Network


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

Hotspot


Dive into the research topics where Stephen J. Davies is active.

Publication


Featured researches published by Stephen J. Davies.


Journal of Immunology | 2004

IL-13 Activates a Mechanism of Tissue Fibrosis That Is Completely TGF-β Independent

Mallika Kaviratne; Matthias Hesse; Mary Leusink; Allen W. Cheever; Stephen J. Davies; James H. McKerrow; Lalage M. Wakefield; John J. Letterio; Thomas A. Wynn

Fibrosis is a characteristic feature in the pathogenesis of a wide spectrum of diseases. Recently, it was suggested that IL-13-dependent fibrosis develops through a TGF-β1 and matrix metalloproteinase-9-dependent (MMP-9) mechanism. However, the significance of this pathway in a natural disorder of fibrosis was not investigated. In this study, we examined the role of TGF-β in IL-13-dependent liver fibrosis caused by Schistosoma mansoni infection. Infected IL-13−/− mice showed an almost complete abrogation of fibrosis despite continued and undiminished production of TGF-β1. Although MMP-9 activity was implicated in the IL-13 pathway, MMP-9−/− mice displayed no reduction in fibrosis, even when chronically infected. To directly test the requirement for TGF-β, studies were also performed with neutralizing anti-TGF-β Abs, soluble antagonists (soluble TGF-βR-Fc), and Tg mice (Smad3−/− and TGF-βRII-Fc Tg) that have disruptions in all or part of the TGF-β signaling cascade. In all cases, fibrosis developed normally and with kinetics similar to wild-type mice. Production of IL-13 was also unaffected. Finally, several genes, including interstitial collagens, several MMPs, and tissue inhibitors of metalloprotease-1 were up-regulated in TGF-β1−/− mice by IL-13, demonstrating that IL-13 activates the fibrogenic machinery directly. Together, these studies provide unequivocal evidence of a pathway of fibrogenesis that is IL-13 dependent but TGF-β1 independent, illustrating the importance of targeting IL-13 directly in the treatment of infection-induced fibrosis.


Journal of Immunology | 2012

Helminth Protection against Autoimmune Diabetes in Nonobese Diabetic Mice Is Independent of a Type 2 Immune Shift and Requires TGF-β

Marc P. Hübner; Yinghui Shi; Marina N. Torrero; Ellen Mueller; David Larson; Kateryna Soloviova; Fabian Gondorf; Achim Hoerauf; Kristin E. Killoran; J. Thomas Stocker; Stephen J. Davies; Kristin V. Tarbell; Edward Mitre

Leading hypotheses to explain helminth-mediated protection against autoimmunity postulate that type 2 or regulatory immune responses induced by helminth infections in the host limit pathogenic Th1-driven autoimmune responses. We tested these hypotheses by investigating whether infection with the filarial nematode Litomosoides sigmodontis prevents diabetes onset in IL-4–deficient NOD mice and whether depletion or absence of regulatory T cells, IL-10, or TGF-β alters helminth-mediated protection. In contrast to IL-4–competent NOD mice, IL-4–deficient NOD mice failed to develop a type 2 shift in either cytokine or Ab production during L. sigmodontis infection. Despite the absence of a type 2 immune shift, infection of IL-4–deficient NOD mice with L. sigmodontis prevented diabetes onset in all mice studied. Infections in immunocompetent and IL-4–deficient NOD mice were accompanied by increases in CD4+CD25+Foxp3+ regulatory T cell frequencies and numbers, respectively, and helminth infection increased the proliferation of CD4+Foxp3+ cells. However, depletion of CD25+ cells in NOD mice or Foxp3+ T cells from splenocytes transferred into NOD.scid mice did not decrease helminth-mediated protection against diabetes onset. Continuous depletion of the anti-inflammatory cytokine TGF-β, but not blockade of IL-10 signaling, prevented the beneficial effect of helminth infection on diabetes. Changes in Th17 responses did not seem to play an important role in helminth-mediated protection against autoimmunity, because helminth infection was not associated with a decreased Th17 immune response. This study demonstrates that L. sigmodontis-mediated protection against diabetes in NOD mice is not dependent on the induction of a type 2 immune shift but does require TGF-β.


PLOS Neglected Tropical Diseases | 2009

A Schistosome cAMP-Dependent Protein Kinase Catalytic Subunit Is Essential for Parasite Viability

Brett E. Swierczewski; Stephen J. Davies

Eukaryotes, protozoan, and helminth parasites make extensive use of protein kinases to control cellular functions, suggesting that protein kinases may represent novel targets for the development of anti-parasitic drugs. Because of their central role in intracellular signaling pathways, cyclic nucleotide–dependent kinases such as cAMP-dependent protein kinase (PKA) represent promising new targets for the treatment of parasitic infections and neoplastic disorders. However, the role of these kinases in schistosome biology has not been characterized and the genes encoding schistosome PKAs have not been identified. Here we provide biochemical evidence for the presence of a PKA signaling pathway in adult Schistosoma mansoni and show that PKA activity is required for parasite viability in vitro. We also provide the first full description of a gene that encodes a PKA catalytic subunit in S. mansoni, named SmPKA-C. Finally we demonstrate, through RNA interference, that SmPKA-C contributes to the PKA activity we detected biochemically and that inhibition of SmPKA-C expression in adult schistosomes results in parasite death. Together our data show that SmPKA-C is a critically important gene product and may represent an attractive therapeutic target for the treatment and control of schistosomiasis.


Journal of Immunology | 2012

Chronic Helminth Infection Reduces Basophil Responsiveness in an IL-10–Dependent Manner

David Larson; Marc P. Hübner; Marina N. Torrero; Christopher P. Morris; Amy Brankin; Brett E. Swierczewski; Stephen J. Davies; Becky M. Vonakis; Edward Mitre

Basophils play a key role in the development and effector phases of type 2 immune responses in both allergic diseases and helminth infections. This study shows that basophils become less responsive to IgE-mediated stimulation when mice are chronically infected with Litomosoides sigmodontis, a filarial nematode, and Schistosoma mansoni, a blood fluke. Although excretory/secretory products from microfilariae of L. sigmodontis suppressed basophils in vitro, transfer of microfilariae into mice did not result in basophil suppression. Rather, reduced basophil responsiveness, which required the presence of live helminths, was found to be dependent on host IL-10 and was accompanied by decreases in key IgE signaling molecules known to be downregulated by IL-10. Given the importance of basophils in the development of type 2 immune responses, these findings help explain the mechanism by which helminths protect against allergy and may have broad implications for understanding how helminth infections alter other disease states in people.


BMC Immunology | 2010

Rapid induction of IgE responses to a worm cysteine protease during murine pre-patent schistosome infection

Lucia Alves de Oliveira Fraga; Erika W Lamb; Elizabeth C Moreno; Mitali Chatterjee; Jan Dvořák; Melaine Delcroix; Mohammed Sajid; Conor R. Caffrey; Stephen J. Davies

BackgroundDuring the pre-patent stage of infection, juvenile Schistosoma blood flukes co-opt signals from the adaptive immune system to facilitate parasite development, but the types of responses that are induced at this early stage of infection, and the parasite antigens they target, have not been characterized.ResultsThrough analysis of experimental pre-patent infections, we show that the S. mansoni cysteine protease SmCB1 is rapidly targeted by an antigen-specific IgE response. The induction of this response is independent of schistosome eggs as infection with male or female worms alone also induced SmCB1-specific IgE. We also show that the SmCB1-specific IgE response is dependent on cognate CD4+ T cell help and IL-4, suggesting that pre-patent Th2 responses provide T cell help for the SmCB1-specific IgE response. Finally, exposed human subjects also produced IgE against SmCB1.ConclusionsOur data demonstrate that, like eggs, schistosome worms also induce functional type 2 responses and that a parasite cysteine protease is an inducer of type 2 responses during the early stages of schistosome infection.


The Journal of Infectious Diseases | 2010

Induction of type 2 responses by schistosome worms during prepatent infection.

Lucia Alves de Oliveira Fraga; Marina N. Torrero; Anna S. Tocheva; Edward Mitre; Stephen J. Davies

During natural schistosome infection, the induction of T helper type 2 (Th2) responses has been ascribed to parasite eggs, because exposure of the host to this life-cycle stage elicits a polarized Th2 response to egg antigens. In the present study, we show that schistosome worms also elicit systemic, antigen-specific type 2 responses during prepatent infection, before egg deposition begins. CD4(+) T cells producing interleukin (IL)-4 were induced by both male and female worms during single-sex infections, demonstrating that this response is independent of exposure to eggs. The Th2 response was accompanied by production of immunoglobulin E and the sensitization of circulating basophils to produce additional IL-4 in response to schistosome antigens. Together, our data show that schistosome worms establish an immunologic milieu where CD4(+) T cells and basophils are both primed to produce IL-4 before eggs are laid, suggesting that worms play a role in establishment of the Th2 response that is critical for host survival and parasite transmission.


Trends in Parasitology | 2011

Schistosoma comparative genomics: integrating genome structure, parasite biology and anthelmintic discovery.

Martin T. Swain; Denis M. Larkin; Conor R. Caffrey; Stephen J. Davies; Alex Loukas; Patrick J. Skelly; Karl F. Hoffmann

Schistosoma genomes provide a comprehensive resource for identifying the molecular processes that shape parasite evolution and for discovering novel chemotherapeutic or immunoprophylactic targets. Here, we demonstrate how intragenus and intergenus comparative genomics can be used to drive these investigations forward, illustrate the advantages and limitations of these approaches and review how post-genomic technologies offer complementary strategies for genome characterisation. Although sequencing and functional characterisation of other schistosome/platyhelminth genomes continues to expedite anthelmintic discovery, we contend that future priorities should equally focus on improving assembly quality, and chromosomal assignment, of existing schistosome/platyhelminth genomes.


PLOS Pathogens | 2010

Blood Fluke Exploitation of Non-Cognate CD4+ T Cell Help to Facilitate Parasite Development

Erika W. Lamb; Colleen D. Walls; John T. Pesce; Diana K. Riner; Sean K. Maynard; Emily T. Crow; Thomas A. Wynn; Brian C. Schaefer; Stephen J. Davies

Schistosoma blood flukes, which infect over 200 million people globally, co-opt CD4+ T cell-dependent mechanisms to facilitate parasite development and egg excretion. The latter requires Th2 responses, while the mechanism underpinning the former has remained obscure. Using mice that are either defective in T cell receptor (TCR) signaling or that lack TCRs that can respond to schistosomes, we show that naïve CD4+ T cells facilitate schistosome development in the absence of T cell receptor signaling. Concurrently, the presence of naïve CD4+ T cells correlates with both steady-state changes in the expression of genes that are critical for the development of monocytes and macrophages and with significant changes in the composition of peripheral mononuclear phagocyte populations. Finally, we show that direct stimulation of the mononuclear phagocyte system restores blood fluke development in the absence of CD4+ T cells. Thus we conclude that schistosomes co-opt innate immune signals to facilitate their development and that the role of CD4+ T cells in this process may be limited to the provision of non-cognate help for mononuclear phagocyte function. Our findings have significance for understanding interactions between schistosomiasis and other co-infections, such as bacterial infections and human immunodeficiency virus infection, which potently stimulate innate responses or interfere with T cell help, respectively. An understanding of immunological factors that either promote or inhibit schistosome development may be valuable in guiding the development of efficacious new therapies and vaccines for schistosomiasis.


International Journal for Parasitology | 2010

Developmental regulation of protein kinase A expression and activity in Schistosoma mansoni.

Brett E. Swierczewski; Stephen J. Davies

cAMP-dependent protein kinases (PKAs) are the main transducers of cAMP signalling in eukaryotic cells. Recently we reported the identification and characterisation of a PKA catalytic subunit (SmPKA-C) in Schistosoma mansoni that is required for adult schistosome viability in vitro. To gain further insights into the role of SmPKA-C in biological processes during the schistosome life cycle, we undertook a quantitative analysis of SmPKA-C mRNA expression in different life cycle stages. Our data shows that SmPKA-C mRNA expression is developmentally regulated, with the highest levels of expression in cercariae and adult female worms. To evaluate the biological role of SmPKA-C in these developmental stages, cercariae and adult worms were treated with various concentrations of PKA inhibitors. Treatment of cercariae with H-89 or PKI 14-22 amide resulted in loss of viability suggesting that, as in adults, PKA is an essential enzyme activity in this infectious larval stage. In adult worms, in vitro exposure to sub-lethal concentrations of H-89 or PKI 14-22 amide resulted in inhibition of egg production in a dose-dependent manner. Furthermore, using a murine model of schistosome infection where S. mansoni fecundity is impaired, we show that reduced rates of egg production in vivo correlate with significant reductions in SmPKA-C mRNA expression and PKA activity. Finally, restoration of parasite egg production in vivo also resulted in normalisation of SmPKA-C mRNA expression and PKA activity. Taken together, our data suggest that PKA signalling is required for cercarial viability and may play a specific role in the reproductive activity of adult worms.


The Journal of Infectious Diseases | 2006

The Common γ Chain Cytokines Interleukin (IL)–2 and IL-7 Indirectly Modulate Blood Fluke Development via Effects on CD4+ T Cells

Rebecca B. Blank; Erika W. Lamb; Anna S. Tocheva; Emily T. Crow; Kee-Chong Lim; James H. McKerrow; Stephen J. Davies

The blood fluke Schistosoma mansoni is a causative agent of schistosomiasis and a major global health problem. After penetrating the skin of the host, schistosomula migrate via the bloodstream to the portal circulation, arriving during the second week of infection [1]. On reaching the liver, migration ceases, and rapid growth and development ensues [2], culminating in the sexual maturation of the parasites, pairing, and the onset of oviposition at ~5 weeks after infection. Eggs are deposited in and transit across the wall of the intestine, emerging into the lumen and passing from the body in feces. Deposition of eggs in host tissues and the resulting inflammatory and immune responses are the primary cause of the pathological and clinical manifestations associated with schistosome infection. Deposition of schistosome eggs in the bowel wall and other tissues induces formation of granulomas around the eggs, a process that is mediated by CD4+ Th2 responses to egg antigens [3]. Formation of granulomas around schistosome eggs appears to facilitate the transit of eggs across the intestinal wall and their egress from the body [4]. Interestingly, CD4+ T cells also facilitate S. mansoni development during the prepatent period, because parasite development is severely impaired in immunodeficient mice and normal development is restored by adoptive transfer of CD4+ T cells into immunodeficient mice [5]. Taken together, these findings may explain why patients with schistosomiasis who are coinfected with HIV excrete fewer eggs in their feces than patients who are HIV negative [6]. Observations from patients with schistosome-HIV coinfection highlight a general recurring theme in helminth-HIV coinfections—that, in contrast to bacteria, fungi, and protozoa, immunosuppression by HIV does not lead to fulminant helminth infections [7]. Rather, HIV coinfection either has little effect on helminth infections, as is the case with most intestinal helminths [7], or may create conditions that are unfavorable to helminth development in the host, as is the case for Strongyloides stercoralis infection [8]. The latter finding correlates well with the notable lack of association between HIV infection and disseminated strongyloidiasis [7]. Attenuated schistosome development was also reported in IL-7–deficient (IL-7−/−) mice [9], suggesting that this host factor may play a direct role in promoting parasite development. Because IL-7 is not expressed by T lymphocytes but is produced by endothelial and epithelial cells such as thymic stromal cells, it is possible that IL-7 represents a distinct mechanism by which host factors modulate schistosome development. However, IL-7 is critical for the thymic development of T lymphocytes and their subsequent survival in the periphery, as demonstrated by the severely lymphopenic phenotype of IL-7−/− [10] and IL-7 receptor (IL-7R)−/− [11] mice. We, therefore, hypothesized that the defective schistosome development reported in IL-7−/− mice was the indirect result of impaired T cell development and T lymphopenia in the absence of IL-7 signaling rather than the result of a direct effect that IL-7 has on schistosomes. To test this hypothesis, we examined schistosome infections in mice that lack components of the IL-7R complex (IL-7Rα and common γ [γc] chains) because IL-7 signaling is abrogated in these mice [11, 12] but IL-7 synthesis is intact, as demonstrated by their ability to sustain homeostatic proliferation of adoptively transferred IL-7R–expressing T cells [13]. Our data support the conclusion that the effects that IL-7 has on schistosome development are indeed mediated indirectly via the effects that IL-7 has on T cell homeostasis. Furthermore, our analyses identify an indirect role that another γc cytokine, IL-2, plays in schistosome development. Thus, although expression of Th1 or Th2 phenotypes and their associated effector cytokines by CD4+ T cells does not play a role in schistosome development [5, 14], autocrine production of the T cell growth factor IL-2 by CD4+ T cells is essential to the mechanism by which these cells modulate blood fluke development.

Collaboration


Dive into the Stephen J. Davies's collaboration.

Top Co-Authors

Avatar

Edward Mitre

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Brett E. Swierczewski

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

David A. Florin

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Edgar D. Rowton

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

George Schultz

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kee-Chong Lim

University of California

View shared research outputs
Top Co-Authors

Avatar

Lisa W. Keep

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Marina N. Torrero

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Phillip G. Lawyer

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge