Network


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

Hotspot


Dive into the research topics where Mariam Quiñones is active.

Publication


Featured researches published by Mariam Quiñones.


Science | 2012

Compartmentalized Control of Skin Immunity by Resident Commensals

Shruti Naik; Nicolas Bouladoux; Christoph Wilhelm; Michael J. Molloy; Rosalba Salcedo; Wolfgang Kastenmüller; Clayton Deming; Mariam Quiñones; Lily Koo; Sean Conlan; Sean P. Spencer; Jason A. Hall; Amiran K. Dzutsev; Heidi Kong; Daniel J. Campbell; Giorgio Trinchieri; Julia A. Segre; Yasmine Belkaid

Skin Specifics Much of the recent attention paid to the trillions of bacteria that colonize our bodies has been given to the bacteria that reside in the gut. Naik et al. (p. 1115, published online 26 July) report that colonization of the skin with commensal bacteria is important for tuning effector T cell responses in the skin and for protective immunity against cutaneous infection with the parasite Leishmania major in mice. In contrast, selective depletion of the gut microbiota, which plays an important role in modulating immune responses in the gut, had no impact on T cell responses in the skin. The skin microbiota play a selective role in modulating immunity in the skin of mice. Intestinal commensal bacteria induce protective and regulatory responses that maintain host-microbial mutualism. However, the contribution of tissue-resident commensals to immunity and inflammation at other barrier sites has not been addressed. We found that in mice, the skin microbiota have an autonomous role in controlling the local inflammatory milieu and tuning resident T lymphocyte function. Protective immunity to a cutaneous pathogen was found to be critically dependent on the skin microbiota but not the gut microbiota. Furthermore, skin commensals tuned the function of local T cells in a manner dependent on signaling downstream of the interleukin-1 receptor. These findings underscore the importance of the microbiota as a distinctive feature of tissue compartmentalization, and provide insight into mechanisms of immune system regulation by resident commensal niches in health and disease.


Nature | 2015

Commensal–dendritic-cell interaction specifies a unique protective skin immune signature

Shruti Naik; Nicolas Bouladoux; Jonathan L. Linehan; Seong-Ji Han; Oliver J. Harrison; Christoph Wilhelm; Sean Conlan; Sarah Himmelfarb; Allyson L. Byrd; Clayton Deming; Mariam Quiñones; Jason M. Brenchley; Heidi H. Kong; Roxanne Tussiwand; Kenneth M. Murphy; Miriam Merad; Julia A. Segre; Yasmine Belkaid

The skin represents the primary interface between the host and the environment. This organ is also home to trillions of microorganisms that play an important role in tissue homeostasis and local immunity. Skin microbial communities are highly diverse and can be remodelled over time or in response to environmental challenges. How, in the context of this complexity, individual commensal microorganisms may differentially modulate skin immunity and the consequences of these responses for tissue physiology remains unclear. Here we show that defined commensals dominantly affect skin immunity and identify the cellular mediators involved in this specification. In particular, colonization with Staphylococcus epidermidis induces IL-17A+ CD8+ T cells that home to the epidermis, enhance innate barrier immunity and limit pathogen invasion. Commensal-specific T-cell responses result from the coordinated action of skin-resident dendritic cell subsets and are not associated with inflammation, revealing that tissue-resident cells are poised to sense and respond to alterations in microbial communities. This interaction may represent an evolutionary means by which the skin immune system uses fluctuating commensal signals to calibrate barrier immunity and provide heterologous protection against invasive pathogens. These findings reveal that the skin immune landscape is a highly dynamic environment that can be rapidly and specifically remodelled by encounters with defined commensals, findings that have profound implications for our understanding of tissue-specific immunity and pathologies.


BMC Genomics | 2011

Directional gene expression and antisense transcripts in sexual and asexual stages of Plasmodium falciparum

María José López-Barragán; Jacob Lemieux; Mariam Quiñones; Kim C. Williamson; Alvaro Molina-Cruz; Kairong Cui; Carolina Barillas-Mury; Keji Zhao; Xin-Zhuan Su

BackgroundIt has been shown that nearly a quarter of the initial predicted gene models in the Plasmodium falciparum genome contain errors. Although there have been efforts to obtain complete cDNA sequences to correct the errors, the coverage of cDNA sequences on the predicted genes is still incomplete, and many gene models for those expressed in sexual or mosquito stages have not been validated. Antisense transcripts have widely been reported in P. falciparum; however, the extent and pattern of antisense transcripts in different developmental stages remain largely unknown.ResultsWe have sequenced seven bidirectional libraries from ring, early and late trophozoite, schizont, gametocyte II, gametocyte V, and ookinete, and four strand-specific libraries from late trophozoite, schizont, gametocyte II, and gametocyte V of the 3D7 parasites. Alignment of the cDNA sequences to the 3D7 reference genome revealed stage-specific antisense transcripts and novel intron-exon splicing junctions. Sequencing of strand-specific cDNA libraries suggested that more genes are expressed in one direction in gametocyte than in schizont. Alternatively spliced genes, antisense transcripts, and stage-specific expressed genes were also characterized.ConclusionsIt is necessary to continue to sequence cDNA from different developmental stages, particularly those of non-erythrocytic stages. The presence of antisense transcripts in some gametocyte and ookinete genes suggests that these antisense RNA may play an important role in gene expression regulation and parasite development. Future gene expression studies should make use of directional cDNA libraries. Antisense transcripts may partly explain the observed discrepancy between levels of mRNA and protein expression.


Journal of Clinical Investigation | 2013

Probiotic/prebiotic supplementation of antiretrovirals improves gastrointestinal immunity in SIV-infected macaques

Nichole R. Klatt; Xiaoyong Sun; Carol L. Vinton; Nicholas T. Funderburg; David R. Morcock; Mariam Quiñones; Clayton Deming; Molly R. Perkins; Daria J. Hazuda; Michael D. Miller; Michael M. Lederman; Julie Segre; Jeffrey D. Lifson; Elias K. Haddad; Jacob D. Estes; Jason M. Brenchley

HIV infection results in gastrointestinal (GI) tract damage, microbial translocation, and immune activation, which are not completely ameliorated with suppression of viremia by antiretroviral (ARV) therapy. Furthermore, increased morbidity and mortality of ARV-treated HIV-infected individuals is associated with these dysfunctions. Thus, to enhance GI tract physiology, we treated SIV-infected pigtail macaques with ARVs, probiotics, and prebiotics or with ARVs alone. This synbiotic treatment resulted in increased frequency and functionality of GI tract APCs, enhanced reconstitution and functionality of CD4+ T cells, and reduced fibrosis of lymphoid follicles in the colon. Thus, ARV synbiotic supplementation in HIV-infected individuals may improve GI tract immunity and thereby mitigate inflammatory sequelae, ultimately improving prognosis.


Cell | 2015

Microbiota-Dependent Sequelae of Acute Infection Compromise Tissue-Specific Immunity

Denise Morais da Fonseca; Timothy W. Hand; Seong-Ji Han; Michael Y. Gerner; Arielle Glatman Zaretsky; Allyson L. Byrd; Oliver J. Harrison; Alexandra M. Ortiz; Mariam Quiñones; Giorgio Trinchieri; Jason M. Brenchley; Igor E. Brodsky; Ronald N. Germain; Gwendalyn J. Randolph; Yasmine Belkaid

Infections have been proposed as initiating factors for inflammatory disorders; however, identifying associations between defined infectious agents and the initiation of chronic disease has remained elusive. Here, we report that a single acute infection can have dramatic and long-term consequences for tissue-specific immunity. Following clearance of Yersinia pseudotuberculosis, sustained inflammation and associated lymphatic leakage in the mesenteric adipose tissue deviates migratory dendritic cells to the adipose compartment, thereby preventing their accumulation in the mesenteric lymph node. As a consequence, canonical mucosal immune functions, including tolerance and protective immunity, are persistently compromised. Post-resolution of infection, signals derived from the microbiota maintain inflammatory mesentery remodeling and consequently, transient ablation of the microbiota restores mucosal immunity. Our results indicate that persistent disruption of communication between tissues and the immune system following clearance of an acute infection represents an inflection point beyond which tissue homeostasis and immunity is compromised for the long-term. VIDEO ABSTRACT.


Cell Host & Microbe | 2013

Intraluminal containment of commensal outgrowth in the gut during infection-induced dysbiosis.

Michael J. Molloy; John Grainger; Nicolas Bouladoux; Timothy W. Hand; Lily Koo; Shruti Naik; Mariam Quiñones; Amiran K. Dzutsev; Ji Liang Gao; Giorgio Trinchieri; Philip M. Murphy; Yasmine Belkaid

Shifts in commensal microbiota composition are emerging as a hallmark of gastrointestinal inflammation. In particular, outgrowth of γ-proteobacteria has been linked to the etiology of inflammatory bowel disease and the pathologic consequences of infections. Here we show that following acute Toxoplasma gondii gastrointestinal infection of mice, control of commensal outgrowth is a highly coordinated process involving both the host response and microbial signals. Notably, neutrophil emigration to the intestinal lumen results in the generation of organized intraluminal structures that encapsulate commensals and limit their contact with the epithelium. Formation of these luminal casts depends on the high-affinity N-formyl peptide receptor, Fpr1. Consequently, after infection, mice deficient in Fpr1 display increased microbial translocation, poor commensal containment, and increased mortality. Altogether, our study describes a mechanism by which the host rapidly contains commensal pathobiont outgrowth during infection. Further, these results reveal Fpr1 as a major mediator of host commensal interaction during dysbiosis.


Mucosal Immunology | 2015

Dysbiotic bacteria translocate in progressive SIV infection

Zachary Klase; Alexandra M. Ortiz; Claire Deleage; Joseph C. Mudd; Mariam Quiñones; Elias Schwartzman; Nichole R. Klatt; Jacob D. Estes; Jason M. Brenchley

Infection of gut-resident CD4+ memory T cells during acute human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infection is associated with rapid loss of these cells and damage to the epithelial barrier. Damage to the epithelial barrier allows translocation of microbial products from the intestinal lumen into the body. Immune activation caused by these microbial products has been associated with disease progression. Although microbial translocation has been demonstrated in SIV-infected nonhuman primates, the identity of translocating bacteria has not been determined. In this study we examined the communities of bacteria both within the gastrointestinal (GI) tract and systemic tissues of both healthy and experimentally SIV-infected Asian macaques. Although there were only modest changes in the GI tract-associated microbiome resulting from infection, there is substantial dysbiosis after administration of antiretrovirals. Analysis of bacterial DNA isolated from tissues of infected animals revealed a preference for the phylum Proteobacteria, suggesting that they preferentially translocate. Consistent with this finding, we observed increased metabolic activity of Proteobacterial species within the colonic lumen of SIV-infected animals. Overall, these data provide insights into disease progression and suggest that therapies aimed at altering the composition and metabolic activity of the GI tract microbiome could benefit chronically HIV-infected individuals, particularly those on antiretroviral therapies.


BMC Medicine | 2010

The normal breast microenvironment of premenopausal women differentially influences the behavior of breast cancer cells in vitro and in vivo

Jodie M. Fleming; Tyler C. Miller; Mariam Quiñones; Zhen G Xiao; Xia Xu; Matthew J. Meyer; Erika Ginsburg; Timothy D. Veenstra; Barbara K. Vonderhaar

BackgroundBreast cancer studies frequently focus on the role of the tumor microenvironment in the promotion of cancer; however, the influence of the normal breast microenvironment on cancer cells remains relatively unknown. To investigate the role of the normal breast microenvironment on breast cancer cell tumorigenicity, we examined whether extracellular matrix molecules (ECM) derived from premenopausal African-American (AA) or Caucasian-American (CAU) breast tissue would affect the tumorigenicity of cancer cells in vitro and in vivo. We chose these two populations because of the well documented predisposition of AA women to develop aggressive, highly metastatic breast cancer compared to CAU women.MethodsThe effects of primary breast fibroblasts on tumorigenicity were analyzed via real-time PCR arrays and mouse xenograft models. Whole breast ECM was isolated, analyzed via zymography, and its effects on breast cancer cell aggressiveness were tested in vitro via soft agar and invasion assays, and in vivo via xenograft models. Breast ECM and hormone metabolites were analyzed via mass spectrometry.ResultsMouse mammary glands humanized with premenopausal CAU fibroblasts and injected with primary breast cancer cells developed significantly larger tumors compared to AA humanized glands. Examination of 164 ECM molecules and cytokines from CAU-derived fibroblasts demonstrated a differentially regulated set of ECM proteins and increased cytokine expression. Whole breast ECM was isolated; invasion and soft agar assays demonstrated that estrogen receptor (ER)-, progesterone receptor (PR)/PR- cells were significantly more aggressive when in contact with AA ECM, as were ER+/PR+ cells with CAU ECM. Using zymography, protease activity was comparatively upregulated in CAU ECM. In xenograft models, CAU ECM significantly increased the tumorigenicity of ER+/PR+ cells and enhanced metastases. Mass spectrometry analysis of ECM proteins showed that only 1,759 of approximately 8,000 identified were in common. In the AA dataset, proteins associated with breast cancer were primarily related to tumorigenesis/neoplasia, while CAU unique proteins were involved with growth/metastasis. Using a novel mass spectrometry method, 17 biologically active hormones were measured; estradiol, estriol and 2-methoxyestrone were significantly higher in CAU breast tissue.ConclusionsThis study details normal premenopausal breast tissue composition, delineates potential mechanisms for breast cancer development, and provides data for further investigation into the role of the microenvironment in cancer disparities.


PeerJ | 2014

Unipro UGENE NGS pipelines and components for variant calling, RNA-seq and ChIP-seq data analyses

Olga Golosova; Ross Henderson; Yuriy Vaskin; Andrei Gabrielian; German Grekhov; Vijayaraj Nagarajan; Andrew J. Oler; Mariam Quiñones; Darrell E. Hurt; Mikhail Fursov; Yentram Huyen

The advent of Next Generation Sequencing (NGS) technologies has opened new possibilities for researchers. However, the more biology becomes a data-intensive field, the more biologists have to learn how to process and analyze NGS data with complex computational tools. Even with the availability of common pipeline specifications, it is often a time-consuming and cumbersome task for a bench scientist to install and configure the pipeline tools. We believe that a unified, desktop and biologist-friendly front end to NGS data analysis tools will substantially improve productivity in this field. Here we present NGS pipelines “Variant Calling with SAMtools”, “Tuxedo Pipeline for RNA-seq Data Analysis” and “Cistrome Pipeline for ChIP-seq Data Analysis” integrated into the Unipro UGENE desktop toolkit. We describe the available UGENE infrastructure that helps researchers run these pipelines on different datasets, store and investigate the results and re-run the pipelines with the same parameters. These pipeline tools are included in the UGENE NGS package. Individual blocks of these pipelines are also available for expert users to create their own advanced workflows.


BMC Genomics | 2014

In depth annotation of the Anopheles gambiae mosquito midgut transcriptome

Alejandro Padrón; Alvaro Molina-Cruz; Mariam Quiñones; José M. C. Ribeiro; Urvashi N. Ramphul; Janneth Rodrigues; Kui Shen; Ashley Haile; Jose Luis Ramirez; Carolina Barillas-Mury

BackgroundGenome sequencing of Anopheles gambiae was completed more than ten years ago and has accelerated research on malaria transmission. However, annotation needs to be refined and verified experimentally, as most predicted transcripts have been identified by comparative analysis with genomes from other species. The mosquito midgut—the first organ to interact with Plasmodium parasites—mounts effective antiplasmodial responses that limit parasite survival and disease transmission. High-throughput Illumina sequencing of the midgut transcriptome was used to identify new genes and transcripts, contributing to the refinement of An. gambiae genome annotation.ResultsWe sequenced ~223 million reads from An. gambiae midgut cDNA libraries generated from susceptible (G3) and refractory (L35) mosquito strains. Mosquitoes were infected with either Plasmodium berghei or Plasmodium falciparum, and midguts were collected after the first or second Plasmodium infection. In total, 22,889 unique midgut transcript models were generated from both An. gambiae strain sequences combined, and 76% are potentially novel. Of these novel transcripts, 49.5% aligned with annotated genes and appear to be isoforms or pre-mRNAs of reference transcripts, while 50.5% mapped to regions between annotated genes and represent novel intergenic transcripts (NITs). Predicted models were validated for midgut expression using qRT-PCR and microarray analysis, and novel isoforms were confirmed by sequencing predicted intron-exon boundaries. Coding potential analysis revealed that 43% of total midgut transcripts appear to be long non-coding RNA (lncRNA), and functional annotation of NITs showed that 68% had no homology to current databases from other species. Reads were also analyzed using de novo assembly and predicted transcripts compared with genome mapping-based models. Finally, variant analysis of G3 and L35 midgut transcripts detected 160,742 variants with respect to the An. gambiae PEST genome, and 74% were new variants. Intergenic transcripts had a higher frequency of variation compared with non-intergenic transcripts.ConclusionThis in-depth Illumina sequencing and assembly of the An. gambiae midgut transcriptome doubled the number of known transcripts and tripled the number of variants known in this mosquito species. It also revealed existence of a large number of lncRNA and opens new possibilities for investigating the biological function of many newly discovered transcripts.

Collaboration


Dive into the Mariam Quiñones's collaboration.

Top Co-Authors

Avatar

Yasmine Belkaid

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Nicolas Bouladoux

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Oler

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Clayton Deming

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Giorgio Trinchieri

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jason M. Brenchley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Julia A. Segre

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shruti Naik

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Darrell E. Hurt

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lily Koo

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge