Network


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

Hotspot


Dive into the research topics where Junko Yano is active.

Publication


Featured researches published by Junko Yano.


Infection and Immunity | 2002

Vaginal and Oral Epithelial Cell Anti-Candida Activity

Fatema Nomanbhoy; Chad Steele; Junko Yano; Paul L. Fidel

ABSTRACT Candida albicans is the causative agent of acute and recurrent vulvovaginal candidiasis (VVC), a common mucosal infection affecting significant numbers of women in their reproductive years. While any murine host protective role for cell-mediated immunity (CMI), humoral immunity, and innate resistance by neutrophils against the vaginal infection appear negligible, significant in vitro growth inhibition of Candida species by vaginal and oral epithelial cell-enriched cells has been observed. Both oral and vaginal epithelial cell anti-Candida activity has a strict requirement for cell contact to C. albicans with no role for soluble factors, and oral epithelial cells inhibit C. albicans through a cell surface carbohydrate moiety. The present study further evaluated the inhibitory mechanisms by murine vaginal epithelial cells and the fate of C. albicans by oral and vaginal epithelial cells. Similar to human oral cells, anti-Candida activity produced by murine vaginal epithelial cells is unaffected by enzymatic cleavage of cell surface proteins and lipids but sensitive to periodic acid cleavage of surface carbohydrates. Analysis of specific membrane carbohydrate moieties, however, showed no role for sulfated polysaccharides, sialic acid residues, or glucose and mannose-containing carbohydrates, also similar to oral cells. Staining for live and dead Candida in the coculture with fluorescein diacetate (FDA) and propidium iodide (PI), respectively, showed a clear predominance of live organisms, suggesting a static rather than cidal action. Together, the results suggest that oral and vaginal epithelial cells retard or arrest the growth rather than kill C. albicans through an as-yet-unidentified carbohydrate moiety in a noninflammatory manner.


Infection and Immunity | 2010

Epithelial cell-derived S100 calcium-binding proteins as key mediators in the hallmark acute neutrophil response during Candida vaginitis

Junko Yano; Elizabeth A. Lilly; Melissa M. Barousse; Paul L. Fidel

ABSTRACT Vulvovaginal candidiasis (VVC), caused by Candida species, is a significant problem in women of childbearing age. Similar to clinical observations, a robust vaginal polymorphonuclear neutrophil (PMN) migration occurs in a subset of mice without affecting vaginal fungal burden. We hypothesize that the vaginal PMN infiltrate and accompanying inflammation are not protective but instead are responsible for the symptoms of infection. The purpose of this study was to identify the signal(s) associated with the PMN response in the established mouse model. Vaginal lavage fluid from inoculated mice were categorized base on PMN counts, evaluated for PMN chemotactic activity and analyzed by SDS-PAGE and mass spectrometry (MS) for unique protein identification. The lavage fluid from inoculated mice with high, but not low, PMN levels showed increased chemotactic activity. Likewise, SDS-PAGE of lavage fluid with high PMN levels showed distinct protein patterns. MS revealed that bands at 6 and 14 kDa matched the PMN chemotactic calcium-binding proteins (CBPs), S100A8 and S100A9, respectively. The presence of the CBPs in lavage fluid was confirmed by Western blots and enzyme-linked immunosorbent assay. Vaginal tissues and epithelial cells from inoculated mice with high PMN levels stained more intensely and exhibited increased mRNA transcripts for both proteins compared to those in mice with low PMN levels. Subsequent antibody neutralization showed significant abrogation of the chemotactic activity when the lavage fluid was treated with anti-S100A8, but not anti-S100A9, antibodies. These results reveal that the PMN chemotactic CBP S100A8 and S100A9 are produced by vaginal epithelial cells following interaction with Candida and that S100A8 is a strong candidate responsible for the robust PMN migration during experimental VVC.


Cytokine | 2012

Cytokines in the host response to Candida vaginitis: Identifying a role for non-classical immune mediators, S100 alarmins

Junko Yano; Mairi C. Noverr; Paul L. Fidel

Vulvovaginal candidiasis (VVC), caused by Candida albicans, affects a significant number of women during their reproductive years. More than two decades of research have been focused on the mechanisms associated with susceptibility or resistance to symptomatic infection. Adaptive immunity by Th1-type CD4(+) T cells and downstream cytokine responses are considered the predominant host defense mechanisms against mucosal Candida infections. However, numerous clinical and animal studies have indicated no or limited protective role of cells and cytokines of the Th1 or Th2 lineage against vaginal infection. The role for Th17 is only now begun to be investigated in-depth for VVC with results already showing significant controversy. On the other hand, a clinical live-challenge study and an established animal model have shown that a symptomatic condition is intimately associated with the vaginal infiltration of polymorphonuclear leukocytes (PMNs) but with no effect on vaginal fungal burden. Subsequent studies identified S100A8 and S100A9 alarmins as key chemotactic mediators of the acute PMN response. These chemotactic danger signals appear to be secreted by vaginal epithelial cells upon interaction and early adherence of Candida. Thus, instead of a putative immunodeficiency against Candida involving classical immune cells and cytokines of the adaptive response, the pathological inflammation in VVC is now considered a consequence of a non-productive innate response initiated by non-classical immune mediators.


PLOS ONE | 2012

The Acute Neutrophil Response Mediated by S100 Alarmins during Vaginal Candida Infections Is Independent of the Th17-Pathway

Junko Yano; Jay K. Kolls; Kyle I. Happel; Floyd L. Wormley; Karen L. Wozniak; Paul L. Fidel

Vulvovaginal candidiasis (VVC) caused by Candida albicans affects a significant number of women during their reproductive ages. Clinical observations revealed that a robust vaginal polymorphonuclear neutrophil (PMN) migration occurs in susceptible women, promoting pathological inflammation without affecting fungal burden. Evidence to date in the mouse model suggests that a similar acute PMN migration into the vagina is mediated by chemotactic S100A8 and S100A9 alarmins produced by vaginal epithelial cells in response to Candida. Based on the putative role for the Th17 response in mucosal candidiasis as well as S100 alarmin induction, this study aimed to determine whether the Th17 pathway plays a role in the S100 alarmin-mediated acute inflammation during VVC using the experimental mouse model. For this, IL-23p19−/−, IL-17RA−/− and IL-22−/− mice were intravaginally inoculated with Candida, and vaginal lavage fluids were evaluated for fungal burden, PMN infiltration, the presence of S100 alarmins and inflammatory cytokines and chemokines. Compared to wild-type mice, the cytokine-deficient mice showed comparative levels of vaginal fungal burden and PMN infiltration following inoculation. Likewise, inoculated mice of all strains with substantial PMN infiltration exhibited elevated levels of vaginal S100 alarmins in both vaginal epithelia and secretions in the vaginal lumen. Finally, cytokine analyses of vaginal lavage fluid from inoculated mice revealed equivalent expression profiles irrespective of the Th17 cytokine status or PMN response. These data suggest that the vaginal S100 alarmin response to Candida does not require the cells or cytokines of the Th17 lineage, and therefore, the immunopathogenic inflammatory response during VVC occurs independently of the Th17-pathway.


Mbio | 2015

Transcriptomic Analysis of Vulvovaginal Candidiasis Identifies a Role for the NLRP3 Inflammasome

Vincent M. Bruno; Amol C. Shetty; Junko Yano; Paul L. Fidel; Mairi C. Noverr; Brian M. Peters

ABSTRACT Treatment of vulvovaginal candidiasis (VVC), caused most frequently by Candida albicans, represents a significant unmet clinical need. C. albicans, as both a commensal and a pathogenic organism, has a complex and poorly understood interaction with the vaginal environment. Understanding the complex nature of this relationship is necessary for the development of desperately needed therapies to treat symptomatic infection. Using transcriptome sequencing (RNA-seq), we characterized the early murine vaginal and fungal transcriptomes of the organism during VVC. Network analysis of host genes that were differentially expressed between infected and naive mice predicted the activation or repression of several signaling pathways that have not been previously associated with VVC, including NLRP3 inflammasome activation. Intravaginal challenge of Nlrp3−/− mice with C. albicans demonstrated severely reduced levels of polymorphonuclear leukocytes (PMNs), alarmins, and inflammatory cytokines, including interleukin-1β (IL-1β) (the hallmarks of VVC immunopathogenesis) in vaginal lavage fluid. Intravaginal administration of wild-type (WT) mice with glyburide, a potent inhibitor of the NLRP3 inflammasome, reduced PMN infiltration and IL-1β to levels comparable to those observed in Nlrp3−/− mice. Furthermore, RNA-seq analysis of C. albicans genes indicated robust expression of hypha-associated secreted aspartyl proteinases 4, 5, and 6 (SAP4–6), which are known inflammasome activators. Despite colonization similar to that of the WT strain, ΔSAP4–6 triple and ΔSAP5 single mutants induced significantly less PMN influx and IL-1β during intravaginal challenge. Our findings demonstrate a novel role for the inflammasome in the immunopathogenesis of VVC and implicate the hypha-associated SAPs as major C. albicans virulence determinants during vulvovaginal candidiasis. IMPORTANCE Vaginitis, most commonly caused by the fungus Candida albicans, results in significant quality-of-life issues for all women of reproductive age. Recent efforts have suggested that vaginitis results from an immunopathological response governed by host innate immunity, although an explanatory mechanism has remained undefined. Using comprehensive genomic, immunological, and pharmacological approaches, we have elucidated the NLRP3 inflammasome as a crucial molecular mechanism contributing to host immunopathology. We have also demonstrated that C. albicans hypha-associated secreted aspartyl proteinases (SAP4–6 and SAP5, more specifically) contribute to disease immunopathology. Ultimately, this study enhances our understanding of the complex interplay between host and fungus at the vaginal mucosa and provides proof-of-principle evidence for therapeutic targeting of inflammasomes for symptomatic vulvovaginal candidiasis. Vaginitis, most commonly caused by the fungus Candida albicans, results in significant quality-of-life issues for all women of reproductive age. Recent efforts have suggested that vaginitis results from an immunopathological response governed by host innate immunity, although an explanatory mechanism has remained undefined. Using comprehensive genomic, immunological, and pharmacological approaches, we have elucidated the NLRP3 inflammasome as a crucial molecular mechanism contributing to host immunopathology. We have also demonstrated that C. albicans hypha-associated secreted aspartyl proteinases (SAP4–6 and SAP5, more specifically) contribute to disease immunopathology. Ultimately, this study enhances our understanding of the complex interplay between host and fungus at the vaginal mucosa and provides proof-of-principle evidence for therapeutic targeting of inflammasomes for symptomatic vulvovaginal candidiasis.


PLOS Pathogens | 2014

Candida Vaginitis: When Opportunism Knocks, the Host Responds

Brian M. Peters; Junko Yano; Mairi C. Noverr; Paul L. Fidel

Candida albicans, an opportunistic polymorphic fungus and resident of the normal vaginal microbiota, is the leading causative agent of vulvovaginal candidiasis (VVC) and presents major quality of life issues for women worldwide [1]. Candida vaginitis is characterized by itching, burning, pain, and redness of the vulva and vaginal mucosa and often accompanied by vaginal discharge. Predisposing factors for primary VVC include high-estrogen oral contraceptive use, hormone replacement therapy, antibiotic usage, and underlying diabetes mellitus. It is estimated that 75% of all women of childbearing age will be afflicted by VVC at least once in their lifetime [2]. Of these, approximately 5–8% (approximately 150 million worldwide) suffer from recurrent VVC (RVVC), resulting in idiopathic chronic episodes of vaginal irritation that require antifungal maintenance therapy (e.g., azoles) to partially control symptoms [1]. Although these treatments are typically effective at reducing organism burden and symptoms, the static function of azole activity and fungal recalcitrance to clearance are key factors resulting in recurrence [3]. It is proposed that RVVC and VVC both involve similar immunopathologies but that the triggers occur with greater sensitivity in individuals with RVVC. Continuously rising vaginitis-related healthcare costs are estimated at


Infection and Immunity | 2014

Vaginal Epithelial Cell-Derived S100 Alarmins Induced by Candida albicans via Pattern Recognition Receptor Interactions Are Sufficient but Not Necessary for the Acute Neutrophil Response during Experimental Vaginal Candidiasis

Junko Yano; Glen E. Palmer; Karen E. Eberle; Brian M. Peters; Thomas Vogl; Andrew N. J. McKenzie; Paul L. Fidel

1.8 billion annually in the United States alone [4]. These unsustainable costs further necessitate a comprehensive understanding of vaginitis and the host and fungal factors that contribute to its immunopathology. The Role of Host Immunity in Candida Vaginitis: Historical and Contemporary Perspectives Susceptibility to oral, chronic mucocutaneous, and gastrointestinal candidiasis has been clearly linked to deficiencies in cell-mediated immunity (CMI) [5]. Therefore, susceptibility to Candida vaginitis was also long believed to result from defects in the adaptive immune response. However, numerous clinical studies examining women with RVVC and the use of an experimental mouse model to evaluate roles for CMI or humoral immunity (HI) revealed no appreciable protection provided by local or systemic adaptive immune mechanisms [6], [7], [8]. In support of these findings, relatively high production of immunoregulatory factors (e.g., TGF-β, T-regs, and Υ/δ T-cells) in the vagina may partly explain the lack of functional local CMI [9], [10]. Despite a lack of supportive evidence for a role of adaptive immunity in vaginitis, the newly characterized Th17 axis of CMI, which links innate and adaptive immune responses, has been shown to be critical for local protection against oropharyngeal candidiasis (OPC) [11]. Accordingly, animal models were used to determine its potential role in mucosal immunity during vaginitis. However, discrepant findings amongst mouse models have led to contradictory conclusions: one study using a less stringent pharmacologic approach to Th17 blockade demonstrated a modest role for Th17 involvement [12], while a more rigorous approach using Th17 axis-knockout mice showed no such function [12], [13]. Thus, the role of Th17 responses during vaginitis remains unresolved and lacks any supportive clinical evidence. As for mucosal HI, some animal models have demonstrated modest antibody-mediated protection against vaginitis [14]. It is conceivable that protective human antibodies do exist but occur naturally at concentrations in vaginal secretions too low to sufficiently mediate protection. While exhaustive efforts have found no major role for adaptive immunity in susceptibility to vaginitis, recent studies have identified the importance of innate immunity in regulating vaginitis symptomatology. A paramount study using women volunteers challenged with live C. albicans determined that vaginitis symptoms were associated with polymorphonuclear leukocyte (PMN) recruitment into the vagina and that organism burden alone was not predictive of disease [15]. Moreover, depletion of PMNs in mice did not result in increased fungal load but did decrease histological evidence of vaginal inflammation [16], [17]. Most recently, a family of calcium-binding proteins termed S100A8 and S100A9 “alarmins” have been implicated in the innate vaginal epithelial cell response to C. albicans (see Figure 1) [18]. Because these proteins have vigorous PMN chemotactic activity, it was hypothesized that epithelial expression of S100s may play a key role in controlling PMN migration into the vaginal lumen. However, while this was confirmed, studies using mice lacking expression of S100A8/9 proteins determined that these factors were sufficient but not necessary for driving the PMN response [19]. Collectively, these exciting new studies highlight the immunopathological response as a crucial element of vaginitis pathogenesis. Future clinical studies, however, are required to confirm the presence and function of alarmins during human infection. Figure 1 Working model of the immunopathogenesis of C. albicans vaginitis. As alluded to above, resultant findings from animal models must be translatable to the human host. One important point to consider is that laboratory rodents, unlike humans, do not naturally harbor C. albicans as commensal organisms. Although the estrogen-dependent mouse model of vaginitis closely mimics clinical infection, observed antifungal immune responses are considered primary and may be exaggerated as compared to human infection, in which repeated exposure, immunotolerance, or higher signaling thresholds to Candida may be encountered. Despite this shortcoming, the mouse model of vaginitis has been an indispensible tool for dissecting the immunological mechanisms associated with this highly complex disease.


Journal of Visualized Experiments | 2011

Protocols for vaginal inoculation and sample collection in the experimental mouse model of Candida vaginitis.

Junko Yano; Paul L. Fidel

ABSTRACT Vulvovaginal candidiasis (VVC), caused by Candida albicans, affects women worldwide. Animal and clinical studies suggest that the immunopathogenic inflammatory condition of VVC is initiated by S100 alarmins in response to C. albicans, which stimulate polymorphonuclear neutrophil (PMN) migration to the vagina. The purpose of this study was to extend previous in vitro data and determine the requirement for the alarmin S100A8 in the PMN response and to evaluate pattern recognition receptors (PRRs) that initiate the response. For the former, PMN migration was evaluated in vitro or in vivo in the presence or absence of S100 alarmins initiated by several approaches. For the latter, vaginal epithelial cells were evaluated for PRR expression and C. albicans-induced S100A8 and S100A9 mRNAs, followed by evaluation of the PMN response in inoculated PRR-deficient mice. Results revealed that, consistent with previously reported in vitro data, eukaryote-derived S100A8, but not prokaryote-derived recombinant S100A8, induced significant PMN chemotaxis in vivo. Conversely, a lack of biologically active S100A8 alarmin, achieved by antibody neutralization or by using S100A9−/− mice, had no effect on the PMN response in vivo. In PRR analyses, whereas Toll-like receptor 4 (TLR4)- and SIGNR1-deficient vaginal epithelial cells showed a dramatic reduction in C. albicans-induced S100A8/S100A9 mRNAs in vitro, inoculated mice deficient in these PRRs showed PMN migration similar to that in wild-type controls. These results suggest that S100A8 alarmin is sufficient, but not necessary, to induce PMN migration during VVC and that the vaginal PMN response to C. albicans involves PRRs in addition to SIGNR1 and TLR4, or other induction pathways.


Molecular Oral Microbiology | 2010

Annexin-A1 identified as the oral epithelial cell anti-Candida effector moiety

Elizabeth A. Lilly; Junko Yano; Paul L. Fidel

Vulvovaginal candidiasis (VVC), caused by Candida species, is a fungal infection of the lower female genital tract that affects approximately 75% of otherwise healthy women during their reproductive years. Predisposing factors include antibiotic usage, uncontrolled diabetes and disturbance in reproductive hormone levels due to pregnancy, oral contraceptives or hormone replacement therapies. Recurrent VVC (RVVC), defined as three or more episodes per year, affects a separate 5 to 8% of women with no predisposing factors. An experimental mouse model of VVC has been established and used to study the pathogenesis and mucosal host response to Candida. This model has also been employed to test potential antifungal therapies in vivo. The model requires that the animals be maintained in a state of pseudoestrus for optimal Candida colonization/infection. Under such conditions, inoculated animals will have detectable vaginal fungal burden for weeks to months. Past studies show an extremely high parallel between the animal model and human infection relative to immunological and physiological properties. Differences, however, include a lack of Candida as normal vaginal flora and a neutral vaginal pH in the mice. Here, we demonstrate a series of key methods in the mouse vaginitis model that include vaginal inoculation, rapid collection of vaginal specimens, assessment of vaginal fungal burden, and tissue preparations for cellular extraction/isolation. This is followed by representative results for constituents of vaginal lavage fluid, fungal burden, and draining lymph node leukocyte yields. With the use of anesthetics, lavage samples can be collected at multiple time points on the same mice for longitudinal evaluation of infection/colonization. Furthermore, this model requires no immunosuppressive agents to initiate infection, allowing immunological studies under defined host conditions. Finally, the model and each technique introduced here could potentially give rise to use of the methodologies to examine other infectious diseases of the lower female genital tract (bacterial, parasitic, viral) and respective local or systemic host defenses.


Microbiology | 2014

Characterization of IL-22 and antimicrobial peptide production in mice protected against pulmonary Cryptococcus neoformans infection

Karen L. Wozniak; Camaron R. Hole; Junko Yano; Paul L. Fidel; Floyd L. Wormley

Innate and adaptive immunity are considered critical to protection against mucosal candidal infections. Among innate anti-Candida mechanisms, oral and vaginal epithelial cells have antifungal activity. The mechanism is fungistatic, acid-labile and includes a requirement for cell contact by intact, but not necessarily live, epithelial cells. The purpose of this study was to use the acid-labile property to further characterize the effector moiety. Surface material extracted from phosphate-buffered saline (PBS) -treated, but not acid-treated, epithelial cells significantly inhibited the growth of Candida blastoconidia in a dose-dependent manner which was abrogated by prior heat and protease treatment. Proteins extracted from PBS-treated cells bound blastoconidia and hyphae more intensely than those from acid-treated cells. Proteins from PBS-treated cells eluted from Candida revealed two unique bands of approximately 33 and 45 kDa compared with acid-treated cells. Mass spectrometry identified these proteins as Annexin-A1 and actin, respectively. Oral epithelial cells stained positive for Annexin-A1, but not actin. Western blots showed reduced Annexin-A1 in proteins from acid-treated epithelial cells compared with those from PBS-treated epithelial cells. Lastly, it was demonstrated that immunoprecipitation of Annexin-A1 from proteins extracted from PBS-treated oral epithelial cells resulted in abrogation of inhibitory activity. Taken together, these results indicate that Annexin-A1 is a strong candidate for the epithelial cell anti-Candida effector protein.

Collaboration


Dive into the Junko Yano's collaboration.

Top Co-Authors

Avatar

Brian M. Peters

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chad Steele

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Floyd L. Wormley

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Karen L. Wozniak

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Thomas Vogl

University of Münster

View shared research outputs
Top Co-Authors

Avatar

Alika Yu

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Camaron R. Hole

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge