021

RESUMEN

The subcortical maternal complex (SCMC), which is vital in oocyte maturation and embryogenesis, consists of core proteins (NLRP5, TLE6, OOEP), non-core proteins (PADI6, KHDC3L, NLRP2, NLRP7), and other unknown proteins that are encoded by maternal effect genes. Some variants of SCMC genes have been linked to female infertility characterized by embryonic development arrest. However, so far, the candidate non-core SCMC components associated with embryonic development need further exploration and the pathogenic variants that have been identified are still limited. In this study, we discovered two novel variants [p.(Ala131Val) and p.(Met326Val)] of NLRP2 in patients with primary infertility displaying embryonic development arrest from large families. In vitro studies using 293T cells and mouse oocytes, respectively, showed that these variants significantly decreased protein expression and caused the phenotype of embryonic development arrest. Additionally, we combined the 'DevOmics' database with the whole exome sequence data of our cohort and screened out a new candidate non-core SCMC gene ZFP36L2. Its variants [p.(Ala241Pro) and p.(Pro291dup)] were found to be responsible for embryonic development arrest. Co-immunoprecipitation experiments in 293T cells, used to demonstrate the interaction between proteins, verified that ZFP36L2 is one of the human SCMC components, and microinjection of ZFP36L2 complementary RNA variants into mouse oocytes affected embryonic development. Furthermore, the ZFP36L2 variants were associated with disrupted stability of its target mRNAs, which resulted in aberrant H3K4me3 and H3K9me3 levels. These disruptions decreased oocyte quality and further developmental potential. Overall, this is the first report of ZFP36L2 as a non-core component of the human SCMC and we found four novel pathogenic variants in the NLRP2 and ZFP36L2 genes in 4 of 161 patients that caused human embryonic development arrest. These findings contribute to the genetic diagnosis of female infertility and provide new insights into the physiological function of SCMC in female reproduction.


Asunto(s)
Desarrollo Embrionario , Infertilidad Femenina , Humanos , Femenino , Animales , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Desarrollo Embrionario/genética , Ratones , Oocitos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Adulto , Células HEK293 , Tristetraprolina/genética , Tristetraprolina/metabolismo
2.
Cell Death Dis ; 15(8): 574, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39117638

RESUMEN

Necrosome activation following TLR- or cytokine receptor-signaling results in cell death by necroptosis which is characterized by the rupture of cell membranes and the consequent release of intracellular contents to the extracellular milieu. While necroptosis exacerbates various inflammatory diseases, the mechanisms through which the inflammatory responses are regulated are not clear. We show that the necrosome activation of macrophages results in an upregulation of various pathways, including the mitogen-activated protein kinase (MAPK) cascade, which results in an elevation of the inflammatory response and consequent expression of several cytokines and chemokines. Programming for this upregulation of inflammatory response occurs during the early phase of necrosome activation and proceeds independently of cell death but depends on the activation of the receptor-interacting protein kinase-1 (RipK1). Interestingly, necrosome activation also results in an upregulation of IFNß, which in turn exerts an inhibitory effect on the maintenance of inflammatory response through the repression of MAPK-signaling and an upregulation of Zfp36. Activation of the interferon-induced gene factor-3 (ISGF3) results in the expression of ZFP36 (TTP), which induces the post-transcriptional degradation of mRNAs of various inflammatory cytokines and chemokines through the recognition of AU-rich elements in their 3'UTR. Furthermore, ZFP-36 inhibits IFNß-, but not TNFα- induced necroptosis. Overall, these results reveal the molecular mechanism through which IFNß, a pro-inflammatory cytokine, induces the expression of ZFP-36, which in turn inhibits necroptosis and halts the maintenance of the inflammatory response.


Asunto(s)
Citocinas , Péptidos y Proteínas de Señalización Intracelular , Necroptosis , Proteínas Serina-Treonina Quinasas , Tristetraprolina , Tristetraprolina/metabolismo , Tristetraprolina/genética , Animales , Citocinas/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Macrófagos/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Células RAW 264.7 , Regulación hacia Arriba/efectos de los fármacos , Interferón beta/metabolismo , Ratones Endogámicos C57BL , Regulación de la Expresión Génica
3.
Cell Immunol ; 403-404: 104859, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39067169

RESUMEN

Asthma is a chronic inflammatory airway disease, in which inflammatory cytokines play a pivotal role. The zinc finger binding protein 36 (ZFP36) family includes ZFP36, ZFP36L1, and ZFP36L2 and is among the RNA-binding proteins (RBPs) reported to cause inflammation. The present study aimed to clarify the roles of the ZFP36 family in asthma, particularly highlighting the relationship between the ZFP36 family and Th2 cells, which are key players in type 2 inflammation in asthma. Real-time PCR analysis revealed the preferential expression of ZFP36 family mRNAs in human white blood cells. Gene expression analysis using public datasets from the GEO database (https://www.ncbi.nlm.nih.gov/gds) showed significantly suppressed expression of ZFP36 family mRNAs in patients with asthma compared to that in healthy controls. Using multiple cytokine assays, Th2 cell transfection with ZFP36 family siRNAs enhanced the expression of inflammatory cytokines IL-8, IFN-γ, CCL3/MIP-1α, CCL4/MIP-1ß, and TNF-α and cell surface molecules CCR4 (CD194) and PSGL-1 (CD162). Treatment with IL-2, 4, and 15 significantly suppressed, and corticosteroid significantly enhanced the expressions of ZFP36 family mRNAs by Th2 cells. In conclusion, the ZFP36 family expressed by Th2 cells was suppressed in patients with asthma, leading to the enhanced expression of cytokines and cell surface molecules. Suppressed ZFP36 expression in asthma may be involved in the enhancement of airway inflammation, and the ZFP36 family may be a therapeutic target for inflammatory diseases, including asthma.


Asunto(s)
Asma , Citocinas , Células Th2 , Tristetraprolina , Humanos , Asma/inmunología , Asma/metabolismo , Tristetraprolina/metabolismo , Tristetraprolina/genética , Citocinas/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Inflamación/inmunología , Femenino , Masculino , Adulto , Regulación de la Expresión Génica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción , Factor 1 de Respuesta al Butirato
4.
Eur J Immunol ; 54(10): e2451018, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38980256

RESUMEN

CD8+ T cells kill target cells by releasing cytotoxic molecules and proinflammatory cytokines, such as TNF and IFN-γ. The magnitude and duration of cytokine production are defined by posttranscriptional regulation, and critical regulator herein are RNA-binding proteins (RBPs). Although the functional importance of RBPs in regulating cytokine production is established, the kinetics and mode of action through which RBPs control cytokine production are not well understood. Previously, we showed that the RBP ZFP36L2 blocks the translation of preformed cytokine encoding mRNA in quiescent memory T cells. Here, we uncover that ZFP36L2 regulates cytokine production in a time-dependent manner. T cell-specific deletion of ZFP36L2 (CD4-cre) had no effect on T-cell development or cytokine production during early time points (2-6 h) of T-cell activation. In contrast, ZFP36L2 specifically dampened the production of IFN-γ during prolonged T-cell activation (20-48 h). ZFP36L2 deficiency also resulted in increased production of IFN-γ production in tumor-infiltrating T cells that are chronically exposed to antigens. Mechanistically, ZFP36L2 regulates IFN-γ production at late time points of activation by destabilizing Ifng mRNA in an AU-rich element-dependent manner. Together, our results reveal that ZFP36L2 employs different regulatory nodules in effector and memory T cells to regulate cytokine production.


Asunto(s)
Interferón gamma , Activación de Linfocitos , Tristetraprolina , Interferón gamma/inmunología , Interferón gamma/metabolismo , Animales , Tristetraprolina/genética , Tristetraprolina/metabolismo , Ratones , Activación de Linfocitos/inmunología , Linfocitos T CD8-positivos/inmunología , Ratones Noqueados , Ratones Endogámicos C57BL , Regulación de la Expresión Génica/inmunología
5.
Immunology ; 173(3): 511-519, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39046234

RESUMEN

The human innate immune system recognizes dsRNA as a pathogen-associated molecular pattern that induces a potent inflammatory response. The primary source of pathogenic dsRNA is cells infected with replicating viruses, but can also be released from uninfected necrotic cells. Here, we show that the dsRNA poly(I:C) challenge in human macrophages activates the p38 MAPK-MK2 signalling pathway and subsequently the phosphorylation of tristetraprolin (TTP/ZFP36). The latter is an mRNA decay-promoting protein that controls the stability of AU-rich mRNAs (AREs) that code for many inflammatory mediators. Hydroxychloroquine (HCQ), a common anti-malaria drug, is used to treat inflammatory and autoimmune disorders and, controversially, during acute COVID-19 disease. We found that HCQ reduced the dsRNA-dependent phosphorylation of p38 MAPK and its downstream kinase MK2. Subsequently, HCQ reduced the abundance and protein stability of the inactive (phosphorylated) form of TTP. HCQ reduced the levels and the mRNA stability of poly (I:C)-induced cytokines and inflammatory mRNAs like TNF, IL-6, COX-2, and IL-8 in THP-1 and primary blood monocytes. Our results demonstrate a new mechanism of the anti-inflammatory role of HCQ at post-transcriptional level (TTP phosphorylation) in a model of dsRNA activation, which usually occurs in viral infections or RNA release from necrotic tissue.


Asunto(s)
Hidroxicloroquina , Estabilidad del ARN , ARN Bicatenario , Tristetraprolina , Proteínas Quinasas p38 Activadas por Mitógenos , Humanos , Tristetraprolina/metabolismo , Estabilidad del ARN/efectos de los fármacos , Hidroxicloroquina/farmacología , Fosforilación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , ARN Bicatenario/metabolismo , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , SARS-CoV-2/inmunología , Poli I-C/farmacología , ARN Mensajero/metabolismo , ARN Mensajero/genética , Citocinas/metabolismo , COVID-19/virología , COVID-19/inmunología , Células THP-1 , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Transducción de Señal/efectos de los fármacos , Elementos Ricos en Adenilato y Uridilato , Tratamiento Farmacológico de COVID-19 , Proteínas Serina-Treonina Quinasas
6.
J Assist Reprod Genet ; 41(8): 1955-1963, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38829516

RESUMEN

PURPOSE: To explore the pathogenesis of oocyte maturation defects. METHODS: Whole exome sequencing was conducted to identify potential variants, which were then confirmed within the pedigree through Sanger sequencing. The functional characterization of the identified variants responsible for the disease, including their subcellular localization, protein levels, and interactions with other proteins, was verified through transient transfection in HeLa cells in vitro. Additionally, we employed real-time RT-PCR and single-cell RNA sequencing to examine the impact of ZFP36L2 pathogenic variants on mRNA metabolism in both HeLa cells and mouse or human oocytes. RESULTS: A novel compound heterozygous variant in ZFP36L2 (c.186T > G, p.His62Gln and c.869 C > T, p.Pro290Leu) was discovered in a patient with oocyte maturation defects. Our findings indicate that these variants lead to compromised binding capacity of the ZFP36L2-CONT6L complex and impaired mRNA degradation in HeLa cells and mouse oocytes. Furthermore, we characterized the changes in the human oocyte transcriptome associated with ZFP36L2 variants, with a particular emphasis on cell division, mitochondrial function, and ribosome metabolism. CONCLUSIONS: This study broadens the mutation spectrum of ZFP36L2 and constitutes the first report of human oocyte transcriptome alterations linked to ZFP36L2 variants. In conjunction with existing knowledge of ZFP36L2, our research lays the groundwork for genetic counseling aimed at addressing female infertility.


Asunto(s)
Infertilidad Femenina , Oocitos , Humanos , Femenino , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Oocitos/patología , Ratones , Células HeLa , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Animales , Secuenciación del Exoma , Linaje , Heterocigoto , Oogénesis/genética , Tristetraprolina/genética , Tristetraprolina/metabolismo , Mutación/genética , Adulto
7.
Cell Rep ; 43(4): 114069, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38602876

RESUMEN

The integrated stress response (ISR) is a key cellular signaling pathway activated by environmental alterations that represses protein synthesis to restore homeostasis. To prevent sustained damage, the ISR is counteracted by the upregulation of growth arrest and DNA damage-inducible 34 (GADD34), a stress-induced regulatory subunit of protein phosphatase 1 that mediates translation reactivation and stress recovery. Here, we uncover a novel ISR regulatory mechanism that post-transcriptionally controls the stability of PPP1R15A mRNA encoding GADD34. We establish that the 3' untranslated region of PPP1R15A mRNA contains an active AU-rich element (ARE) recognized by proteins of the ZFP36 family, promoting its rapid decay under normal conditions and stabilization for efficient expression of GADD34 in response to stress. We identify the tight temporal control of PPP1R15A mRNA turnover as a component of the transient ISR memory, which sets the threshold for cellular responsiveness and mediates adaptation to repeated stress conditions.


Asunto(s)
Regiones no Traducidas 3' , Proteína Fosfatasa 1 , Animales , Humanos , Ratones , Regiones no Traducidas 3'/genética , Adaptación Fisiológica/genética , Elementos Ricos en Adenilato y Uridilato/genética , Células HEK293 , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 1/genética , Estabilidad del ARN/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , Estrés Fisiológico/genética , Tristetraprolina/metabolismo , Tristetraprolina/genética
8.
BMC Res Notes ; 17(1): 82, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38504371

RESUMEN

OBJECTIVE: Tristetraprolin (TTP/ZFP36) family proteins exhibit antiinflammatory effects by destabilizing proinflammatory mRNAs. Previous studies showed that bacterial endotoxin lipopolysaccharides (LPS) stimulated TTP and tumor necrosis factor (TNF) gene expression, but less was known about LPS effects on TTP homologues and other proinflammatory gene expression in macrophages. The objective was to investigate LPS regulation of TTP family gene and TTP-targeted gene expression in mouse RAW264.7 macrophages using much higher concentrations of LPS and much longer treatment time than previous studies. RESULTS: MTT assay showed that LPS was not toxic to the cells under LPS treatment up to 1000 ng/mL for 2-24 h. LPS mildly affected the soluble protein content in the cells. qPCR assay showed that LPS stimulated TTP mRNA rapidly but not sustainably with 40, 10, and 3 fold of the DMSO control after 2, 8 and 24 h treatment, respectively. Immunoblotting confirmed qPCR results on LPS stimulation of TTP gene expression in the mouse macrophages. LPS exhibited minimal effects on ZFP36L1, ZFP36L2 and ZFP36L3 mRNA levels. LPS increased mRNA levels of TNF, COX2, GM-CSF, INFγ and IL12b up to 311, 418, 11, 9 and 4 fold, respectively. This study demonstrated that LPS did not affect macrophage viability, dramatically increased antiinflammatory TTP gene expression as well as proinflammatory TNF and COX2 gene expression but had only mild effects on TTP homologues and other proinflammatory cytokine gene expression in the mouse macrophages.


Asunto(s)
Lipopolisacáridos , Tristetraprolina , Ratones , Animales , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Tristetraprolina/genética , Tristetraprolina/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Macrófagos/metabolismo , Antiinflamatorios/farmacología , Expresión Génica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación de la Expresión Génica
9.
Mol Cell Biol ; 44(3): 87-102, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38520226

RESUMEN

Although LncRNA AA465934 expression is reduced in high glucose (HG)-treated podocytes, its role in HG-mediated podocyte injury and diabetic nephropathy (DN) remains unknown. Herein, we investigated the role of AA465934 in HG-mediated podocyte injury and DN using a spontaneous type II diabetic nephropathy (T2DN) model. The model was created by injecting AA465934 overexpressed adeno-associated virus (AAV) or control into mice. The levels of renal function, proteinuria, renal structural lesions, and podocyte apoptosis were then examined. Furthermore, AA465934 and autophagy levels, as well as tristetraprolin (TTP) and high mobility group box 1 (HMGB1) expression changes were detected. We also observed podocyte injury and the binding ability of TTP to E3 ligase proviral insertion in murine lymphomas 2 (PIM2), AA465934, or HMGB1. According to the results, AA465934 improved DN progression and podocyte damage in T2DN mice. In addition, AA465934 bound to TTP and inhibited its degradation by blocking TTP-PIM2 binding. Notably, TTP knock-down blocked the ameliorating effects of AA465934 and TTP bound HMGB1 mRNA, reducing its expression. Overexpression of HMGB1 inhibited the ability of AA465934 and TTP to improve podocyte injury. Furthermore, AA465934 bound TTP, inhibiting TTP-PIM2 binding, thereby suppressing TTP degradation, downregulating HMGB1, and reversing autophagy downregulation, ultimately alleviating HG-mediated podocyte injury and DN. Based on these findings, we deduced that the AA465934/TTP/HMGB1/autophagy axis could be a therapeutic avenue for managing podocyte injury and DN.


Asunto(s)
Nefropatías Diabéticas , Proteína HMGB1 , Podocitos , ARN Largo no Codificante , Animales , Ratones , Apoptosis , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Regulación hacia Abajo , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Podocitos/metabolismo , Podocitos/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Tristetraprolina/genética , Tristetraprolina/metabolismo
10.
Sci Rep ; 14(1): 3113, 2024 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326511

RESUMEN

Loss of inflammatory effector function, such as cytokine production and proliferation, is a fundamental driver of failure in T cell therapies against solid tumors. Here, we used CRISPR/Cas9 to genetically disrupt ZFP36, an RNA binding protein that regulates the stability of mRNAs involved in T cell inflammatory function, such as the cytokines IL2 and IFNγ, in human T cells engineered with a clinical-stage mesothelin-targeting CAR to determine whether its disruption could enhance antitumor responses. ZFP36 disruption slightly increased antigen-independent activation and cytokine responses but did not enhance overall performance in vitro or in vivo in a xenograft tumor model with NSG mice. While ZFP36 disruption does not reduce the function of CAR-T cells, these results suggest that singular disruption of ZFP36 is not sufficient to improve their function and may benefit from a multiplexed approach.


Asunto(s)
Inmunoterapia Adoptiva , Mesotelina , Humanos , Animales , Ratones , Inmunoterapia Adoptiva/métodos , Linfocitos T/metabolismo , Inmunidad , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Tristetraprolina/genética
11.
Cell Prolif ; 57(6): e13609, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38351596

RESUMEN

The association between Porphyromonas gingivalis infection and oral squamous cell carcinoma (OSCC) has been established by numerous epidemiological studies. However, the underlying mechanism specific to this connection remains unclear. By bioinformatical analysis, we identified ZFP36 as a potentially significant co-expressed gene in both the OSCC gene database and the persistent infection model of P. gingivalis. To further investigate the role of ZFP36, we established a cell model that human immortalized oral epithelial cells (HIOECs) that were sustainedly infected by P. gingivalis (MOI = 1) for a duration of 30 weeks. Our findings indicated that sustained infection with P. gingivalis inhibited the expression of ZFP36 protein and induced changes in the biological behaviour of HIOECs. The mechanism investigation demonstrated the potential role of ZFP36 in regulating the cancer-related biological behaviour of HIOECs. Subsequent studies revealed that highly expressed CCAT1 could serve as a molecular scaffold in the formation of the ZFP36/CCAT1/MK2 complex. This complex formation enhanced the binding abundance of MK2 and ZFP36, thereby promoting the inhibition of ZFP36 protein phosphorylation. To summarize, low expression of ZFP36 protein under persistent P. gingivalis infection enhances the cancer-related biological behaviour of HIOECs.


Asunto(s)
Infecciones por Bacteroidaceae , Células Epiteliales , Porphyromonas gingivalis , Tristetraprolina , Humanos , Porphyromonas gingivalis/patogenicidad , Células Epiteliales/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Infecciones por Bacteroidaceae/microbiología , Infecciones por Bacteroidaceae/metabolismo , Tristetraprolina/metabolismo , Tristetraprolina/genética , Neoplasias de la Boca/patología , Neoplasias de la Boca/microbiología , Neoplasias de la Boca/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/microbiología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Fosforilación
12.
STAR Protoc ; 5(1): 102807, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38165801

RESUMEN

Here, we present a protocol for modulating the function of the Cth2 mRNA-binding protein (RBP) in Saccharomyces cerevisiae. We describe steps to amplify and integrate mutations in Cth2 that affect its stability and function. Next, we detail the functional assay to verify the activity of the wild-type and mutant versions of Cth2 in yeast cells. This protocol can be adopted to modify the function of other RBPs with their respective functional mutations. For complete details on the use and execution of this protocol, please refer to Patnaik et al. (2022).1.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Portadoras/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tristetraprolina/genética , Tristetraprolina/metabolismo , Hierro/metabolismo
13.
Geroscience ; 46(1): 1271-1284, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37535204

RESUMEN

Tristetraprolin (TTP), encoded by Zfp36 in mice, is one of the best-characterized tandem zinc-finger mRNA binding proteins involved in mRNA deadenylation and decay. TTPΔARE mice lack an AU-rich motif in the 3'-untranslated regions of TTP mRNA, leading to increased TTP mRNA stability and more TTP protein, resulting in elevated mRNA decay rates of TTP targets. We examined the effect of TTP overexpression on the hematopoietic system in both young and middle-aged mice using TTPΔARE mice and found alterations in blood cell frequencies, with loss of platelets and B220 cells and gains of eosinophils and T cells. TTPΔARE mice also have skewed primitive populations in the bone marrow, with increases in myeloid-biased hematopoietic stem cells (HSCs) but decreases in granulocyte/macrophage-biased multipotent progenitors (MPP3) in both young and middle-aged mice. Changes in the primitive cells' frequencies were associated with transcriptional alterations in the TTP overexpression cells specific to age as well as cell type. Regardless of age, there was a consistent elevation of transcripts regulated by TNFα and TGFß signaling pathways in both the stem and multipotent progenitor populations. HSCs with TTP overexpression had decreased reconstitution potential in murine transplants but generated hematopoietic environments that mitigated the inflammatory response to the collagen antibody-induced arthritis (CAIA) challenge, which models rheumatoid arthritis and other autoimmune disorders. This dampening of the inflammatory response was even present when there was only a small frequency of TTP overexpressing cells present in the middle-aged mice. We provide an analysis of the early hematopoietic compartments with elevated TTP expression in both young and middle-aged mice which inhibits the reconstitution potential of the HSCs but generates a hematopoietic system that provides dominant repression of induced inflammation.


Asunto(s)
Sistema Hematopoyético , Tristetraprolina , Animales , Ratones , Regiones no Traducidas 3' , Modelos Animales de Enfermedad , Sistema Hematopoyético/metabolismo , Inflamación/genética , Ratones Noqueados , Tristetraprolina/genética , Tristetraprolina/metabolismo
14.
RNA Biol ; 21(1): 1-15, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38111129

RESUMEN

Inhibition of apoptosis is one of the hallmarks of cancer and is a target of various therapeutic interventions. BIRC5 is an inhibitor of apoptosis that is aberrantly expressed in cancer leading to sustained growth of tumours. Post-transcriptional control mechanisms involving RNA-binding proteins and AU-rich elements (AREs) are fundamental to many cellular processes and changes in the expression or function of these proteins can promote an aberrant and pathological phenotype. BIRC5 mRNA has an ARE in its 3' UTR making it a candidate for regulation by the RNA binding proteins tristetraprolin (TTP) and HuR (ELAVL1). In this study, we investigated the binding of TTP and HuR by RNA-immunoprecipitation assays and found that these proteins were associated with BIRC5 mRNA to varying extents. Consequently, BIRC5 expression decreased when TTP was overexpressed and apoptosis was induced. In the absence of TTP, BIRC5 mRNA was stabilized, protein expression increased and the number of apoptotic cells declined. As an ARE-mRNA stabilizing protein, recombinant HuR led to upregulation of BIRC5 expression, whereas HuR silencing was concomitant with downregulation of BIRC5 mRNA and protein and increased cell death. Survival analyses demonstrated that increased TTP and low BIRC5 expression predicted an overall better prognosis compared to dysregulated TTP and high BIRC5. Thus, the results present a novel target of ARE-mediated post-transcriptional regulation.


Asunto(s)
Neoplasias de la Mama , Tristetraprolina , Humanos , Femenino , Tristetraprolina/genética , Tristetraprolina/metabolismo , Survivin/genética , Survivin/metabolismo , Neoplasias de la Mama/genética , Regiones no Traducidas 3' , Apoptosis/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Estabilidad del ARN/genética
15.
Life Sci Alliance ; 7(1)2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37903626

RESUMEN

Members of the tristetraprolin (TTP) family of RNA-binding proteins can bind to and promote the decay of specific transcripts containing AU-rich motifs. ZFP36 (TTP) is best known for regulating pro-inflammatory cytokine expression in myeloid cells; however, its mammalian paralogues ZFP36L1 and ZFP36L2 have not been viewed as important in controlling inflammation. We knocked out these genes in myeloid cells in mice, singly and together. Single-gene myeloid-specific knockouts resulted in almost no spontaneous phenotypes. In contrast, mice with myeloid cell deficiency of all three genes developed severe inflammation, with a median survival of 8 wk. Macrophages from these mice expressed many more stabilized transcripts than cells from myeloid-specific TTP knockout mice; many of these encoded pro-inflammatory cytokines and chemokines. The failure of weight gain, arthritis, and early death could be prevented completely by two normal alleles of any of the three paralogues, and even one normal allele of Zfp36 or Zfp36l2 was enough to prevent the inflammatory phenotype. Our findings emphasize the importance of all three family members, acting in concert, in myeloid cell function.


Asunto(s)
Inflamación , Tristetraprolina , Ratones , Animales , Tristetraprolina/genética , Tristetraprolina/metabolismo , Inflamación/genética , Inflamación/metabolismo , Células Mieloides/metabolismo , Macrófagos/metabolismo , Ratones Noqueados , Citocinas/metabolismo , Mamíferos/metabolismo
16.
FASEB J ; 38(1): e23338, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38038723

RESUMEN

Tristetraprolin (TTP; also known as NUP475, GOS24, or TIS11), encoded by Zfp36, is an RNA-binding protein that regulates target gene expression by promoting mRNA decay and preventing translation. Although previous studies have indicated that TTP deficiency is associated with systemic inflammation and a catabolic-like skeletal phenotype, the mechanistic underpinnings remain unclear. Here, using both TTP-deficient (TTPKO) and myeloid-specific TTPKO (cTTPKO) mice, we reveal that global absence or loss of TTP in the myeloid compartment results in a reduced bone microarchitecture, whereas gain-of-function TTP knock-in (TTPKI) mice exhibit no significant loss of bone microarchitecture. Flow cytometry analysis revealed a significant immunosuppressive immune cell phenotype with increased monocytic myeloid-derived suppressor cells (M-MDSCs) in TTPKO and cTTPKO mice, whereas no significant changes were observed in TTPKI mice. Single-cell transcriptomic analyses of bone marrow myeloid progenitor cell populations indicated a dramatic increase in early MDSC marker genes for both cTTPKO and TTPKO bone marrow populations. Consistent with these phenotypic and transcriptomic data, in vitro osteoclastogenesis analysis of bone marrow M-MDSCs from cTTPKO and TTPKO displayed enhanced osteoclast differentiation and functional capacity. Focused transcriptomic analyses of differentiated M-MDSCs showed increased osteoclast-specific transcription factors and cell fusion gene expression. Finally, functional data showed that M-MDSCs from TTP loss-of-function mice were capable of osteoclastogenesis and bone resorption in a context-dependent manner. Collectively, these findings indicate that TTP plays a central role in regulating osteoclastogenesis through multiple mechanisms, including induction of M-MDSCs that appear to regulate skeletal phenotype.


Asunto(s)
Células Supresoras de Origen Mieloide , Tristetraprolina , Animales , Ratones , Osteoclastos/metabolismo , Osteogénesis , Fenotipo , Tristetraprolina/genética
17.
mBio ; 14(5): e0174223, 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37707056

RESUMEN

IMPORTANCE: Our findings define a novel role for ZIKV-induced TTP expression in regulating IFNß/IFNλ production in primary hBMECs and Sertoli cells. These cells comprise key physiological barriers subverted by ZIKV to access brain and testicular compartments and serve as reservoirs for persistent replication and dissemination. We demonstrate for the first time that the ARE-binding protein TTP is virally induced and post-transcriptionally regulates IFNß/IFNλ secretion. In ZIKV-infected hBMEC and Sertoli cells, TTP knockout increased IFNß/IFNλ secretion, while TTP expression blocked IFNß/IFNλ secretion. The TTP-directed blockade of IFN secretion permits ZIKV spread and persistence in hBMECs and Sertoli cells and may similarly augment ZIKV spread across IFNλ-protected placental barriers. Our work highlights the importance of post-transcriptional ZIKV regulation of IFN expression and secretion in cells that regulate viral access to protected compartments and defines a novel mechanism of ZIKV-regulated IFN responses which may facilitate neurovirulence and sexual transmission.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Embarazo , Masculino , Femenino , Humanos , Células de Sertoli/metabolismo , Virus Zika/fisiología , Infección por el Virus Zika/metabolismo , Tristetraprolina , Placenta/metabolismo , Replicación Viral
18.
Cell Death Dis ; 14(8): 527, 2023 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-37587140

RESUMEN

Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, with high morbidity and mortality worldwide. Although the dysregulation of BARX1 expression has been shown to be associated with malignant cancers, including NSCLC, the underlying mechanism remains elusive. In this study, we identified BARX1 as a common differentially expressed gene in lung squamous cell carcinoma and adenocarcinoma. Importantly, we uncovered a novel mechanism behind the regulation of BARX1, in which ZFP36 interacted with 3'UTR of BARX1 mRNA to mediate its destabilization. Loss of ZFP36 led to the upregulation of BARX1, which further promoted the proliferation, migration and invasion of NSCLC cells. In addition, the knockdown of BARX1 inhibited tumorigenicity in mouse xenograft. We demonstrated that BARX1 promoted the malignant phenotypes by transactivating a set of master oncogenes involved in the cell cycle, DNA synthesis and metastasis. Overall, our study provides insights into the mechanism of BARX1 actions in NSCLC and aids a better understanding of NSCLC pathogenesis.


Asunto(s)
Adenocarcinoma , Carcinoma de Pulmón de Células no Pequeñas , Proteínas de Homeodominio , Neoplasias Pulmonares , Factores de Transcripción , Tristetraprolina , Animales , Humanos , Ratones , Carcinoma de Pulmón de Células no Pequeñas/genética , Proteínas de Homeodominio/genética , Neoplasias Pulmonares/genética , Oncogenes , Fenotipo , Factores de Transcripción/genética , Tristetraprolina/genética
19.
Biochim Biophys Acta Gene Regul Mech ; 1866(3): 194959, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37453649

RESUMEN

Sterol synthesis is an iron-dependent metabolic pathway in eukaryotes. Consequently, fungal ergosterol biosynthesis (ERG) is down-regulated in response to iron deficiency. In this report, we show that, upon iron limitation or overexpression of the iron-regulated mRNA-binding protein Cth2, the yeast Saccharomyces cerevisiae down-regulates the three initial enzymatic steps of ergosterol synthesis (ERG1, ERG7 and ERG11). Mechanistically, we show that Cth2 protein limits the translation and promotes the decrease in the mRNA levels of these specific ERG genes, which contain consensus Cth2-binding sites defined as AU-rich elements (AREs). Thus, expression of CTH2 leads to the accumulation of initial sterol intermediates, such as squalene, and to the drop of ergosterol levels. Changes in CTH2 expression levels disturb the response of yeast cells to stresses related to membrane integrity such as high ethanol and sorbitol concentrations. Therefore, CTH2 should be considered as a critical regulatory factor of ergosterol biosynthesis during iron deficiency.


Asunto(s)
Deficiencias de Hierro , Proteínas de Saccharomyces cerevisiae , Humanos , Ergosterol/metabolismo , Hierro/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Esteroles/metabolismo , Tristetraprolina/genética , Tristetraprolina/metabolismo
20.
Biomolecules ; 13(4)2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-37189372

RESUMEN

Gossypol is a complex plant polyphenol reported to be cytotoxic and anti-inflammatory, but little is known about its effect on gene expression in macrophages. The objective of this study was to explore gossypol's toxicity and its effect on gene expression involved in the inflammatory response, glucose transport and insulin signaling pathways in mouse macrophages. Mouse RAW264.7 macrophages were treated with multiple concentrations of gossypol for 2-24 h. Gossypol toxicity was estimated by MTT assay and soluble protein content. qPCR analyzed the expression of anti-inflammatory tristetraprolin family (TTP/ZFP36), proinflammatory cytokine, glucose transporter (GLUT) and insulin signaling genes. Cell viability was greatly reduced by gossypol, accompanied with a dramatic reduction in soluble protein content in the cells. Gossypol treatment resulted in an increase in TTP mRNA level by 6-20-fold and increased ZFP36L1, ZFP36L2 and ZFP36L3 mRNA levels by 26-69-fold. Gossypol increased proinflammatory cytokine TNF, COX2, GM-CSF, INFγ and IL12b mRNA levels up to 39-458-fold. Gossypol treatment upregulated mRNA levels of GLUT1, GLUT3 and GLUT4 genes as well as INSR, AKT1, PIK3R1 and LEPR, but not APP genes. This study demonstrated that gossypol induced macrophage death and reduced soluble protein content, which was accompanied with the massive stimulation of anti-inflammatory TTP family and proinflammatory cytokine gene expression, as well as the elevation of gene expression involved in glucose transport and the insulin signaling pathway in mouse macrophages.


Asunto(s)
Gosipol , Polifenoles , Ratones , Animales , Polifenoles/farmacología , Polifenoles/metabolismo , Gosipol/farmacología , Macrófagos/metabolismo , Insulina/metabolismo , Antiinflamatorios/farmacología , Citocinas/metabolismo , Expresión Génica , ARN Mensajero/metabolismo , Muerte Celular , Glucosa/metabolismo , Tristetraprolina/genética , Tristetraprolina/metabolismo , Tristetraprolina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...