Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Rocio T. Martinez-Nunez is active.

Publication


Featured researches published by Rocio T. Martinez-Nunez.


Journal of Biological Chemistry | 2009

MicroRNA-155 modulates the pathogen binding ability of dendritic cells (DCs) by down-regulation of DC-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN).

Rocio T. Martinez-Nunez; Fethi Louafi; Peter S. Friedmann; Tilman Sanchez-Elsner

MicroRNA-155 (miR-155) has been involved in the response to inflammation in macrophages and lymphocytes. Here we show how miR-155 participates in the maturation of human dendritic cells (DC) and modulates pathogen binding by down-regulating DC-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN), after directly targeting the transcription factor PU.1. During the maturation of DCs, miR-155 increases up to 130-fold, whereas PU.1 protein levels decrease accordingly. We establish that human PU.1 is a direct target for miR-155 and localize the target sequence for miR-155 in the 3′-untranslated region of PU.1. Also, overexpression of miR-155 in the THP1 monocytic cell line decreases PU.1 protein levels and DC-SIGN at both the mRNA and protein levels. We prove a link between the down-regulation of PU.1 and reduced transcriptional activity of the DC-SIGN promoter, which is likely to be the basis for its reduced mRNA expression, after miR-155 overexpression. Finally, we show that, by reducing DC-SIGN in the cellular membrane, miR-155 is involved in regulating pathogen binding as dendritic cells exhibited the lower binding capacity for fungi and HIV protein gp-120 when the levels of miR-155 were higher. Thus, our results suggest a mechanism by which miR-155 regulates proteins involved in the cellular immune response against pathogens that could have clinical implications in the way pathogens enter the human organism.


Journal of Biological Chemistry | 2011

The Interleukin 13 (IL-13) Pathway in Human Macrophages Is Modulated by MicroRNA-155 via Direct Targeting of Interleukin 13 Receptor α1 (IL13Rα1)

Rocio T. Martinez-Nunez; Fethi Louafi; Tilman Sanchez-Elsner

Macrophages play a central role in the balance and efficiency of the immune response and are at the interface between innate and adaptive immunity. Their phenotype is a delicate equilibrium between the M1 (classical, pro-Th1) and M2 (alternative, pro-Th2) profiles. This balance is regulated by cytokines such as interleukin 13 (IL-13), a typical pro-M2-Th2 cytokine that has been related to allergic disease and asthma. IL-13 binds to IL-13 receptor α1 (IL13Rα1), a component of the Type II IL-4 receptor, and exerts its effects by activating the transcription factor signal transducer and activator of transcription 6 (STAT6) through phosphorylation. MicroRNAs are short (∼22 nucleotide) inhibitory non-coding RNAs that block the translation or promote the degradation of their specific mRNA targets. By bioinformatics analysis, we found that microRNA-155 (miR-155) is predicted to target IL13Rα1. This suggested that miR-155 might be involved in the regulation of the M1/M2 balance in macrophages by modulating IL-13 effects. miR-155 has been implicated in the development of a healthy immune system and function as well as in the inflammatory pro-Th1/M1 immune profile. Here we have shown that in human macrophages, miR-155 directly targets IL13Rα1 and reduces the levels of IL13Rα1 protein, leading to diminished activation of STAT6. Finally we also demonstrate that miR-155 affects the IL-13-dependent regulation of several genes (SOCS1, DC-SIGN, CCL18, CD23, and SERPINE) involved in the establishment of an M2/pro-Th2 phenotype in macrophages. Our work shows a central role for miR-155 in determining the M2 phenotype in human macrophages.


Journal of Biological Chemistry | 2010

MicroRNA-155 targets SMAD2 and modulates the response of macrophages to transforming growth factor-{beta}

Fethi Louafi; Rocio T. Martinez-Nunez; Tilman Sanchez-Elsner

Transforming growth factor-beta (TGF-β) is a pleiotropic cytokine with important effects on processes such as fibrosis, angiogenesis, and immunosupression. Using bioinformatics, we identified SMAD2, one of the mediators of TGF-β signaling, as a predicted target for a microRNA, microRNA-155 (miR-155). MicroRNAs are a class of small non-coding RNAs that have emerged as an important class of gene expression regulators. miR-155 has been found to be involved in the regulation of the immune response in myeloid cells. Here, we provide direct evidence of binding of miR-155 to a predicted binding site and the ability of miR-155 to repress SMAD2 protein expression. We employed a lentivirally transduced monocyte cell line (THP1–155) containing an inducible miR-155 transgene to show that endogenous levels of SMAD2 protein were decreased after sustained overexpression of miR-155. This decrease in SMAD2 led to a reduction in both TGF-β-induced SMAD-2 phosphorylation and SMAD-2-dependent activation of the expression of the CAGA12LUC reporter plasmid. Overexpression of miR-155 altered the cellular responses to TGF-β by changing the expression of a set of genes that is involved in inflammation, fibrosis, and angiogenesis. Our study provides firm evidence of a role for miR-155 in directly repressing SMAD2 expression, and our results demonstrate the relevance of one of the two predicted target sites in SMAD2 3′-UTR. Altogether, our data uncover an important role for miR-155 in modulating the cellular response to TGF-β with possible implications in several human diseases where homeostasis of TGF-β might be altered.


Journal of Biological Chemistry | 2008

Structural Requirements for Multimerization of the Pathogen Receptor Dendritic Cell-specific ICAM3-grabbing Non-integrin (CD209) on the Cell Surface

Diego Serrano-Gómez; Elena Sierra-Filardi; Rocio T. Martinez-Nunez; Esther Caparrós; Rafael Delgado; Mari Angeles Muñoz-Fernández; M. A. Abad; Jesús Jiménez-Barbero; Manuel Leal; Angel L. Corbí

The myeloid C-type lectin dendritic cell-specific ICAM3-grabbing non-integrin (DC-SIGN, CD209) recognizes oligosaccharide ligands on clinically relevant pathogens (HIV, Mycobacterium, and Aspergillus). Alternative splicing and genomic polymorphism generate DC-SIGN mRNA variants, which have been detected at sites of pathogen entrance and transmission. We present evidence that DC-SIGN neck variants are expressed on dendritic and myeloid cells at the RNA and protein levels. Structural analysis revealed that multimerization of DC-SIGN within a cellular context depends on the lectin domain and the number and arrangement of the repeats within the neck region, whose glycosylation negatively affects oligomer formation. Naturally occurring DC-SIGN neck variants differ in multimerization competence in the cell membrane, exhibit altered sugar binding ability, and retain pathogen-interacting capacity, implying that pathogen-induced cluster formation predominates over the basal multimerization capability. Analysis of DC-SIGN neck polymorphisms indicated that the number of allelic variants is higher than previously thought and that multimerization of the prototypic molecule is modulated in the presence of allelic variants with a different neck structure. Our results demonstrate that the presence of allelic variants or a high level of expression of neck domain splicing isoforms might influence the presence and stability of DC-SIGN multimers on the cell surface, thus providing a molecular explanation for the correlation between DC-SIGN polymorphisms and altered susceptibility to HIV-1 and other pathogens.


PLOS ONE | 2014

A microRNA network dysregulated in asthma controls IL-6 production in bronchial epithelial cells.

Rocio T. Martinez-Nunez; Victor P. Bondanese; Fethi Louafi; Ana S. Francisco-Garcia; Hitasha Rupani; Nicole Bedke; Stephen T. Holgate; Peter H. Howarth; Donna E. Davies; Tilman Sanchez-Elsner

MicroRNAs are short non-coding single stranded RNAs that regulate gene expression. While much is known about the effects of individual microRNAs, there is now growing evidence that they can work in co-operative networks. MicroRNAs are known to be dysregulated in many diseases and affect pathways involved in the pathology. We investigated dysregulation of microRNA networks using asthma as the disease model. Asthma is a chronic inflammatory disease of the airways characterized by bronchial hyperresponsiveness and airway remodelling. The airway epithelium is a major contributor to asthma pathology and has been shown to produce an excess of inflammatory and pro-remodelling cytokines such as TGF-β, IL-6 and IL-8 as well as deficient amounts of anti-viral interferons. After performing microRNA arrays, we found that microRNAs -18a, -27a, -128 and -155 are down-regulated in asthmatic bronchial epithelial cells, compared to cells from healthy donors. Interestingly, these microRNAs are predicted in silico to target several components of the TGF-β, IL-6, IL-8 and interferons pathways. Manipulation of the levels of individual microRNAs in bronchial epithelial cells did not have an effect on any of these pathways. Importantly, knock-down of the network of microRNAs miR-18a, -27a, -128 and -155 led to a significant increase of IL-8 and IL-6 expression. Interestingly, despite strong in silico predictions, down-regulation of the pool of microRNAs did not have an effect on the TGF-β and Interferon pathways. In conclusion, using both bioinformatics and experimental tools we found a highly relevant potential role for microRNA dysregulation in the control of IL-6 and IL-8 expression in asthma. Our results suggest that microRNAs may have different roles depending on the presence of other microRNAs. Thus, interpretation of in silico analysis of microRNA function should be confirmed experimentally in the relevant cellular context taking into account interactions with other microRNAs when studying disease.


Immunobiology | 2010

The novel RUNX3/p33 isoform is induced upon monocyte-derived dendritic cell maturation and downregulates IL-8 expression.

Amaya Puig-Kröger; Noemí Aguilera-Montilla; Rocio T. Martinez-Nunez; Ángeles Domínguez-Soto; Fátima Sánchez-Cabo; Enrique Martín-Gayo; Ángel Zaballos; María L. Toribio; Yoram Groner; Yoshiaki Ito; Ana Dopazo; María Teresa Corcuera; María J. Alonso Martín; Miguel A. Vega; Angel L. Corbí

RUNX proteins are heterodimeric factors that play crucial roles during development and differentiation of cells of the immune system. The RUNX3 transcription factor controls lineage decisions during thymopoiesis and T-cell differentiation, and modulates myeloid cell effector functions. We now report the characterization of the human RUNX3/p33 isoform, generated by splicing out a Runt DNA-binding domain-encoding exon, and whose transcriptional activities differ from those of the prototypic RUNX3/p44 molecule. Unlike RUNX3/p44, RUNX3/p33 is induced upon maturation of monocyte-derived dendritic cells (MDDC), and is unable to transactivate the regulatory regions of the CD11a, CD11c and CD49e integrin genes. Overexpression of RUNX3/p33 in myeloid cell lines led to diminished expression of genes involved in inflammatory responses. Moreover, overexpression of RUNX3/p33 down-modulated the basal level of IL-8 production from immature monocyte-derived dendritic cells (MDDC). Besides, siRNA-mediated knock-down of RUNX3 led to diminished levels of IL-8 RNA in immature MDDC, and modulated the neutrophil-recruiting capacity of myeloid cell line supernatants. Since IL-8 promotes neutrophil chemotaxis and degranulation during inflammatory responses, and exerts mitogenic and angiogenic actions within tumor microenvironment, our results imply that myeloid RUNX3 expression regulates the recruitment of leukocytes towards inflammatory foci and might also contribute to human cancer progression.


Antimicrobial Agents and Chemotherapy | 2007

AM3 Modulates Dendritic Cell Pathogen Recognition Capabilities by Targeting DC-SIGN

Diego Serrano-Gómez; Rocio T. Martinez-Nunez; Elena Sierra-Filardi; Nuria Izquierdo; María Colmenares; Jesús Pla; Luis Rivas; Javier Martinez-Picado; Jesús Jiménez-Barbero; José Luis Alonso-Lebrero; Salvador González; Angel L. Corbí

ABSTRACT AM3 (Inmunoferon) is an orally effective immunomodulator that influences the regulatory and effector functions of the immune system whose molecular mechanisms of action are mostly unknown. We hypothesized that the polysaccharide moiety of AM3 (IF-S) might affect immune responses by modulating the lectin-dependent pathogen recognition abilities of human dendritic cells. IF-S inhibited binding of viral, fungal, and parasite pathogens by human monocyte-derived dendritic cells in a dose-dependent manner. IF-S specifically impaired the pathogen recognition capabilities of DC-SIGN, as it reduced the attachment of Candida, Aspergillus, and Leishmania to DC-SIGN transfectants. IF-S also inhibited the interaction of DC-SIGN with both its cellular counterreceptor (intercellular adhesion molecule 3) and the human immunodeficiency virus (HIV) type 1 gp120 protein and blocked the DC-SIGN-dependent capture of HIV virions and the HIV trans-infection capability of DC-SIGN transfectants. IF-S promoted DC-SIGN internalization in DCs without affecting mannose receptor expression, and 1D saturation transfer difference nuclear magnetic resonance demonstrated that IF-S directly interacts with DC-SIGN on the cell surface. Therefore, the polysaccharide moiety of AM3 directly influences pathogen recognition by dendritic cells by interacting with DC-SIGN. Our results indicate that DC-SIGN is the target for an immunomodulator and imply that the adjuvant and immunomodulatory actions of AM3 are mediated, at least in part, by alteration of the DC-SIGN functional activities.


Methods of Molecular Biology | 2016

Studying Isoform-Specific mRNA Recruitment to Polyribosomes with Frac-seq

Rocio T. Martinez-Nunez; Jeremy R. Sanford

Gene expression profiling is widely used as a measure of the protein output of cells. However, it is becoming more evident that there are multiple layers of post-transcriptional gene regulation that greatly impact protein output (Battle et al., Science 347:664-667, 2014; Khan et al., Science 342:1100-1104, 2013; Vogel et al., Mol Syst Biol 6:400, 2010). Alternative splicing (AS) impacts the expression of protein coding genes in several ways. Firstly, AS increases exponentially the coding-capacity of genes generating multiple transcripts from the same genomic sequence. Secondly, alternatively spliced mRNAs are subjected differentially to RNA-degradation via pathways such as nonsense mediated decay (AS-NMD) or microRNAs (Shyu et al., EMBO J 27:471-481, 2008). And thirdly, cytoplasmic export from the nucleus and translation are regulated in an isoform-specific manner, adding an extra layer of regulation that impacts the protein output of the cell (Martin and Ephrussi, Cell 136:719-730, 2009; Sterne-Weiler et al., Genome Res 23:1615-1623, 2013). These data highlight the need of a method that allows analyzing both the nuclear events (AS) and the cytoplasmic fate (polyribosome-binding) of individual mRNA isoforms.In order to determine how alternative splicing determines the polyribosome association of mRNA isoforms we developed Frac-seq. Frac-seq combines subcellular fractionation and high throughput RNA sequencing (RNA-seq). Frac-seq gives a window onto the translational fate of specific alternatively spliced isoforms on a genome-wide scale. There is evidence of preferential translation of specific mRNA isoforms (Coldwell and Morley, Mol Cell Biol 26:8448-8460, 2006; Sanford et al., Genes Dev 18:755-768; Zhong et al., Mol Cell 35:1-10, 2009; Michlewski et al., Mol Cell 30:179-189, 2008); the advantage of Frac-seq is that it allows analyzing the binding of alternatively spliced isoforms to polyribosomes and comparing their relative abundance to the cytosolic fraction. Polyribosomes are resolved by sucrose gradient centrifugation of cytoplasmic extracts, subsequent reading and extraction. The total mRNA fraction is taken prior ultracentrifugation as a measure of all mRNAs present in the sample. Both populations of RNAs are then isolated using phenol-chloroform precipitation; polyadenylated RNAs are selected and converted into libraries and sequenced. Bioinformatics analysis is then performed to measure alternatively spliced isoforms; several tools can be used such as MISO, RSEM, or Cufflinks (Katz et al., Nat Methods 7:1009-1015, 2010; Li and Dewey, BMC Bioinformatics 12:323, 2011; Trapnell et al., Nat Protoc 7:562-578, 2012). Comparison of total mRNAs and polyribosome-bound mRNAs can be used as a measure of the polyribosome association of specific isoforms based on the presence/absence of specific alternative splicing events in each fraction. Frac-seq shows that not all isoforms from a gene are equally loaded into polyribosomes, that mRNA preferential loading does not always correlate to its expression in the cytoplasm and that the presence of specific events such as microRNA binding sites or Premature Termination Codons determine the loading of specific isoforms into polyribosomes.


Nucleic Acids Research | 2017

Modulation of nonsense mediated decay by rapamycin

Rocio T. Martinez-Nunez; Andrew Wallace; Doyle Coyne; Linnea Jansson; Miles Rush; Hanane Ennajdaoui; Sol Katzman; Joanne L. Bailey; Katrin Deinhardt; Tilman Sanchez-Elsner; Jeremy R. Sanford

Abstract Rapamycin is a naturally occurring macrolide whose target is at the core of nutrient and stress regulation in a wide range of species. Despite well-established roles as an inhibitor of cap-dependent mRNA translation, relatively little is known about its effects on other modes of RNA processing. Here, we characterize the landscape of rapamycin-induced post-transcriptional gene regulation. Transcriptome analysis of rapamycin-treated cells reveals genome-wide changes in alternative mRNA splicing and pronounced changes in NMD-sensitive isoforms. We demonstrate that despite well-documented attenuation of cap-dependent mRNA translation, rapamycin can augment NMD of certain transcripts. Rapamycin-treatment significantly reduces the levels of both endogenous and exogenous Premature Termination Codon (PTC)-containing mRNA isoforms and its effects are dose-, UPF1- and 4EBP-dependent. The PTC-containing SRSF6 transcript exhibits a shorter half-life upon rapamycin-treatment as compared to the non-PTC isoform. Rapamycin-treatment also causes depletion of PTC-containing mRNA isoforms from polyribosomes, underscoring the functional relationship between translation and NMD. Enhanced NMD activity also correlates with an enrichment of the nuclear Cap Binding Complex (CBC) in rapamycin-treated cells. Our data demonstrate that rapamycin modulates global RNA homeostasis by NMD.


Genes | 2018

MicroRNA-31 and MicroRNA-155 Are Overexpressed in Ulcerative Colitis and Regulate IL-13 Signaling by Targeting Interleukin 13 Receptor α-1

Markus Gwiggner; Rocio T. Martinez-Nunez; Simon R. Whiteoak; Victor P. Bondanese; Andy Claridge; Jane E. Collins; J. Cummings; Tilman Sanchez-Elsner

Interleukin-13 (IL-13) is an important Type 2 T helper (Th2) cytokine, controlling biological functions in epithelium and has been linked to asthma, atopic dermatitis and ulcerative colitis (UC). Interleukin-13 signals through IL-13 receptor α-1 (IL13RA1 (gene) and IL13Rα1 (protein)), a receptor that can be regulated by microRNAs (miRs). MicroRNAs are small non-coding single-stranded RNAs with a role in several pathologies. However, their relevance in the pathophysiology of UC, a chronic inflammatory condition of the colonic mucosa, is poorly characterised. Here, we determined the expression of IL13Rα1 in UC, its potential regulation by miRs and the subsequent effect on IL-13 signalling. Inflamed mucosa of UC patients showed decreased mRNA and protein expression of IL13RA1 when compared to healthy controls. We show that miR-31 and miR-155 are upregulated in inflamed UC mucosa and that both directly target the 3′ untranslated region of IL13RA1 mRNA. Transfection of miR-31 and miR-155 mimics reduced the expression of IL13RA1 mRNA and protein, and blocked IL-13-dependent phosphorylation of signal transducer and activator of transcription 6 (STAT6) in HT-29 cells, a gut epithelium cell line. Interleukin-13 activation of suppressor of cytokine signaling 1 (SOCS1) and eotaxin-3 (CCL26) expression was also diminished. MicroRNA-31/microRNA-155 mimics also downregulated IL13RA1 in ex vivo human inflamed UC biopsies. We propose that miR-31 and miR-155 have an important role in limiting IL-13 signalling in UC disease.

Collaboration


Dive into the Rocio T. Martinez-Nunez's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hitasha Rupani

University of Southampton

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fethi Louafi

University of Southampton

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Angel L. Corbí

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Miles Rush

University of Southampton

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Diego Serrano-Gómez

Spanish National Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge