Network


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

Hotspot


Dive into the research topics where Edwin V. Oaks is active.

Publication


Featured researches published by Edwin V. Oaks.


Infection and Immunity | 2006

IpaD Localizes to the Tip of the Type III Secretion System Needle of Shigella flexneri

Marianela Espina; Andrew J. Olive; Roma Kenjale; David S. Moore; S. Fernando Ausar; Robert W. Kaminski; Edwin V. Oaks; C. Russell Middaugh; William D. Picking; Wendy L. Picking

ABSTRACT Shigella flexneri, the causative agent of shigellosis, is a gram-negative bacterial pathogen that initiates infection by invading cells within the colonic epithelium. Contact with host cell surfaces induces a rapid burst of protein secretion via the Shigella type III secretion system (TTSS). The first proteins secreted are IpaD, IpaB, and IpaC, with IpaB and IpaC being inserted into the host cell membrane to form a pore for translocating late effectors into the target cell cytoplasm. The resulting pathogen-host cross talk results in localized actin polymerization, membrane ruffling, and, ultimately, pathogen entry. IpaD is essential for host cell invasion, but its role in this process is just now coming to light. IpaD is a multifunctional protein that controls the secretion and presentation of IpaB and IpaC at the pathogen-host interface. We show here that antibodies recognizing the surface-exposed N terminus of IpaD neutralize Shigellas ability to promote pore formation in erythrocyte membranes. We further show that MxiH and IpaD colocalize on the bacterial surface. When TTSS needles were sheared from the Shigella surface, IpaD was found at only the needle tips. Consistent with this, IpaD localized to the exposed tips of needles that were still attached to the bacterium. Molecular analyses then showed that the IpaD C terminus is required for this surface localization and function. Furthermore, mutations that prevent IpaD surface localization also eliminate all IpaD-related functions. Thus, this study demonstrates that IpaD localizes to the TTSA needle tip, where it functions to control the secretion and proper insertion of translocators into host cell membranes.


Infection and Immunity | 2000

Isolation and Characterization of a Shigella flexneri Invasin Complex Subunit Vaccine

K. Ross Turbyfill; Antoinette B. Hartman; Edwin V. Oaks

ABSTRACT The invasiveness and virulence of Shigella spp. are largely due to the expression of plasmid-encoded virulence factors, among which are the invasion plasmid antigens (Ipa proteins). After infection, the host immune response is directed primarily against lipopolysaccharide (LPS) and the virulence proteins (IpaB, IpaC, and IpaD). Recent observations have indicated that the Ipa proteins (IpaB, IpaC, and possibly IpaD) form a multiprotein complex capable of inducing the phagocytic event which internalizes the bacterium. We have isolated a complex of invasins and LPS from water-extractable antigens of virulent shigellae by ion-exchange chromatography. Western blot analysis of the complex indicates that all of the major virulence antigens of Shigella, including IpaB, IpaC, and IpaD, and LPS are components of this macromolecular complex. Mice or guinea pigs immunized intranasally with purified invasin complex (invaplex), without any additional adjuvant, mounted a significant immunoglobulin G (IgG) and IgA antibody response against theShigella virulence antigens and LPS. The virulence-specific response was very similar to that previously noted in primates infected with shigellae. Guinea pigs (keratoconjunctivitis model) or mice (lethal lung model) immunized intranasally on days 0, 14, and 28 and challenged 3 weeks later with virulent shigellae were protected from disease (P < 0.01 for both animal models).


Expert Review of Vaccines | 2009

Inactivated and subunit vaccines to prevent shigellosis.

Robert W. Kaminski; Edwin V. Oaks

Shigellosis remains a formidable disease globally, with children of the developing world bearing the greatest number of infections. The need for an affordable, safe and efficacious vaccine has persisted for decades. Vaccines to prevent shigellosis can be divided into living and nonliving approaches. Several nonliving Shigella vaccines are currently at different stages of development and show substantial promise. Outlined here is an overview of multiple nonliving vaccine technologies, highlighting their current status and recent advances in testing. In addition, gaps in the knowledge base regarding immune mechanisms of protection are explored.


Vaccine | 2011

Safety and immunogenicity of an intranasal Shigella flexneri 2a Invaplex 50 vaccine

Mark S. Riddle; Robert W. Kaminski; Carlos Williams; Chad K. Porter; Shahida Baqar; Alexis A. Kordis; Theron Gilliland; Joyce Lapa; Melissa Coughlin; Chris Soltis; Erica Jones; Jackie Saunders; Paul B. Keiser; Ryan T. Ranallo; Robert Gormley; M.R. Nelson; K. Ross Turbyfill; David R. Tribble; Edwin V. Oaks

BACKGROUND Shigella flexneri 2a lipopolysaccharide 50 is a nasally delivered subunit vaccine consisting of a macromolecular complex composed of LPS, IpaB, IpaC and IpaD. The current study examined vaccine safety and immunogenicity across a dose range and the clinical performance of a new intranasal delivery device. METHODS Volunteers (N=36) were randomized to receive vaccine via the Dolphin™ (Valois of America, Congers, New York) intranasal spray device at one of three doses (240, 480, and 690 μg) on days 0, 14, and 28. Another group (N=8) received the 240 μg dose via pipette. Vaccine safety was actively monitored and antigen-specific humoral and mucosal immune responses were determined. RESULTS There were no serious adverse events and the majority of adverse events (98%) were mild. Antibody secreting cells (ASC), plasma, and mucosal immune responses to Shigella antigens were detected at all three dose levels with the 690 μg dose inducing the highest magnitude and frequency of responses. Vaccination with comparable doses of Invaplex 50 via the Dolphin™ resulted in higher plasma and ASC immune responses as compared to pipette delivery. CONCLUSION In this trial the S. flexneri 2a Invaplex 50 vaccine was safe, well-tolerated and induced robust levels of antigen-specific intestinal IgA and ASC responses. The spray device performed well and offered an advantage over pipette intranasal delivery.


Vaccine | 2008

Immunogenicity and efficacy of highly purified invasin complex vaccine from Shigella flexneri 2a

K. Ross Turbyfill; Robert W. Kaminski; Edwin V. Oaks

Development of a subunit vaccine for shigellosis requires identification of protective antigens and delivering these antigens in a manner that stimulates immunity comparable to that induced by natural infection. The Shigella invasin complex (Invaplex) vaccine is an ion-exchange-purified extract from virulent Shigella that consists of LPS and several other proteins, including the invasins IpaB and IpaC. Intranasal delivery of Invaplex stimulates protective immunity in small animal models for shigellosis. To identify the active component(s) of Invaplex responsible for its immunogenicity and efficacy, size-exclusion chromatography (SEC) was used to separate Invaplex into several different fractions. A high-molecular mass complex with a molecular mass between 669 MDa and 2 MDa consisted primarily of LPS, IpaB and IpaC and was considered to be a highly purified (HP) form of Invaplex. Using the mouse lung model to evaluate the immunogenicity and efficacy of the SEC fractions it was clearly demonstrated that the high-molecular mass complex of the invasins and LPS was responsible for the protective capacity of parent native Invaplex. Other smaller mass SEC fractions were mostly non-immunogenic and did not stimulate solid protection. In guinea pigs, the HP Invaplex stimulated an enhanced immune response as compared to the parent Invaplex and was fully protective. Isolation and characterization of the immunogenic and protective moiety within Invaplex will allow better standardization of the Invaplex product and may allow future development of an Invaplex assembled from purified components.


Infection and Immunity | 2006

Mucosal Adjuvant Properties of the Shigella Invasin Complex

Robert W. Kaminski; K. Ross Turbyfill; Edwin V. Oaks

ABSTRACT The Shigella invasin complex (Invaplex) is an effective mucosal vaccine capable of protecting against Shigella challenge in animal models. The major antigenic constituents of Invaplex are the Ipa proteins and lipopolysaccharide. The cell-binding capacity of the Ipa proteins prompted the investigation into the adjuvanticity of Invaplex. Using ovalbumin (OVA) as a model antigen, intranasal immunization with OVA combined with Invaplex was found to enhance anti-OVA serum immunoglobulin G (IgG) and IgA responses and induce OVA-specific mucosal antibody responses at sites located both proximal and distal to the immunization site. The immune responses induced with OVA and Invaplex were comparable in both magnitude and duration to the immune responses induced after immunization with OVA and cholera toxin. The OVA-specific immune response was characterized by high levels of serum IgG1 and increased production of interleukin-4 (IL-4), IL-5, or IL-10 from lymphoid cells of immunized animals, suggesting a Th2 response. In addition to enhancing the immunogenicity of OVA, Invaplex-specific immune responses were also induced, indicating the potential for the development of a combination vaccine consisting of Invaplex and other immunogens. Preexisting Invaplex-specific immunity did not interfere with the capacity to enhance the immunogenicity of a second, unrelated vaccine antigen, suggesting that Invaplex could be used as a mucosal adjuvant in multiple vaccine regimens.


Vaccine | 2010

Safety and immunogenicity of a Shigella flexneri 2a Invaplex 50 intranasal vaccine in adult volunteers.

David R. Tribble; Robert W. Kaminski; J. Cantrell; M.R. Nelson; Chad K. Porter; Shahida Baqar; C. Williams; R. Arora; J. Saunders; M. Ananthakrishnan; J. Sanders; G. Zaucha; R. Turbyfill; Edwin V. Oaks

Shigellosis is a leading cause of diarrhea worldwide prompting vaccine development. The Shigella flexneri Invaplex 50 is a macromolecular complex containing IpaB, IpaC, and LPS, formulated from an aqueous extract of virulent Shigella delivered via nasal administration. Preclinical vaccine testing demonstrated safety, immunogenicity and efficacy. An open-label dose-escalating phase 1 study evaluated a 3-dose (2-week intervals) regimen via nasal pipette delivery. Thirty-two subjects were enrolled into one of four vaccine dose groups (10, 50, 240, or 480 microg). The vaccine was well tolerated with minor short-lived nasal symptoms without evidence of dose effect. Antibody-secreting cell (ASC) responses were elicited at doses > or =50 microg with the highest IgG ASC, Invaplex 50 (100%) and S. flexneri 2a LPS (71%), as well as, serologic responses (43%) occurring with the 240 microg dose. Fecal IgA responses, Invaplex 50 (38.5%) and LPS (30.8%), were observed at doses > or =240 microg. The Invaplex 50 nasal vaccine was safe with encouraging mucosal immune responses. Follow-on studies will optimize dose, delivery mechanism and assess efficacy in a S. flexneri 2a challenge study.


Transactions of The Royal Society of Tropical Medicine and Hygiene | 2013

Surveillance for enteric pathogens in a case-control study of acute diarrhea in Western Kenya

Brett E. Swierczewski; Elizabeth Odundo; Margaret C. Koech; Janet N. Ndonye; Ronald Kirera; Cliff P. Odhiambo; Erick K. Cheruiyot; Max Wu; James Eric Lee; Chunlin Zhang; Edwin V. Oaks

BACKGROUND Acute diarrhea remains a major public health problem in East African nations such as Kenya. Surveillance for a broad range of enteric pathogens is necessary to accurately predict the frequency of pathogens and potential changes in antibiotic resistance patterns. METHOD Stool samples were collected from September 2009 to September 2011; 193 and 239 samples, from age-matched cases and asymptomatic controls, were collected, respectively, from Kericho and Kisumu District Hospitals in western Kenya. Bacterial pathogens were identified by conventional microbiological methods; antibiotic susceptibility of bacterial isolates was ascertained using the MicroScan WalkAway 40 Plus. An enzyme immunoassay kit was used to detect rotavirus, and ova and parasite examination was conducted by microscopy and an enzyme immunoassay. RESULTS Rotavirus (10.2% and 10.5%) and Shigella (11% and 8%) were isolated significantly more often in the cases than the controls from Kericho and Kisumu District Hospitals respectively. The diarrheagenic Escherichia coli, Campylobacter jejuni and Salmonella were found most often in the cases while Giardia lamblia and Entamoeba histolytica/E. dispar were found more often in the controls. Most pathogens were isolated from children under 5 years old. More than 50% of the Shigella, Salmonella and diarrheagenic E. coli isolates were multidrug resistant to ampicillin, tetracycline and trimethoprim/sulfamethoxazole with several enteroaggregative and enterotoxigenic E. coli isolates producing extended-spectrum beta-lactamases. CONCLUSION Accurate epidemiologic information on acute diarrheal illness in Kenya will be critical for augmenting existing diarrhea management policies in terms of treatment and to strengthen future community awareness and health promotion programs.


Clinical and Vaccine Immunology | 2014

Development and preclinical evaluation of a trivalent, formalin-inactivated Shigella whole-cell vaccine.

Robert W. Kaminski; M. Wu; K. R. Turbyfill; K. Clarkson; B. Tai; August L. Bourgeois; L. L. Van De Verg; Richard I. Walker; Edwin V. Oaks

ABSTRACT Studies were undertaken to manufacture a multivalent Shigella inactivated whole-cell vaccine that is safe, effective, and inexpensive. By using several formalin concentrations, temperatures, and incubation periods, an optimized set of inactivation conditions was established for Shigella flexneri 2a, S. sonnei, and S. flexneri 3a to produce inactivated whole cells expressing a full repertoire of Ipa proteins and lipopolysaccharide (LPS). The inactivation conditions selected were treatment with 0.2% formalin (S. flexneri 2a and 3a) or 0.6% formalin (S. sonnei) for 48 h at 25°C. Vaccine formulations prepared under different inactivation conditions, in different doses (10E5, 10E7, and 10E9 cells), and with or without the inclusion of double-mutant heat-labile toxin (dmLT) were evaluated in mice. Two intranasal immunizations with ≥10E7 inactivated whole cells resulted in high levels of anti-Invaplex and moderate levels of LPS-specific IgG and IgA in serum and in lung and intestinal wash samples. Addition of dmLT to the vaccine formulations did not significantly enhance humoral immunogenicity. Minimal humoral responses for IpaB, IpaC, or IpaD were detected after immunization with inactivated whole Shigella cells regardless of the vaccine inactivation conditions. In guinea pigs, monovalent formulations of S. flexneri 2a of 3a or S. sonnei consisting of 10E8, 10E9, or 10E10 cells were protective in a keratoconjunctivitis assay. A trivalent formulation provided protection against all three serotypes (S. flexneri 2a, P = 0.018; S. flexneri 3a, P = 0.04; S. sonnei, P < 0.0001). The inactivated Shigella whole-cell vaccine approach incorporates an uncomplicated manufacturing process that is compatible with multivalency and the future development of a broadly protective Shigella vaccine.


Clinical and Vaccine Immunology | 2009

Mucosal adjuvanticity of a Shigella invasin complex with dna-based vaccines.

Robert W. Kaminski; K. Ross Turbyfill; Chien-Chung Chao; Wei-Mei Ching; Edwin V. Oaks

ABSTRACT Protection against many infectious diseases may require the induction of cell-mediated and mucosal immunity. Immunization with plasmid DNA-based vaccines has successfully induced cell-mediated immune responses in small animals but is less potent in humans. Therefore, several methods are under investigation to augment DNA vaccine immunogenicity. In the current study, a mucosal adjuvant consisting of an invasin protein-lipopolysaccharide complex (Invaplex) isolated from Shigella spp. was evaluated as an adjuvant for DNA-based vaccines. Coadministration of plasmid DNA encoding the Orientia tsutsugamushi r56Karp protein with Invaplex resulted in enhanced cellular and humoral responses in intranasally immunized mice compared to immunization with DNA without adjuvant. Mucosal immunoglobulin A, directed to plasmid-encoded antigen, was detected in lung and intestinal compartments after Invaplex-DNA immunization followed by a protein booster. Moreover, immunization with Invaplex elicited Shigella-specific immune responses, highlighting its potential use in a combination vaccine strategy. The capacity of Invaplex to enhance the immunogenicity of plasmid-encoded genes suggested that Invaplex promoted the uptake and expression of the delivered genes. To better understand the native biological activities of Invaplex related to its adjuvanticity, interactions between Invaplex and mammalian cells were characterized. Invaplex rapidly bound to and was internalized by nonphagocytic, eukaryotic cells in an endocytic process dependent on actin polymerization and independent of microtubule formation. Invaplex also mediated transfection with several plasmid DNA constructs, which could be inhibited with monoclonal antibodies specific for IpaB and IpaC or Invaplex-specific polyclonal sera. The cellular binding and transport capabilities of Invaplex likely contribute to the adjuvanticity and immunogenicity of Invaplex.

Collaboration


Dive into the Edwin V. Oaks's collaboration.

Top Co-Authors

Avatar

Robert W. Kaminski

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Charles K. Stover

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Antoinette B. Hartman

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

K. Ross Turbyfill

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Kevin Ross Turbyfill

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Malabi M. Venkatesan

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Brett E. Swierczewski

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Kristen A. Clarkson

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Wendy L. Picking

University of Texas at Austin

View shared research outputs
Researchain Logo
Decentralizing Knowledge