Network


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

Hotspot


Dive into the research topics where Michael E. Woodman is active.

Publication


Featured researches published by Michael E. Woodman.


Infection and Immunity | 2007

Coordinated Expression of Borrelia burgdorferi Complement Regulator-Acquiring Surface Proteins during the Lyme Disease Spirochete's Mammal-Tick Infection Cycle

Tomasz Bykowski; Michael E. Woodman; Anne E. Cooley; Catherine A. Brissette; Volker Brade; Reinhard Wallich; Peter Kraiczy; Brian Stevenson

ABSTRACT The Lyme disease spirochete, Borrelia burgdorferi, is largely resistant to being killed by its hosts’ alternative complement activation pathway. One possible resistance mechanism of these bacteria is to coat their surfaces with host complement regulators, such as factor H. Five different B. burgdorferi outer surface proteins having affinities for factor H have been identified: complement regulator-acquiring surface protein 1 (BbCRASP-1), encoded by cspA; BbCRASP-2, encoded by cspZ; and three closely related proteins, BbCRASP-3, -4, and -5, encoded by erpP, erpC, and erpA, respectively. We now present analyses of the recently identified BbCRASP-2 and cspZ expression patterns throughout the B. burgdorferi infectious cycle, plus novel analyses of BbCRASP-1 and erp-encoded BbCRASPs. Our results, combined with data from earlier studies, indicate that BbCRASP-2 is produced primarily during established mammalian infection, while BbCRASP-1 is produced during tick-to-mammal and mammal-to-tick transmission stages but not during established mammalian infection, and Erp-BbCRASPs are produced from the time of transmission from infected ticks into mammals until they are later acquired by other feeding ticks. Transcription of cspZ and synthesis of BbCRASP-2 were severely repressed during cultivation in laboratory medium relative to mRNA levels observed during mammalian infection, and cspZ expression was influenced by culture temperature and pH, observations which will assist identification of the mechanisms employed by B. burgdorferi to control expression of this borrelial infection-associated protein.


Infection and Immunity | 2005

Borrelia burgdorferi Regulates Expression of Complement Regulator-Acquiring Surface Protein 1 during the Mammal-Tick Infection Cycle

Kate von Lackum; Jennifer C. Miller; Tomasz Bykowski; Sean P. Riley; Michael E. Woodman; Volker Brade; Peter Kraiczy; Brian Stevenson; Reinhard Wallich

ABSTRACT During the natural mammal-tick infection cycle, the Lyme disease spirochete Borrelia burgdorferi comes into contact with components of the alternative complement pathway. B. burgdorferi, like many other human pathogens, has evolved the immune evasion strategy of binding two host-derived fluid-phase regulators of complement, factor H and factor H-like protein 1 (FHL-1). The borrelial complement regulator-acquiring surface protein 1 (CRASP-1) is a surface-exposed lipoprotein that binds both factor H and FHL-1. Analysis of CRASP-1 expression during the mammal-tick infectious cycle indicated that B. burgdorferi expresses this protein during mammalian infection, supporting the hypothesized role for CRASP-1 in immune evasion. However, CRASP-1 synthesis was repressed in bacteria during colonization of vector ticks. Analysis of cultured bacteria indicated that CRASP-1 is differentially expressed in response to changes in pH. Comparisons of CRASP-1 expression patterns with those of other infection-associated B. burgdorferi proteins, including the OspC, OspA, and Erp proteins, indicated that each protein is regulated through a unique mechanism.


International Journal of Medical Microbiology | 2008

Borrelia burgdorferi complement regulator-acquiring surface proteins (BbCRASPs): Expression patterns during the mammal-tick infection cycle.

Tomasz Bykowski; Michael E. Woodman; Anne E. Cooley; Catherine A. Brissette; Reinhard Wallich; Volker Brade; Peter Kraiczy; Brian Stevenson

Host complement is widely distributed throughout mammalian body fluids and can be activated immediately as part of the first line of defense against invading pathogens. The agent of Lyme disease, Borrelia burgdorferi sensu lato (s.l.), is naturally resistant to that innate immune defense system of its hosts. One resistance mechanism appears to involve binding fluid-phase regulators of complement to distinct borrelial outer surface molecules known as CRASPs (complement regulator acquiring surface proteins). Using sensitive molecular biology techniques, expression patterns of all three classes of genes encoding the CRASPs of B. burgdorferi sensu stricto (BbCRASPs) have been analyzed throughout the natural tick-mammal infection cycle. Each class shows a different expression profile in vivo and the results are summarized herein. Studies on the expression of B. burgdorferi genes using animal models of infection have advanced our knowledge on the ability of the causative agent to circumvent innate immune defenses, the contributions of CRASPs to spirochete infectivity, and the pathogenesis of Lyme disease.


PLOS ONE | 2010

Interleukin-10 Mediated Autoregulation of Murine B-1 B-Cells and Its Role in Borrelia hermsii Infection

Vishal J. Sindhava; Michael E. Woodman; Brian Stevenson; Subbarao Bondada

B cells are typically characterized as positive regulators of the immune response, primarily by producing antibodies. However, recent studies indicate that various subsets of B cells can perform regulatory functions mainly through IL-10 secretion. Here we discovered that peritoneal B-1 (B-1P) cells produce high levels of IL-10 upon stimulation with several Toll-like receptor (TLR) ligands. High levels of IL-10 suppressed B-1P cell proliferation and differentiation response to all TLR ligands studied in an autocrine manner in vitro and in vivo. IL-10 that accumulated in cultures inhibited B-1P cells at second and subsequent cell divisions mainly at the G1/S interphase. IL-10 inhibits TLR induced B-1P cell activation by blocking the classical NF-κB pathway. Co-stimulation with CD40 or BAFF abrogated the IL-10 inhibitory effect on B-1P cells during TLR stimulation. Finally, B-1P cells adoptively transferred from the peritoneal cavity of IL-10−/− mice showed better clearance of Borrelia hermsii than wild-type B-1P cells. This study described a novel autoregulatory property of B-1P cells mediated by B-1P cell derived IL-10, which may affect the function of B-1P cells in infection and autoimmunity.


International Journal of Medical Microbiology | 2008

Lyme borreliosis spirochete Erp proteins, their known host ligands, and potential roles in mammalian infection.

Catherine A. Brissette; Anne E. Cooley; Logan H. Burns; Sean P. Riley; Ashutosh Verma; Michael E. Woodman; Tomasz Bykowski; Brian Stevenson

Lyme borreliae naturally maintain numerous distinct DNA elements of the cp32 family, each of which carries a mono- or bicistronic erp locus. The encoded Erp proteins are surface-exposed outer membrane lipoproteins that are produced at high levels during mammalian infection but largely repressed during colonization of vector ticks. Recent studies have revealed that some Erp proteins can serve as bacterial adhesins, binding host proteins such as the complement regulator factor H and the extracellular matrix component laminin. These results suggest that Erp proteins play roles in multiple aspects of mammalian infection.


Current protocols in microbiology | 2008

Direct PCR of Intact Bacteria (Colony PCR)

Michael E. Woodman

This protocol describes an efficient method for screening intact bacteria for the presence of desired DNA sequences using polymerase chain reaction (PCR). This method is commonly referred to as colony PCR. Curr. Protoc. Microbiol. 9:A.3D.1‐A.3D.6.


Infection and Immunity | 2007

Borrelia burgdorferi Binding of Host Complement Regulator Factor H Is Not Required for Efficient Mammalian Infection

Michael E. Woodman; Anne E. Cooley; Jennifer C. Miller; John J. Lazarus; Kathryn Tucker; Tomasz Bykowski; Marina Botto; Jens Hellwage; R. Mark Wooten; Brian Stevenson

ABSTRACT The causative agent of Lyme disease, Borrelia burgdorferi, is naturally resistant to its hosts alternative pathway of complement-mediated killing. Several different borrelial outer surface proteins have been identified as being able to bind host factor H, a regulator of the alternative pathway, leading to a hypothesis that such binding is important for borrelial resistance to complement. To test this hypothesis, the development of B. burgdorferi infection was compared between factor H-deficient and wild-type mice. Factor B- and C3-deficient mice were also studied to determine the relative roles of the alternative and classical/lectin pathways in B. burgdorferi survival during mammalian infection. While it was predicted that B. burgdorferi should be impaired in its ability to infect factor H-deficient animals, quantitative analyses of bacterial loads indicated that those mice were infected at levels similar to those of wild-type and factor B- and C3-deficient mice. Ticks fed on infected factor H-deficient or wild-type mice all acquired similar numbers of bacteria. Indirect immunofluorescence analysis of B. burgdorferi acquired by feeding ticks from the blood of infected mice indicated that none of the bacteria had detectable levels of factor H on their outer surfaces, even though such bacteria express high levels of surface proteins capable of binding factor H. These findings demonstrate that the acquisition of host factor H is not essential for mammalian infection by B. burgdorferi and indicate that additional mechanisms are employed by the Lyme disease spirochete to evade complement-mediated killing.


Journal of Leukocyte Biology | 2009

Roles for phagocytic cells and complement in controlling relapsing fever infection.

Michael E. Woodman; Anne E. Cooley; Rita Avdiushko; Amy Bowman; Marina Botto; R. Mark Wooten; Nico van Rooijen; Donald A. Cohen; Brian Stevenson

Relapsing fever spirochetes, such as Borrelia hermsii, proliferate to high levels in their hosts’ bloodstream until production of IgM against borrelial surface proteins promotes bacterial clearance. The mechanisms by which B. hermsii survives in host blood, as well as the immune mediators that control this infection, remain largely unknown. It has been hypothesized that B. hermsii is naturally resistant to killing by the alternative pathway of complement activation as a result of its ability to bind factor H, a host complement regulator. However, we found that Cfh−/− mice were infected to levels identical to those seen in wild‐type mice. Moreover, only a small minority of B. hermsii in the blood of wild‐type mice had detectable levels of factor H adhered to their outer surfaces. In vitro, complement was found to play a statistically significant role in antibody‐mediated inactivation of B. hermsii, although in vivo studies indicated that complement is not essential for host control of B. hermsii. Depletion of mφ and DC from mice had significant impacts on B. hermsii infection, and depleted mice were unable to control bloodstream infections, leading to death. Infection studies using muMT indicated a significant antibody‐independent role for mφ and/or DC in host control of relapsing fever infection. Together, these findings indicate mφ and/or DC play a critical role in the production of B. hermsii‐specific IgM and for antibody‐independent control of spirochete levels.


Current Protocols Essential Laboratory Techniques | 2008

Culture of Escherichia coli and Related Bacteria

Sean P. Riley; Michael E. Woodman; Brian Stevenson

This appendix provides general techniques, equipment, and media used for the growth of many commonly encountered bacteria. For specific growth conditions, readers should refer to units regarding the laboratory maintenance of the organism of interest.


Current protocols in microbiology | 2008

APPENDIX 3D Direct PCR of Intact Bacteria (Colony PCR)

Michael E. Woodman

This protocol describes an efficient method for screening intact bacteria for the presence of desired DNA sequences using polymerase chain reaction (PCR). This method is commonly referred to as colony PCR. Curr. Protoc. Microbiol. 9:A.3D.1‐A.3D.6.

Collaboration


Dive into the Michael E. Woodman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sean P. Riley

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge