Network


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

Hotspot


Dive into the research topics where Rhiannon R. Penkert is active.

Publication


Featured researches published by Rhiannon R. Penkert.


Clinical and Vaccine Immunology | 2015

Safety and Immunogenicity of an Intranasal Sendai Virus-Based Human Parainfluenza Virus Type 1 Vaccine in 3- to 6-Year-Old Children

Elisabeth E. Adderson; Kristen Branum; Robert Sealy; Bart G. Jones; Sherri Surman; Rhiannon R. Penkert; Pamela Freiden; Karen S. Slobod; Aditya H. Gaur; Randall T. Hayden; Kim Allison; Nanna Howlett; Jill Utech; Jim Allay; James Knight; Susan Sleep; Michael Meagher; Charles J. Russell; Allen Portner; Julia L. Hurwitz

ABSTRACT Human parainfluenza virus type 1 (hPIV-1) is the most common cause of laryngotracheobronchitis (croup), resulting in tens of thousands of hospitalizations each year in the United States alone. No licensed vaccine is yet available. We have developed murine PIV-1 (Sendai virus [SeV]) as a live Jennerian vaccine for hPIV-1. Here, we describe vaccine testing in healthy 3- to 6-year-old hPIV-1-seropositive children in a dose escalation study. One dose of the vaccine (5 × 105, 5 × 106, or 5 × 107 50% egg infectious doses) was delivered by the intranasal route to each study participant. The vaccine was well tolerated by all the study participants. There was no sign of vaccine virus replication in the airway in any participant. Most children exhibited an increase in antibody binding and neutralizing responses toward hPIV-1 within 4 weeks from the time of vaccination. In several children, antibody responses remained above incoming levels for at least 6 months after vaccination. Data suggest that SeV may provide a benefit to 3- to 6-year-old children, even when vaccine recipients have preexisting cross-reactive antibodies due to previous exposures to hPIV-1. Results encourage the testing of SeV administration in young seronegative children to protect against the serious respiratory tract diseases caused by hPIV-1 infections.


Clinical and Vaccine Immunology | 2016

Vitamin Supplementation at the Time of Immunization with a Cold-Adapted Influenza Virus Vaccine Corrects Poor Mucosal Antibody Responses in Mice Deficient for Vitamins A and D

Sherri Surman; Rhiannon R. Penkert; Bart G. Jones; Robert Sealy; Julia L. Hurwitz

ABSTRACT Vitamin A and D deficiencies and insufficiencies are prevalent worldwide in developed and developing countries. Vitamin metabolites are functionally intertwined in that they are high-affinity ligands for related receptors of the nuclear receptor superfamily. The effects of vitamin A deficiencies (VAD) on antibody responses to respiratory virus vaccines have already been demonstrated. Of particular concern was the reduction in IgA, a first line of defense against pathogens in the respiratory tract. Here, we describe the individual and combined effects of vitamin A and D deficiencies in mice immunized with an attenuated influenza virus vaccine. Relative to VAD, vitamin D deficiency (VDD) had a limited effect, but double deficiencies for vitamins A and D (VAD+VDD) further reduced antibody responses in the respiratory tract. The administration of supplemental vitamins A and D to VAD+VDD mice at the time of vaccination restored responses in a dose-dependent manner. Results suggest that vitamin supplementation programs may be beneficial in a clinical setting to promote healthy immune responses to respiratory virus vaccines in vitamin-deficient individuals.


Experimental Biology and Medicine | 2016

Original Research: Parvovirus B19 infection in children with sickle cell disease in the hydroxyurea era

Jane S. Hankins; Rhiannon R. Penkert; Paul T. Lavoie; Li Tang; Yilun Sun; Julia L. Hurwitz

Parvovirus B19 infection causes transient aplastic crisis in sickle cell disease (SCD) due to a temporary interruption in the red blood cell production. Toxicity from hydroxyurea includes anemia and reticulocytopenia, both of which also occur during a transient aplastic crisis event. Hydroxyurea inhibits proliferation of hematopoietic cells and may be immunosuppressive. We postulated that hydroxyurea could exacerbate parvovirus B19-induced aplastic crisis and inhibit the development of specific immune responses in children with SCD. We conducted a retrospective review of parvovirus B19 infection in 330 children with SCD. Altogether there were 120 known cases of aplastic crisis attributed to parvovirus B19 infection, and 12% of children were on hydroxyurea treatment during the episode. We evaluated hematological and immune responses. Children with HbSS or HbSβ0-thalassemia treated with hydroxyurea, when compared with untreated children, required fewer transfusions and had higher Hb concentration nadir during transient aplastic crisis. Duration of hospital stays was no different between hydroxyurea-treated and untreated groups. Children tested within a week following aplastic crisis were positive for parvovirus-specific IgG. Immune responses lasted for the duration of the observation period, up to 13 years after transient aplastic crisis, and there were no repeat aplastic crisis episodes. The frequencies of parvovirus-specific antibodies in all children with SCD increased with age, as expected due to the increased likelihood of a parvovirus exposure, and were comparable to frequencies reported for healthy children. Approximately one-third of children had a positive parvovirus B19-specific IgG test without a documented history of transient aplastic crisis, and 64% of them were treated with hydroxyurea. Hydroxyurea may reduce requirements for blood transfusions and may attenuate symptoms during transient aplastic crisis episodes caused by parvovirus B19 infections. Children with SCD, whether treated or untreated with hydroxyurea, generate sustained and protective parvovirus B19-specific immune responses.


Viral Immunology | 2016

Hotspots for Vitamin-Steroid-Thyroid Hormone Response Elements Within Switch Regions of Immunoglobulin Heavy Chain Loci Predict a Direct Influence of Vitamins and Hormones on B Cell Class Switch Recombination.

Julia L. Hurwitz; Rhiannon R. Penkert; Beisi Xu; Yiping Fan; Janet F. Partridge; Robert W. Maul; Patricia J. Gearhart

Vitamin A deficiencies are common throughout the world and have a significant negative influence on immune protection against viral infections. Mouse models demonstrate that the production of IgA, a first line of defense against viruses at mucosal sites, is inhibited in the context of vitamin A deficiency. In vitro, the addition of vitamin A to activated B cells can enhance IgA expression, but downregulate IgE. Previous reports have demonstrated that vitamin A modifies cytokine patterns, and in so doing may influence antibody isotype expression by an indirect mechanism. However, we have now discovered hundreds of potential response elements among Sμ, Sɛ, and Sα switch sites within immunoglobulin heavy chain loci. These hotspots appear in both mouse and human loci and include targets for vitamin receptors and related proteins (e.g., estrogen receptors) in the nuclear receptor superfamily. Full response elements with direct repeats are relatively infrequent or absent in Sγ regions although half-sites are present. Based on these results, we pose a hypothesis that nuclear receptors have a direct effect on the immunoglobulin heavy chain class switch recombination event. We propose that vitamin A may alter S site accessibility to activation-induced deaminase and nonhomologous end-joining machinery, thereby influencing the isotype switch, antibody production, and protection against viral infections at mucosal sites.


Cytokine | 2017

Vitamin A differentially regulates cytokine expression in respiratory epithelial and macrophage cell lines.

Rhiannon R. Penkert; Bart G. Jones; Hans Häcker; Janet F. Partridge; Julia L. Hurwitz

HIGHLIGHTSRetinoic acid (RA) decreased IL‐6 production in LETs (an epithelial cell line).RA increased IL‐6 production in MACs (a macrophage cell line).Transcript levels for IL‐6, MCP‐1, GMCSF, and IL‐10 were increased only in MACs.In contrast, transcript levels for RAR&bgr; were increased only in LETs.The cytokine outcomes of RA signaling are clearly dependent on cell target. ABSTRACT Vitamin A is an essential nutrient for the protection of children from respiratory tract disease. Supplementation with vitamin A is frequently prescribed in the clinical setting, in part to combat deficiencies among children in developing countries, and in part to treat respiratory infections in clinical trials. This vitamin influences immune responses via multiple, and sometimes seemingly contradictory mechanisms. For example, in separate reports, vitamin A was shown to decrease Th17 T‐cell activity by downregulating IL‐6, and to promote B cell production of IgA by upregulating IL‐6. To explain these apparent contradictions, we evaluated the effects of retinoic acid (RA), a key metabolite of vitamin A, on cell lines of respiratory tract epithelial cells (LETs) and macrophages (MACs). When triggered with LPS or Sendai virus, a mouse respiratory pathogen, these two cell lines experienced opposing influences of RA on IL‐6. Both IL‐6 protein production and transcript levels were downregulated by RA in LETs, but upregulated in MACs. RA also increased transcript levels of MCP‐1, GMCSF, and IL‐10 in MACs, but not in LETs. Conversely, when LETs, but not MACs, were exposed to RA, there was an increase in transcripts for RAR&bgr;, an RA receptor with known inhibitory effects on cell metabolism. Results help explain past discrepancies in the literature by demonstrating that the effects of RA are cell target dependent, and suggest close attention be paid to cell‐specific effects in clinical trials involving vitamin A supplements.


International Immunology | 2016

Vitamin A deficient mice exhibit increased viral antigens and enhanced cytokine/chemokine production in nasal tissues following respiratory virus infection despite the presence of FoxP3+ T cells.

Rhiannon R. Penkert; Sherri Surman; Bart G. Jones; Robert Sealy; Peter Vogel; Geoffrey Neale; Julia L. Hurwitz

The World Health Organization (WHO) estimates that 250 million children under the age of five suffer from vitamin A deficiencies (VAD). Individuals with VAD experience higher rates of mortality and increased morbidity during enteric and respiratory infections compared with those who are vitamin A sufficient. Previously, our laboratory has demonstrated that VAD mice have significantly impaired virus-specific IgA and CD8(+) T-cell responses in the airways. Here, we demonstrate that VAD mice experience enhanced cytokine/chemokine gene expression and release in the respiratory tract 10 days following virus infection compared with control vitamin A sufficient animals. Cytokines/chemokines that are reproducibly up-regulated at the gene expression and protein levels include IFNγ and IL-6. Despite previous indications that cytokine dysregulation in VAD animals might reflect low forkhead box P3 (FoxP3)-positive regulatory T-cell frequencies, we found no reduction in FoxP3(+) T cells in VAD respiratory tissues. As an alternative explanation for the high cytokine levels, we found that the extent of virus infection and the persistence of viral antigens were increased on day 10 post-infection in VAD animals compared with controls, and consequently that respiratory tract tissues had an increased potential to activate virus-specific T cells. Results encourage cautious management of viral infections in patients with VAD, as efforts to enhance FoxP3(+) T cell frequencies and quell immune effectors could potentially exacerbate disease if the virus has not been cleared.


Vaccine | 2017

Prevnar-13 vaccine failure in a mouse model for vitamin A deficiency

Rhiannon R. Penkert; Amy R. Iverson; Jason W. Rosch; Julia L. Hurwitz

Streptococcus pneumoniae (pneumococcus) is responsible for serious pediatric respiratory infections, and kills close to one million children under the age of five each year. Unfortunately, the Prevnar-13 vaccine (PCV-13) that is used to protect children from the serious consequences of pneumococcus infections is not always successful. Given that vitamin A deficiency (VAD) is known to affect children in both developed and developing countries, we asked if VAD could be responsible, at least in part, for PCV-13 vaccine failures. In a mouse model for VAD, we found that PCV-13 failed to elicit binding and neutralizing antibody activities. Unlike vaccinated, vitamin-replete animals, vaccinated VAD animals were not protected from lethal pneumococcus infections. Results suggest that children with VAD may be susceptible to serious pneumococcal infections even after having received the PCV-13 vaccine.


The Journal of Pediatrics | 2017

Low Retinol-Binding Protein and Vitamin D Levels Are Associated with Severe Outcomes in Children Hospitalized with Lower Respiratory Tract Infection and Respiratory Syncytial Virus or Human Metapneumovirus Detection

Julia L. Hurwitz; Bart G. Jones; Rhiannon R. Penkert; Shane Gansebom; Yilun Sun; Li Tang; Anna M. Bramley; Seema Jain; Jonathan A. McCullers; Sandra R. Arnold

Retinol binding protein and vitamin D were measured in children aged <5 years hospitalized with lower respiratory tract infection and respiratory syncytial virus and/or human metapneumovirus detections. Low vitamin levels were observed in 50% of the children and were associated with significantly elevated risk of the need for intensive care unit admission and invasive mechanical ventilation.


Haematologica | 2017

Inflammatory molecule reduction with hydroxyurea therapy in children with sickle cell anemia

Rhiannon R. Penkert; Julia L. Hurwitz; Paul G. Thomas; Jason W. Rosch; Jola Dowdy; Yilun Sun; Li Tang; Jane S. Hankins

Children with sickle cell anemia (SCA) suffer from a chronic state of inflammation due to repeated tissue insult from sickling red blood cells. The clinical consequences are severe and involve aberration of inflammatory systems, with associated increased activation of multiple cell types, including


Vaccine | 2017

Saccharomyces cerevisiae-derived virus-like particle parvovirus B19 vaccine elicits binding and neutralizing antibodies in a mouse model for sickle cell disease

Rhiannon R. Penkert; Neal S. Young; Sherri Surman; Robert Sealy; Jason W. Rosch; Philip R. Dormitzer; Ethan C. Settembre; Sumana Chandramouli; Susan Wong; Jane S. Hankins; Julia L. Hurwitz

Parvovirus B19 infections are typically mild in healthy individuals, but can be life threatening in individuals with sickle cell disease (SCD). A Saccharomyces cerevisiae-derived B19 VLP vaccine, now in pre-clinical development, is immunogenic in wild type mice when administered with the adjuvant MF59. Because SCD alters the immune response, we evaluated the efficacy of this vaccine in a mouse model for SCD. Vaccinated mice with SCD demonstrated similar binding and neutralizing antibody responses to those of heterozygous littermate controls following a prime-boost-boost regimen. Due to the lack of a mouse parvovirus B19 challenge model, we employed a natural mouse pathogen, Sendai virus, to evaluate SCD respiratory tract responses to infection. Normal mucosal and systemic antibody responses were observed in these mice. Results demonstrate that mice with SCD can respond to a VLP vaccine and to a respiratory virus challenge, encouraging rapid development of the B19 vaccine for patients with SCD.

Collaboration


Dive into the Rhiannon R. Penkert's collaboration.

Top Co-Authors

Avatar

Julia L. Hurwitz

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Bart G. Jones

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Robert Sealy

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Sherri Surman

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Li Tang

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jane S. Hankins

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jason W. Rosch

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Yilun Sun

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Paul G. Thomas

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Aditya H. Gaur

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge