Network


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

Hotspot


Dive into the research topics where Jenny Katz is active.

Publication


Featured researches published by Jenny Katz.


Molecular Immunology | 2009

Glycogen synthase kinase-3β (GSK3β) inhibition suppresses the inflammatory response to Francisella infection and protects against tularemia in mice.

Ping Zhang; Jenny Katz; Suzanne M. Michalek

Francisella tularensis, the causative agent of tularemia, is currently considered a category A bioterrorism agent due to its high virulence. Infection with F. tularensis results in an inflammatory response that plays an important role in the pathogenesis of the disease; however, the cellular mechanisms regulating this response are poorly understood. Glycogen synthase kinase-3beta (GSK3beta) is a serine/threonine protein kinase that has recently emerged as a key regulatory switch in the modulation of the inflammatory response. In this study, we investigated the effect of GSK3beta inhibition in regulating F. tularensis LVS-induced inflammatory responses. F. tularensis LVS infection of murine peritoneal macrophages induced a TLR2 dependent phosphorylation of GSK3beta. Inhibition of GSK3beta resulted in a significant decrease in the production of pro-inflammatory cytokine IL-6, IL-12p40 and TNF-alpha, as well as a significant increase in the production of the anti-inflammatory cytokine IL-10. GSK3beta regulated the F. tularensis LVS-induced cytokine response by differentially affecting the activation of transcription factors NF-kappaB and CREB. Inhibition of GSK3beta by lithium in vivo suppressed the inflammatory response in mice infected with F. tularensis LVS and conferred a survival advantage. In addition, we show that the production of IFN-gamma contributed to the development of tularemia and to the fatal outcome of the infected animals, depending on the timing and the relative level of the IFN-gamma produced. IFN-gamma potentiated F. tularensis LVS-induced cytokine production by increasing GSK3beta activity and the nuclear translocation of NF-kappaB. Taken together, these results demonstrate a regulatory function of GSK3beta in modulating inflammatory responses that can be detrimental to the host during an F. tularensis LVS infection, and suggest that inhibition of GSK3beta may represent a novel therapeutic approach in the treatment of tularemia.


Journal of Biological Chemistry | 2011

TLR2-dependent modulation of osteoclastogenesis by Porphyromonas gingivalis through differential induction of NFATc1 and NF-κB

Ping Zhang; Jianzhong Liu; Qing-an Xu; Gregory J. Harber; Xu Feng; Suzanne M. Michalek; Jenny Katz

Osteolytic diseases, including rheumatoid arthritis, osteomyelitis, and periodontitis, are usually associated with bacterial infections. However, the precise mechanisms by which bacteria induce bone loss still remain unclear. Evidence exists that Toll-like receptor (TLR) signaling regulates both inflammation and bone metabolism and that the receptor activator of NF-κB ligand (RANKL) and its receptor RANK are the key regulators for bone remodeling and for the activation of osteoclasts. Here, we investigate the direct effects of the periodontal pathogen Porphyromonas gingivalis on osteoclast differentiation and show that P. gingivalis differentially modulates RANKL-induced osteoclast formation contingent on the state of differentiation of osteoclast precursors. In addition, although an optimal induction of cytokines by P. gingivalis is dependent on TLR2 and TLR4, as well as myeloid differentiation factor 88 and Toll/IL-1R domain-containing adaptor-inducing IFN-β, P. gingivalis utilizes TLR2/ myeloid differentiation factor 88 in modulating osteoclast differentiation. P. gingivalis modulates RANKL-induced osteoclast formation by differential induction of NFATc1 and c-Fos. More importantly, RANKL-mediated lineage commitment also has an impact on P. gingivalis-induced cytokine production. RANKL inhibits P. gingivalis-induced cytokine production by down-regulation of TLR/NF-κB and up-regulation of NFATc1. Our findings reveal novel aspects of the interactions between TLR and RANK signaling and provide a new model for understanding the mechanism underlying the pathogenesis of bacteria-mediated bone loss.


Archives of Oral Biology | 1991

Effect of immune bovine milk on Streptococcus mutans in human dental plaque

Steven J. Filler; Richard L. Gregory; Suzanne M. Michalek; Jenny Katz; Jerry R. McGhee

The use of a mouthrinse of bovine milk containing antibodies to Streptococcus mutans resulted in an initial reduction in the numbers of recoverable Strep. mutans in a group of 9 individuals. Ten volunteers who used control bovine milk that contained no antibody activity to Strep. mutans had variable levels of plaque Strep. mutans. In addition, after culture on Mitis Salivarius and Golds agar, the plaque Strep. mutans from subjects who used the immune bovine milk rinse formed smaller colonies than those from pre-treatment plaque and from all plaque samples of subjects who used the control rinse.


Current Topics in Microbiology and Immunology | 1989

Liposomes as Oral Adjuvants

Suzanne M. Michalek; Noel K. Childers; Jenny Katz; Francis R. Denys; A. K. Berry; John H. Eldridge; Jerry R. McGhee; Roy Curtiss

In this brief review, emphasis was placed on the effectiveness of liposomes as carriers/vehicles of soluble antigens and as adjuvants for mucosal responses when used as oral vaccines. Evidence was provided that oral administration of antigen in liposomes resulted in an augmented mucosal response, compared to the response obtained when the oral vaccine consisted of antigen alone. Specific mucosal responses were further enhanced by the use of lipophilic MDP in the antigen/liposome vaccines. In order to better understand the properties of liposomes important for their functional activities, a rapid and reproducible method employing flow cytometry was described which can be conveniently used for the characterization of liposome preparations. Finally, evidence was presented which further supports the potential of recombinant DNA techniques in developing effective and safe oral vaccines against a variety of infectious diseases.


Molecular Immunology | 2009

Requirement of TLR4 and CD14 in dendritic cell activation by Hemagglutinin B from Porphyromonas gingivalis

Dalia E. Gaddis; Suzanne M. Michalek; Jenny Katz

Porphyromonas gingivalis is a Gram-negative anaerobic bacterium that is one of the causative agents of chronic adult periodontal disease. Among the potential virulence factors of P. gingivalis are the hemagglutinins. Recombinant Hemagglutinin B (rHagB) from P. gingivalis has been shown to activate the immune system by inducing specific antibodies that protect against experimental periodontal bone loss following P. gingivalis infection. Since different microbial products can stimulate dendritic cells (DC) through Toll-like receptors (TLRs), subsequently leading to T cell activation and antibody production, we wanted to investigate the immunostimulatory effect of rHagB on DC and the role of TLR signaling in this process. Using an endotoxin free rHagB preparation, our results show that stimulation of murine bone marrow-derived DC with rHagB leads to upregulation of the costimulatory molecules CD86 and CD40, activation of p38 and ERK MAP kinases, transcription factors NF-kappaB, CREB and IRF-3 and the production of IL-6, TNF-alpha, IL-12p40 and to a lesser extent IL-10 and IFN-beta. This activation process was absolutely dependent on TLR4 and CD14. While upregulation of CD86 was independent of the adaptor molecule MyD88, CD40 upregulation and optimal cytokine (IL-6, TNF-alpha, IL-12p40, IL-10 and IFN-beta) production required both MyD88 and TRIF molecules. These results are of importance since they are the first to provide insights into the interaction of rHagB with DC and TLRs. The information from this study will aid in the design of effective vaccines strategies against chronic adult periodontal disease.


Infection and Immunity | 2011

Contribution of a Streptococcus mutans Antigen Expressed by a Salmonella Vector Vaccine in Dendritic Cell Activation

Qing-an Xu; Jenny Katz; Ping Zhang; Amit R. Ashtekar; Dalia E. Gaddis; Mingwen Fan; Suzanne M. Michalek

ABSTRACT A Salmonella vector vaccine expressing the saliva-binding region (SBR) of the adhesin AgI/II of Streptococcus mutans has been shown to induce a mixed Th1/Th2 anti-SBR immune response in mice and to require Toll-like receptor 2 (TLR2), TLR4, and MyD88 signaling for the induction of mucosal anti-SBR antibody responses. Since dendritic cells (DC) are critical in innate and adaptive immunity, the present study assessed the role of SBR expression by the vector vaccine in DC activation. Bone marrow-derived DC from wild-type and TLR2, TLR4, and MyD88 knockout mice were stimulated with Salmonella vector BRD509, the SBR-expressing Salmonella vector vaccine BRD509(pSBRT7), or SBR protein, and the DC responses to different stimuli were compared by assessing costimulatory molecule expression, cytokine production, and signaling pathways. The DC response to both BRD509(pSBRT7) and BRD509 was dependent mainly on TLR4. BRD509(pSBRT7) and BRD509 induced upregulation of CD80, CD86, CD40, and major histocompatibility complex class II (MHC II) expression. Lower levels of interleukin-10 (IL-10) and IL-12p40 were produced by BRD509(pSBRT7)-stimulated DC than by BRD509-stimulated DC. Furthermore, BRD509(pSBRT7)-stimulated DC showed decreased p38 phosphorylation compared to that induced by DC stimulated with BRD509. However, BRD509(pSBRT7)-treated DC produced a higher level of IL-6 than BRD509-stimulated cells. The low IL-12p40 and high IL-6 cytokine profile expressed by BRD509(pSBRT7)-stimulated DC may represent a shift toward a Th2 response, as suggested by the increased expression in Jagged-1. These results provide novel evidence that a heterologous protein expressed by a Salmonella vector vaccine can differentially affect DC activation.


Journal of Biological Chemistry | 2015

IL-1R/TLR2 through MyD88 Divergently Modulates Osteoclastogenesis through Regulation of Nuclear Factor of Activated T Cells c1 (NFATc1) and B Lymphocyte-induced Maturation Protein-1 (Blimp1)

Zhihong Chen; Lingkai Su; Qingan Xu; Jenny Katz; Suzanne M. Michalek; Mingwen Fan; Xu Feng; Ping Zhang

Background: Interleukin-1 and Porphyromonas gingivalis both signal through the IL-1R/TLR superfamily but have distinct effects on osteoclastogenesis. Results: IL-1 and P. gingivalis lipopolysaccharide (LPS-PG) differentially regulate osteoclast genes and osteoclastogenic transcription factor NFATc1, as well as transcription repressor Blimp1 and anti-osteoclastogenic genes. Conclusion: Multiple signaling molecules are involved in distinct IL-1R/TLR2-mediated effects on osteoclastogenesis. Significance: This is a novel mechanism for understanding infection/inflammation-mediated osteoclastogenesis. Toll-like receptors (TLR) and the receptor for interleukin-1 (IL-1R) signaling play an important role in bacteria-mediated bone loss diseases including periodontitis, rheumatoid arthritis, and osteomyelitis. Recent studies have shown that TLR ligands inhibit the receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation from un-committed osteoclast precursors, whereas IL-1 potentiates RANKL-induced osteoclast formation. However, IL-1R and TLR belong to the same IL-1R/TLR superfamily, and activate similar intracellular signaling pathways. Here, we investigate the molecular mechanisms underlying the distinct effects of IL-1 and Porphyromonas gingivalis lipopolysaccharide (LPS-PG) on RANKL-induced osteoclast formation. Our results show that LPS-PG and IL-1 differentially regulate RANKL-induced activation of osteoclast genes encoding Car2, Ctsk, MMP9, and TRAP, as well as expression of NFATc1, a master transcription factor of osteoclastogenesis. Regulation of osteoclast genes and NFATc1 by LPS-PG and IL-1 is dependent on MyD88, an important signaling adaptor for both TLR and IL-1R family members. Furthermore, LPS-PG and IL-1 differentially regulate RANKL-costimulatory receptor OSCAR (osteoclast-associated receptor) expression and Ca2+ oscillations induced by RANKL. Moreover, LPS-PG completely abrogates RANKL-induced gene expression of B lymphocyte-induced maturation protein-1 (Blimp1), a global transcriptional repressor of anti-osteoclastogenic genes encoding Bcl6, IRF8, and MafB. However, IL-1 enhances RANKL-induced blimp1 gene expression but suppresses the gene expression of bcl6, irf8, and mafb. Our study reveals the involvement of multiple signaling molecules in the differential regulation of RANKL-induced osteoclastogenesis by TLR2 and IL-1 signaling. Understanding the signaling cross-talk among TLR, IL-1R, and RANK is critical for identifying therapeutic strategies to control bacteria-mediated bone loss.


Archive | 1990

Oral Streptococcus mutans vaccines: diversity of approaches for the induction of mucosal responses

Suzanne M. Michalek; Noel K. Childers; Susan Jackson; Jenny Katz; Jiri Mestecky

In this paper, we have summarized our studies in humans and experimental animals concerned with the development of oral vaccines effective in inducing mucosal IgA responses. In our studies, we have used antigens of or anti-idiotypic antibodies that mimic Streptococcus mutans, the principal etiologic agent of dental caries, which has allowed an assessment of the induction of a mucosal immune response and a determination of the effectiveness of the response in protection against S. mutans infection. Evidence is provided indicating: 1) the usefulness of liposomes as carriers of purified antigen and as an adjuvant for the induction of mucosal responses to orally administered vaccine antigens, 2) the ability of anti-idiotypic antibodies to induce a protective mucosal response and 3) the induction of a primary and secondary salivary IgA anti-S. mutans serotype carbohydrate (CHO) immune response in humans after swallowing capsules containing S. mutans CHO in liposomes.


Archive | 1990

Rat T cell subpopulations: Effect of environmental and microbial antigens

Jenny Katz; John H. Eldridge; Suzanne M. Michalek

Extensive studies by Mosmann et al. [1,2] using cloned murine T cells have defined two different subsets of CD4+ T cells based on their lymphokine production and functional properties. However, the existance of similar subsets of CD4+ cells in other systems is still unclear. In the rat, it has been reported [3–5] that the monoclonal antibody OX22 which exhibits specificity for a high molecular weight form of the leukocyte common antigen (L-CA) divides CD4+ T cells into two subpopulations, with the OX22 monoclonal antibody binding to two-thirds of the T helper (Th) cells in the thoracic duct lymph. Recently, Powrie and Mason [6,7] also suggested that upon exposure of OX22+, CD4+ cells to antigen, the OX22 marker is lost and that these OX22−, CD4+ T cells represent memory cells.


Infection and Immunity | 1993

Protective salivary immunoglobulin A responses against Streptococcus mutans infection after intranasal immunization with S. mutans antigen I/II coupled to the B subunit of cholera toxin.

Jenny Katz; C C Harmon; G P Buckner; Gloria J. Richardson; M W Russell; Suzanne M. Michalek

Collaboration


Dive into the Jenny Katz's collaboration.

Top Co-Authors

Avatar

Suzanne M. Michalek

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

John H. Eldridge

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ping Zhang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

C C Harmon

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jerry R. McGhee

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Noel K. Childers

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Dalia E. Gaddis

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Gloria J. Richardson

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Richard L. Gregory

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Roy Curtiss

Arizona State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge