Network


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

Hotspot


Dive into the research topics where Erica Boldenow is active.

Publication


Featured researches published by Erica Boldenow.


Nature Medicine | 2016

Fetal brain lesions after subcutaneous inoculation of Zika virus in a pregnant nonhuman primate

Kristina M. Adams Waldorf; Jennifer Stencel-Baerenwald; Raj P. Kapur; Colin Studholme; Erica Boldenow; Jay Vornhagen; Audrey Baldessari; Manjiri Dighe; Jeff Thiel; Sean Merillat; Blair Armistead; Jennifer Tisoncik-Go; Richard Green; Michael A. Davis; Elyse C. Dewey; Marian R. Fairgrieve; J. Christopher Gatenby; Todd L. Richards; Gwenn A. Garden; Michael S. Diamond; Sandra E. Juul; Richard Grant; La Rene Kuller; Dennis W. W. Shaw; Jason Ogle; G. Michael Gough; Wonsok Lee; Chris English; Robert F. Hevner; William B. Dobyns

We describe the development of fetal brain lesions after Zika virus (ZIKV) inoculation in a pregnant pigtail macaque. Periventricular lesions developed within 10 d and evolved asymmetrically in the occipital–parietal lobes. Fetal autopsy revealed ZIKV in the brain and significant cerebral white matter hypoplasia, periventricular white matter gliosis, and axonal and ependymal injury. Our observation of ZIKV-associated fetal brain lesions in a nonhuman primate provides a model for therapeutic evaluation.


Mbio | 2016

Bacterial Hyaluronidase Promotes Ascending GBS Infection and Preterm Birth

Jay Vornhagen; Phoenicia Quach; Erica Boldenow; Sean Merillat; Christopher Whidbey; Lisa Y. Ngo; Km Adams Waldorf; Lakshmi Rajagopal

ABSTRACT Preterm birth increases the risk of adverse birth outcomes and is the leading cause of neonatal mortality. A significant cause of preterm birth is in utero infection with vaginal microorganisms. These vaginal microorganisms are often recovered from the amniotic fluid of preterm birth cases. A vaginal microorganism frequently associated with preterm birth is group B streptococcus (GBS), or Streptococcus agalactiae. However, the molecular mechanisms underlying GBS ascension are poorly understood. Here, we describe the role of the GBS hyaluronidase in ascending infection and preterm birth. We show that clinical GBS strains associated with preterm labor or neonatal infections have increased hyaluronidase activity compared to commensal strains obtained from rectovaginal swabs of healthy women. Using a murine model of ascending infection, we show that hyaluronidase activity was associated with increased ascending GBS infection, preterm birth, and fetal demise. Interestingly, hyaluronidase activity reduced uterine inflammation but did not impact placental or fetal inflammation. Our study shows that hyaluronidase activity enables GBS to subvert uterine immune responses, leading to increased rates of ascending infection and preterm birth. These findings have important implications for the development of therapies to prevent in utero infection and preterm birth. IMPORTANCE GBS are a family of bacteria that frequently colonize the vagina of pregnant women. In some cases, GBS ascend from the vagina into the uterine space, leading to fetal injury and preterm birth. Unfortunately, little is known about the mechanisms underlying ascending GBS infection. In this study, we show that a GBS virulence factor, HylB, shows higher activity in strains isolated from cases of preterm birth than those isolates from rectovaginal swabs of healthy women. We discovered that GBS rely on HylB to avoid immune detection in uterine tissue, but not placental tissue, which leads to increased rates of fetal injury and preterm birth. These studies provide novel insight into the underlying mechanisms of ascending infection. GBS are a family of bacteria that frequently colonize the vagina of pregnant women. In some cases, GBS ascend from the vagina into the uterine space, leading to fetal injury and preterm birth. Unfortunately, little is known about the mechanisms underlying ascending GBS infection. In this study, we show that a GBS virulence factor, HylB, shows higher activity in strains isolated from cases of preterm birth than those isolates from rectovaginal swabs of healthy women. We discovered that GBS rely on HylB to avoid immune detection in uterine tissue, but not placental tissue, which leads to increased rates of fetal injury and preterm birth. These studies provide novel insight into the underlying mechanisms of ascending infection.


Nature Medicine | 2018

Congenital Zika virus infection as a silent pathology with loss of neurogenic output in the fetal brain

Kristina M. Adams Waldorf; Branden R. Nelson; Jennifer Stencel-Baerenwald; Colin Studholme; Raj P. Kapur; Blair Armistead; Christie Walker; Sean Merillat; Jay Vornhagen; Jennifer Tisoncik-Go; Audrey Baldessari; Michelle Coleman; Manjiri Dighe; Dennis W. W. Shaw; Justin A. Roby; Veronica Santana-Ufret; Erica Boldenow; Junwei Li; Xiaohu Gao; Michael A. Davis; Jesica Swanstrom; Kara Jensen; Douglas G. Widman; Ralph S. Baric; Joseph T Medwid; Kathryn A Hanley; Jason Ogle; G. Michael Gough; Wonsok Lee; Chris English

Zika virus (ZIKV) is a flavivirus with teratogenic effects on fetal brain, but the spectrum of ZIKV-induced brain injury is unknown, particularly when ultrasound imaging is normal. In a pregnant pigtail macaque (Macaca nemestrina) model of ZIKV infection, we demonstrate that ZIKV-induced injury to fetal brain is substantial, even in the absence of microcephaly, and may be challenging to detect in a clinical setting. A common and subtle injury pattern was identified, including (i) periventricular T2-hyperintense foci and loss of fetal noncortical brain volume, (ii) injury to the ependymal epithelium with underlying gliosis and (iii) loss of late fetal neuronal progenitor cells in the subventricular zone (temporal cortex) and subgranular zone (dentate gyrus, hippocampus) with dysmorphic granule neuron patterning. Attenuation of fetal neurogenic output demonstrates potentially considerable teratogenic effects of congenital ZIKV infection even without microcephaly. Our findings suggest that all children exposed to ZIKV in utero should receive long-term monitoring for neurocognitive deficits, regardless of head size at birth.


Science immunology | 2016

Group B Streptococcus circumvents neutrophils and neutrophil extracellular traps during amniotic cavity invasion and preterm labor

Erica Boldenow; Claire Gendrin; Lisa Ngo; Craig J. Bierle; Jay Vornhagen; Michelle Coleman; Sean Merillat; Blair Armistead; Christopher Whidbey; Varchita Alishetti; Veronica Santana-Ufret; Jason Ogle; Michael Gough; Sengkeo L. Srinouanprachanh; James W. MacDonald; Theo K. Bammler; Aasthaa Bansal; H. Denny Liggitt; Lakshmi Rajagopal; Kristina M. Adams Waldorf

Preterm birth is a leading cause of neonatal morbidity and mortality. Although microbial invasion of the amniotic cavity (MIAC) is associated with the majority of early preterm births, the temporal events that occur during MIAC and preterm labor are not known. Group B Streptococci (GBS) are β-hemolytic, gram-positive bacteria, which commonly colonize the vagina but have been recovered from the amniotic fluid in preterm birth cases. To understand temporal events that occur during MIAC, we utilized a unique chronically catheterized nonhuman primate model that closely emulates human pregnancy. This model allows monitoring of uterine contractions, timing of MIAC and immune responses during pregnancy-associated infections. Here, we show that adverse outcomes such as preterm labor, MIAC, and fetal sepsis were observed more frequently during infection with hemolytic GBS when compared to nonhemolytic GBS. Although MIAC was associated with systematic progression in chorioamnionitis beginning with chorionic vasculitis and progressing to neutrophilic infiltration, the ability of the GBS hemolytic pigment toxin to induce neutrophil cell death and subvert killing by neutrophil extracellular traps (NETs) in placental membranes in vivo facilitated MIAC and fetal injury. Furthermore, compared to maternal neutrophils, fetal neutrophils exhibit decreased neutrophil elastase activity and impaired phagocytic functions to GBS. Collectively, our studies demonstrate how a unique bacterial hemolytic lipid toxin enables GBS to circumvent neutrophils and NETs in placental membranes to induce fetal injury and preterm labor.Group B streptococci overcome neutrophils in placental membranes, inducing fetal injury and preterm labor. NETting group B strep Group B Streptococcus (GBS) infection in pregnant women can lead to preterm birth and fetal injury. Boldenow et al. report that a hemolytic pigment toxin from GBS contributes to these effects by subverting neutrophils and neutrophil extracellular traps (NETs) in placental membranes. They found in a nonhuman primate model that adverse outcomes were more closely associated with hemolytic than with nonhemolytic GBS, and that GBS hemolytic pigment toxin induced cell death in neutrophils and prevented killing by NETs, allowing GBS to invade the amniotic fluid. This toxin therefore could serve as a target to prevent complications from GBS in pregnant women. Preterm birth is a leading cause of neonatal morbidity and mortality. Although microbial invasion of the amniotic cavity (MIAC) is associated with most early preterm births, the temporal events that occur during MIAC and preterm labor are not known. Group B streptococci (GBS) are β-hemolytic, Gram-positive bacteria, which commonly colonize the vagina but have been recovered from the amniotic fluid in preterm birth cases. To understand temporal events that occur during MIAC, we used a chronically catheterized nonhuman primate model that closely emulates human pregnancy. This model allows monitoring of uterine contractions, timing of MIAC, and immune responses during pregnancy-associated infections. We show that adverse outcomes such as preterm labor, MIAC, and fetal sepsis were observed more frequently during infection with hemolytic GBS when compared with nonhemolytic GBS. Although MIAC was associated with systematic progression in chorioamnionitis beginning with chorionic vasculitis and progressing to neutrophilic infiltration, the ability of the GBS hemolytic pigment toxin to induce neutrophil cell death and subvert killing by neutrophil extracellular traps (NETs) in placental membranes in vivo facilitated MIAC and fetal injury. Furthermore, compared with maternal neutrophils, fetal neutrophils exhibit decreased neutrophil elastase activity and impaired phagocytic functions to GBS. Collectively, our studies demonstrate how a bacterial hemolytic lipid toxin enables GBS to circumvent neutrophils and NETs in placental membranes to induce fetal injury and preterm labor.


Science Advances | 2015

Mast cell degranulation by a hemolytic lipid toxin decreases GBS colonization and infection

Claire Gendrin; Jay Vornhagen; Lisa Ngo; Christopher Whidbey; Erica Boldenow; Veronica Santana-Ufret; Morgan Clauson; Kellie Burnside; Dionne P. Galloway; Kristina M. Adams Waldorf; Adrian M. Piliponsky; Lakshmi Rajagopal

GBS hemolytic lipid/pigment and hyperpigmented GBS activate mast cells triggering the release of preformed and proinflammatory mediators. Ascending infection of microbes from the lower genital tract into the amniotic cavity increases the risk of preterm birth, stillbirth, and newborn infections. Host defenses that are critical for preventing ascending microbial infection are not completely understood. Group B Streptococcus (GBS) are Gram-positive bacteria that frequently colonize the lower genital tract of healthy women but cause severe infections during pregnancy, leading to preterm birth, stillbirth, or early-onset newborn infections. We recently described that the GBS pigment is hemolytic, and increased pigment expression promotes GBS penetration of human placenta. Here, we show that the GBS hemolytic pigment/lipid toxin and hyperpigmented GBS strains induce mast cell degranulation, leading to the release of preformed and proinflammatory mediators. Mast cell–deficient mice exhibit enhanced bacterial burden, decreased neutrophil mobilization, and decreased immune responses during systemic GBS infection. In a vaginal colonization model, hyperpigmented GBS strains showed increased persistence in mast cell–deficient mice compared to mast cell–proficient mice. Consistent with these observations, fewer rectovaginal GBS isolates from women in their third trimester of pregnancy were hyperpigmented/hyperhemolytic. Our work represents the first example of a bacterial hemolytic lipid that induces mast cell degranulation and emphasizes the role of mast cells in limiting genital colonization by hyperpigmented GBS.


Infection and Immunity | 2014

Association and Virulence Gene Expression Vary among Serotype III Group B Streptococcus Isolates following Exposure to Decidual and Lung Epithelial Cells

Michelle L. Korir; David Knupp; Kathryn LeMerise; Erica Boldenow; Rita Loch-Caruso; David M. Aronoff; Shannon D. Manning

ABSTRACT Group B Streptococcus (GBS) causes severe disease in neonates, the elderly, and immunocompromised individuals. GBS species are highly diverse and can be classified by serotype and multilocus sequence typing. Sequence type 17 (ST-17) strains cause invasive neonatal disease more frequently than strains of other STs. Attachment and invasion of host cells are key steps in GBS pathogenesis. We investigated whether four serotype III strains representing ST-17 (two strains), ST-19, and ST-23 differ in their abilities to attach to and invade both decidual cells and lung epithelial cells. Virulence gene expression following host cell association and exposure to amnion cells was also tested. The ST-17 strains differed in their abilities to attach to and invade decidual cells, whereas there were no differences with lung epithelial cells. The ST-19 and ST-23 strains, however, attached to and invaded decidual cells less than both ST-17 strains. Although the ST-23 strain attached to lung epithelial cells better than ST-17 and -19 strains, none of the strains effectively invaded the lung epithelial cells. Notably, the association with host cells resulted in the differential expression of several virulence genes relative to basal expression levels. Similar expression patterns of some genes were observed regardless of cell type used. Collectively, these results show that GBS strains differ in their abilities to attach to distinct host cell types and express key virulence genes that are relevant to the disease process. Enhancing our understanding of pathogenic mechanisms could aid in the identification of novel therapeutic targets or vaccine candidates that could potentially decrease morbidity and mortality associated with neonatal infections.


Reproductive Toxicology | 2015

The trichloroethylene metabolite S-(1,2-dichlorovinyl)-l-cysteine but not trichloroacetate inhibits pathogen-stimulated TNF-α in human extraplacental membranes in vitro.

Erica Boldenow; Iman Hassan; Mark C. Chames; Chuanwu Xi; Rita Loch-Caruso

Extraplacental membranes define the gestational compartment and provide a barrier to infectious microorganisms ascending the gravid female reproductive tract. We tested the hypothesis that bioactive metabolites of trichloroethylene (TCE) decrease pathogen-stimulated innate immune response of extraplacental membranes. Extraplacental membranes were cultured for 4, 8, and 24h with the TCE metabolites trichloroacetate (TCA) or S-(1,2-dichlorovinyl)-l-cysteine (DCVC) in the absence or presence of lipoteichoic acid (LTA) or lipopolysaccharide (LPS) to simulate infection. In addition, membranes were cocultured with DCVC and Group B Streptococcus (GBS). DCVC (5-50μM) significantly inhibited LTA-, LPS-, and GBS-stimulated cytokine release from tissue cultures as early as 4h (P≤0.05). In contrast, TCA (up to 500μM) did not inhibit LTA-stimulated cytokine release from tissue punches. Because cytokines are important mediators for host response to infectious microorganisms these findings suggest that TCE exposure could potentially modify susceptibility to infection during pregnancy.


American Journal of Reproductive Immunology | 2015

Role of Cytokine Signaling in Group B Streptococcus‐Stimulated Expression of Human Beta Defensin‐2 in Human Extraplacental Membranes

Erica Boldenow; Kelly A. Hogan; Mark C. Chames; David M. Aronoff; Chuanwu Xi; Rita Loch-Caruso

Group B Streptococcus (GBS) is a leading cause of neonatal morbidity and mortality. We tested the hypothesis that the choriodecidua plays a role in GBS‐stimulated human beta defensin(HBD)‐2 increases in amnion cells through a secreted factor of choriodecidual origin.


Journal of Clinical Investigation | 2018

Group B streptococcus exploits vaginal epithelial exfoliation for ascending infection

Jay Vornhagen; Blair Armistead; Veronica Santana-Ufret; Claire Gendrin; Sean Merillat; Michelle Coleman; Phoenicia Quach; Erica Boldenow; Varchita Alishetti; Christina Leonhard-Melief; Lisa Y. Ngo; Christopher Whidbey; Kelly S. Doran; Chad Curtis; Kristina M. Adams Waldorf; Elizabeth Nance; Lakshmi Rajagopal

Thirteen percent of pregnancies result in preterm birth or stillbirth, accounting for fifteen million preterm births and three and a half million deaths annually. A significant cause of these adverse pregnancy outcomes is in utero infection by vaginal microorganisms. To establish an in utero infection, vaginal microbes enter the uterus by ascending infection; however, the mechanisms by which this occurs are unknown. Using both in vitro and murine models of vaginal colonization and ascending infection, we demonstrate how a vaginal microbe, group B streptococcus (GBS), which is frequently associated with adverse pregnancy outcomes, uses vaginal exfoliation for ascending infection. GBS induces vaginal epithelial exfoliation by activation of integrin and &bgr;-catenin signaling. However, exfoliation did not diminish GBS vaginal colonization as reported for other vaginal microbes. Rather, vaginal exfoliation increased bacterial dissemination and ascending GBS infection, and abrogation of exfoliation reduced ascending infection and improved pregnancy outcomes. Thus, for some vaginal bacteria, exfoliation promotes ascending infection rather than preventing colonization. Our study provides insight into mechanisms of ascending infection by vaginal microbes.


The Journal of Allergy and Clinical Immunology | 2017

Mast cell chymase decreases the severity of group B Streptococcus infections

Claire Gendrin; Nicholas J. Shubin; Erica Boldenow; Sean Merillat; Morgan Clauson; Danial Power; Kelly S. Doran; Magnus Åbrink; Gunnar Pejler; Lakshmi Rajagopal; Adrian M. Piliponsky

Background: Group B Streptococcus (GBS) or Streptococcus agalactiae are &bgr;‐hemolytic gram‐positive bacteria that colonize the lower genital tracts of women and are frequently associated with infections during pregnancy. Innate immune defenses are critical for controlling GBS dissemination and systemic infection. Mast cells are resident sentinel cells that come into contact with pathogens early during colonization and infection. Objective: We aimed to investigate the contribution of chymase to systemic GBS infection and rates of preterm birth. Methods: Pharmacologic and genetic approaches using mice deficient in mast cell protease (MCPT) 4, the mouse functional homologue of human chymase, were used. Results: Our studies show that mast cells release a protease with chymotrypsin‐like cleavage specificity in response to GBS. Additionally, increased GBS systemic infection and preterm births were observed in MCPT4‐deficient mice versus MCPT4‐sufficient mice. Furthermore, we observed that proteolytic cleavage of the host extracellular matrix protein fibronectin by peritoneal cell–derived mast cell lysates diminished GBS adherence. Consistent with this observation, the increase in GBS dissemination and preterm births observed in MCPT4‐deficient mice was abolished when GBS was deficient in expression of the fibronectin‐binding protein SfbA. Conclusions: Taken together, our results suggest that the protective effect of MCPT4 against GBS dissemination and preterm labor can be attributed in part to MCPT4‐mediated proteolysis of fibronectin. Our studies reveal a novel role of mast cells in defense against bacterial infections.

Collaboration


Dive into the Erica Boldenow's collaboration.

Top Co-Authors

Avatar

Sean Merillat

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Jay Vornhagen

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Blair Armistead

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claire Gendrin

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michelle Coleman

Seattle Children's Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge