Network


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

Hotspot


Dive into the research topics where Jerod A. Skyberg is active.

Publication


Featured researches published by Jerod A. Skyberg.


Frontiers of Biology in China | 2013

Progress in Brucella vaccine development

Xinghong Yang; Jerod A. Skyberg; Ling Cao; Beata Clapp; Theresa Thornburg; David W. Pascual

Brucella spp. are zoonotic, facultative intracellular pathogens, which cause animal and human disease. Animal disease results in abortion of fetuses; in humans, it manifests flu-like symptoms with an undulant fever, with osteoarthritis as a common complication of infection. Antibiotic regimens for human brucellosis patients may last several months and are not always completely effective. While there are no vaccines for humans, several licensed live Brucella vaccines are available for use in livestock. The performance of these animal vaccines is dependent upon the host species, dose, and route of immunization. Newly engineered live vaccines, lacking well-defined virulence factors, retain low residual virulence, are highly protective, and may someday replace currently used animal vaccines. These also have possible human applications. Moreover, due to their enhanced safety and efficacy in animal models, subunit vaccines for brucellosis show great promise for their application in livestock and humans. This review summarizes the progress of brucellosis vaccine development and presents an overview of candidate vaccines.


Infection and Immunity | 2011

Protective live oral brucellosis vaccines stimulate Th1 and th17 cell responses.

Beata Clapp; Jerod A. Skyberg; Xinghong Yang; Theresa Thornburg; Nancy Walters; David W. Pascual

ABSTRACT Zoonotic transmission of brucellosis often results from exposure to Brucella-infected livestock, feral animals, or wildlife or frequently via consumption of unpasteurized milk products or raw meat. Since natural infection of humans often occurs by the oral route, mucosal vaccination may offer a means to confer protection for both mucosal and systemic tissues. Significant efforts have focused on developing a live brucellosis vaccine, and deletion of the znuA gene involved in zinc transport has been found to attenuate Brucella abortus. A similar mutation has been adapted for Brucella melitensis and tested to determine whether oral administration of ΔznuA B. melitensis can confer protection against nasal B. melitensis challenge. A single oral vaccination with ΔznuA B. melitensis rapidly cleared from mice within 2 weeks and effectively protected mice upon nasal challenge with wild-type B. melitensis 16M. In 83% of the vaccinated mice, no detectable brucellae were found in their spleens, unlike with phosphate-buffered saline (PBS)-dosed mice, and vaccination also enhanced the clearance of brucellae from the lungs. Moreover, vaccinated gamma interferon-deficient (IFN-γ−/−) mice also showed protection in both spleens and lungs, albeit protection that was not as effective as in immunocompetent mice. Although IFN-γ, interleukin 17 (IL-17), and IL-22 were stimulated by these live vaccines, only RB51-mediated protection was codependent upon IL-17 in BALB/c mice. These data suggest that oral immunization with the live, attenuated ΔznuA B. melitensis vaccine provides an attractive strategy to protect against inhalational infection with virulent B. melitensis.


PLOS ONE | 2011

Murine and Bovine γδ T Cells Enhance Innate Immunity against Brucella abortus Infections

Jerod A. Skyberg; Theresa Thornburg; MaryClare F. Rollins; Eduardo Huarte; Mark A. Jutila; David W. Pascual

γδ T cells have been postulated to act as a first line of defense against infectious agents, particularly intracellular pathogens, representing an important link between the innate and adaptive immune responses. Human γδ T cells expand in the blood of brucellosis patients and are active against Brucella in vitro. However, the role of γδ T cells in vivo during experimental brucellosis has not been studied. Here we report TCRδ−/− mice are more susceptible to B. abortus infection than C57BL/6 mice at one week post-infection as measured by splenic colonization and splenomegaly. An increase in TCRγδ cells was observed in the spleens of B. abortus-infected C57BL/6 mice, which peaked at two weeks post-infection and occurred concomitantly with diminished brucellae. γδ T cells were the major source of IL-17 following infection and also produced IFN-γ. Depletion of γδ T cells from C57BL/6, IL-17Rα−/−, and GMCSF−/− mice enhanced susceptibility to B. abortus infection although this susceptibility was unaltered in the mutant mice; however, when γδ T cells were depleted from IFN-γ−/− mice, enhanced susceptibility was observed. Neutralization of γδ T cells in the absence of TNF-α did not further impair immunity. In the absence of TNF-α or γδ T cells, B. abortus-infected mice showed enhanced IFN-γ, suggesting that they augmented production to compensate for the loss of γδ T cells and/or TNF-α. While the protective role of γδ T cells was TNF-α-dependent, γδ T cells were not the major source of TNF-α and activation of γδ T cells following B. abortus infection was TNF-α-independent. Additionally, bovine TCRγδ cells were found to respond rapidly to B. abortus infection upon co-culture with autologous macrophages and could impair the intramacrophage replication of B. abortus via IFN-γ. Collectively, these results demonstrate γδ T cells are important for early protection to B. abortus infections.


BMC Microbiology | 2008

Mutational and transcriptional analyses of an avian pathogenic Escherichia coli ColV plasmid

Jerod A. Skyberg; Timothy J. Johnson; Lisa K. Nolan

BackgroundPreviously we described a 184-kb ColV plasmid, pAPEC-O2-ColV, that contributed to the ability of an E. coli to kill avian embryos, grow in human urine, and colonize the murine kidney. Here, the roles of several genes encoded by this plasmid in virulence were assessed using mutational and transcriptional analyses.MethodsGenes chosen for deletion were iss, tsh, iutA, iroN, sitA, and cvaB. In addition, a 35-kb region of the plasmid, containing iss, tsh, and the ColV and iro operons, along with a 15-kb region containing both the aerobactin and sit operons, were deleted. Mutants were compared to the wild-type (APEC O2) for lethality to chick embryos and growth in human urine. Expression of the targeted genes was also assessed under these same conditions using RT-PCRResultsNo significant differences between the mutants and the wild-type in these phenotypic traits were detected. However, genes encoding known or predicted iron transport systems were up-regulated during growth in human urine, as compared to growth in LB broth, while iss, hlyF, and iroN were strongly up-regulated in chick embryos.ConclusionWhile no difference was observed between the mutant strains and their wild-type parent in the phenotypic traits assayed, we reasoned that some compensatory virulence mechanism, insensitivity of the virulence assays, or other factor could have obscured changes in the virulence of the mutants. Indeed we found several of these genes to be up-regulated in human urine and/or in the chick embryo, suggesting that certain genes linked to ColV plasmids are involved in the establishment of avian extraintestinal infection.


Journal of Leukocyte Biology | 2011

Apple polyphenols require T cells to ameliorate dextran sulfate sodium-induced colitis and dampen proinflammatory cytokine expression

Jerod A. Skyberg; Amy Robison; Sarah Golden; MaryClare F. Rollins; Gayle Callis; Eduardo Huarte; Irina Kochetkova; Mark A. Jutila; David W. Pascual

Human IBD, including UC and Crohnˈs disease, is characterized by a chronic, relapsing, and remitting condition that exhibits various features of immunological inflammation and affects at least one/1000 people in Western countries. Polyphenol extracts from a variety of plants have been shown to have immunomodulatory and anti‐inflammatory effects. In this study, treatment with APP was investigated to ameliorate chemically induced colitis. Oral but not peritoneal administration of APP during colitis induction significantly protected C57BL/6 mice against disease, as evidenced by the lack of weight loss, colonic inflammation, and shortening of the colon. APP administration dampened the mRNA expression of IL‐1β, TNF‐α, IL‐6, IL‐17, IL‐22, CXCL9, CXCL10, CXCL11, and IFN‐γ in the colons of mice with colitis. APP‐mediated protection requires T cells, as protection was abated in Rag‐1−/− or TCRα−/− mice but not in IL‐10−/−, IRF‐1−/−, μMT, or TCRδ−/− mice. Administration of APP during colitis to TCRα−/− mice actually enhanced proinflammatory cytokine expression, further demonstrating a requirement for TCRαβ cells in APP‐mediated protection. APP treatment also inhibited CXCR3 expression by TCRαβ cells, but not B or NK cells, in the colons of mice with colitis; however, depletion of CD4+ or CD8+ T cells alone did not abolish APP‐mediated protection. Collectively, these results show that oral administration of APP protects against experimental colitis and diminishes proinflammatory cytokine expression via T cells.


PLOS Pathogens | 2012

Nasal Acai polysaccharides potentiate innate immunity to protect against pulmonary Francisella tularensis and Burkholderia pseudomallei Infections.

Jerod A. Skyberg; MaryClare F. Rollins; Jeff Holderness; Nicole L. Marlenee; Igor A. Schepetkin; Andrew Goodyear; Steven W. Dow; Mark A. Jutila; David W. Pascual

Pulmonary Francisella tularensis and Burkholderia pseudomallei infections are highly lethal in untreated patients, and current antibiotic regimens are not always effective. Activating the innate immune system provides an alternative means of treating infection and can also complement antibiotic therapies. Several natural agonists were screened for their ability to enhance host resistance to infection, and polysaccharides derived from the Acai berry (Acai PS) were found to have potent abilities as an immunotherapeutic to treat F. tularensis and B. pseudomallei infections. In vitro, Acai PS impaired replication of Francisella in primary human macrophages co-cultured with autologous NK cells via augmentation of NK cell IFN-γ. Furthermore, Acai PS administered nasally before or after infection protected mice against type A F. tularensis aerosol challenge with survival rates up to 80%, and protection was still observed, albeit reduced, when mice were treated two days post-infection. Nasal Acai PS administration augmented intracellular expression of IFN-γ by NK cells in the lungs of F. tularensis-infected mice, and neutralization of IFN-γ ablated the protective effect of Acai PS. Likewise, nasal Acai PS treatment conferred protection against pulmonary infection with B. pseudomallei strain 1026b. Acai PS dramatically reduced the replication of B. pseudomallei in the lung and blocked bacterial dissemination to the spleen and liver. Nasal administration of Acai PS enhanced IFN-γ responses by NK and γδ T cells in the lungs, while neutralization of IFN-γ totally abrogated the protective effect of Acai PS against pulmonary B. pseudomallei infection. Collectively, these results demonstrate Acai PS is a potent innate immune agonist that can resolve F. tularensis and B. pseudomallei infections, suggesting this innate immune agonist has broad-spectrum activity against virulent intracellular pathogens.


Journal of Leukocyte Biology | 2012

IFN-γ-deficient mice develop IL-1-dependent cutaneous and musculoskeletal inflammation during experimental brucellosis

Jerod A. Skyberg; Theresa Thornburg; Irina Kochetkova; William Layton; Gayle Callis; MaryClare F. Rollins; Carol Riccardi; Todd Becker; Sarah Golden; David W. Pascual

Human brucellosis exhibits diverse pathological manifestations that can affect almost any organ. In particular, osteoarticular complications are the most common focal manifestation of brucellosis and occur in 40–80% of patients. In immunocompetent mice, Brucella replication is generally restricted to the spleen, liver, and to a lesser extent, LNs, thereby limiting their use for study of focal inflammation often found in brucellosis. Here, we report that nasal, oral, or peritoneal infection of IFN‐γ−/− mice with WT Brucella melitensis or Brucella abortus results in joint and periarticular tissue inflammation. Histological analysis of the affected joints revealed inflammatory infiltrates and debris within the joint space colocalizing with Brucella antigen. Osteoarthritis, necrosis, periarticular soft tissue inflammation, and substantial brucellae burdens were observed. Oral rifampicin was effective in clearing infection and halting further progression of focal inflammation from infected IFN‐γ−/− mice, although some symptoms and swelling remained. Elevated IL‐1β, but not TNF‐α, IL‐6, or IL‐17, was detected in joint homogenates from infected IFN‐γ−/− mice. Whereas more susceptible to systemic infection, IL‐1R−/− mice depleted of IFN‐γ were more resistant to focal inflammation than WT mice similarly depleted of IFN‐γ. Collectively, these results show IFN‐γ−/− mice represent a potential model for study of focal inflammation attributed to Brucella infection and will allow evaluation of intervention strategies targeting IL‐1, IL‐1R, or other inflammatory mediators, with the potential to complement antibiotic‐based therapies.


Journal of Autoimmunity | 2011

Tolerogen-induced interferon-producing killer dendritic cells (IKDCs) protect against EAE

Eduardo Huarte; Agnieszka Rynda-Apple; Carol Riccardi; Jerod A. Skyberg; Sarah Golden; MaryClare F. Rollins; Andrew Ramstead; Larissa Jackiw; Massimo Maddaloni; David W. Pascual

Natural killer (NK) cells and dendritic cells (DCs) have been shown to link the innate and adaptive immune systems. Likewise, a new innate cell subset, interferon-producing killer DCs (IKDCs), shares phenotypic and functional characteristics with both DCs and NK cells. Here, we show IKDCs play an essential role in the resolution of experimental autoimmune encephalomyelitis (EAE) upon treatment with the tolerizing agent, myelin oligodendrocyte glycoprotein (MOG), genetically fused to reovirus protein σ1 (termed MOG-pσ1). Activated IKDCs were recruited subsequent MOG-pσ1 treatment of EAE, and disease resolution was abated upon NK1.1 cell depletion. These IKDCs were able to kill activated CD4(+) T cells and mature dendritic DCs, thus, contributing to EAE remission. In addition, IKDCs were responsible for MOG-pσ1-mediated MOG-specific regulatory T cell recruitment to the CNS. The IKDCs induced by MOG-pσ1 expressed elevated levels of HVEM for interactions with cognate ligand-positive cells: LIGHT(+) NK and T(eff) cells and BTLA(+) B cells. Further characterization revealed these activated IKDCs being MHC class II(high), and upon their adoptive transfer (CD11c(+)NK1.1(+)MHC class II(high)), IKDCs, but not CD11c(+)NK1.1(+)MHC class II(intermediate/low) (unactivated) cells, conferred protection against EAE. These activated IKDCs showed enhanced CD107a, PD-L1, and granzyme B expression and could present OVA, unlike unactivated IKDCs. Thus, these results demonstrate the interventional potency induced HVEM(+) IKDCs to resolve autoimmune disease.


European Journal of Immunology | 2011

Active immunization using a single dose immunotherapeutic abates established EAE via IL-10 and regulatory T cells

Agnieszka Rynda-Apple; Eduardo Huarte; Massimo Maddaloni; Gayle Callis; Jerod A. Skyberg; David W. Pascual

Stimulation of Ag‐specific inducible Treg can enhance resolution of autoimmune disease. Conventional methods to induce Treg often require induction of autoimmune disease or subjection to infection. Reovirus adhesin, protein σ1 (pσ1), can successfully facilitate tolerance when fused to a tolerogen. We tested whether myelin oligodendrocyte glycoprotein (MOG) fused to pσ1 (MOG‐pσ1) can stimulate Ag‐specific Treg. We show that C57BL/6 mice treated nasally with MOG‐pσ1 fail to induce MOG‐specific Abs and delayed‐type hypersensitivity (DTH) responses and resist EAE. Such resistance was attributed to stimulation of Foxp3+ Treg, as well as Th2 cells. MOG‐pσ1s protective capacity was abrogated in IL‐10−/− mice, but restored when adoptively transferred with MOG‐pσ1‐induced Treg. As a therapeutic, MOG‐pσ1 diminished EAE within 24 h of nasal application, unlike recombinant MOG (rMOG), pσ1, or pσ1+rMOG, implicating the importance of Ag specificity by pσ1‐based therapeutics. MOG‐pσ1‐treated mice showed elevated IL‐4, IL‐10, and IL‐28 production by CD4+ T cells, unlike rMOG treated or control mice that produced elevated IFN‐γ or IL‐17, respectively. These data show the feasibility of using pσ1 as a tolerogen platform for Ag‐specific tolerance induction and highlight its potential use as an immunotherapeutic for autoimmunity.


Journal of Leukocyte Biology | 2017

Hematopoietic MyD88 and IL-18 are essential for IFN-γ–dependent restriction of type A Francisella tularensis infection

Jerod A. Skyberg; Carolyn A. Lacey

Francisella tularensis is a highly infectious intracellular bacterium that causes the potentially fatal disease tularemia. We used mice with conditional MyD88 deficiencies to investigate cellular and molecular mechanisms by which MyD88 restricts type A F. tularensis infection. F. tularensis–induced weight loss was predominately dependent on MyD88 signaling in nonhematopoietic cells. In contrast, MyD88 signaling in hematopoietic cells, but not in myeloid and dendritic cells, was essential for control of F. tularensis infection in tissue. Myeloid and dendritic cell MyD88 deficiency also did not markedly impair cytokine production during infection. Although the production of IL‐12 or ‐18 was not significantly reduced in hematopoietic MyD88‐deficient mice, IFN‐γ production was abolished in these animals. In addition, neutralization studies revealed that control of F. tularensis infection mediated by hematopoietic MyD88 was entirely dependent on IFN‐γ. Although IL‐18 production was not significantly affected by MyD88 deficiency, IL‐18 was essential for IFN‐γ production and restricted bacterial replication in an IFN‐γ–dependent manner. Caspase‐1 was also found to be partially necessary for the production of IL‐18 and IFN‐γ and for control of F. tularensis replication. Our collective data show that the response of leukocytes to caspase‐1–dependent IL‐18 via MyD88 is critical, whereas MyD88 signaling in myeloid and dendritic cells is dispensable for IFN‐γ–dependent control of type A F. tularensis infection.

Collaboration


Dive into the Jerod A. Skyberg's collaboration.

Top Co-Authors

Avatar

David W. Pascual

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark A. Jutila

Montana State University

View shared research outputs
Top Co-Authors

Avatar

Eduardo Huarte

Montana State University

View shared research outputs
Top Co-Authors

Avatar

Gayle Callis

Montana State University

View shared research outputs
Top Co-Authors

Avatar

Sarah Golden

Montana State University

View shared research outputs
Top Co-Authors

Avatar

Carol Riccardi

Montana State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge