Network


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

Hotspot


Dive into the research topics where Nicole D. Pecora is active.

Publication


Featured researches published by Nicole D. Pecora.


Molecular Microbiology | 2007

The trans‐envelope Tol–Pal complex is part of the cell division machinery and required for proper outer‐membrane invagination during cell constriction in E. coli

Matthew A. Gerding; Yasuyuki Ogata; Nicole D. Pecora; Hironori Niki; Piet A. J. de Boer

Fission of bacterial cells involves the co‐ordinated invagination of the envelope layers. Invagination of the cytoplasmic membrane (IM) and peptidoglycan (PG) layer is likely driven by the septal ring organelle. Invagination of the outer membrane (OM) in Gram‐negative species is thought to occur passively via its tethering to the underlying PG layer with generally distributed PG‐binding OM (lipo)proteins. The Tol–Pal system is energized by proton motive force and is well conserved in Gram‐negative bacteria. It consists of five proteins that can connect the OM to both the PG and IM layers via protein–PG and protein–protein interactions. Although the system is needed to maintain full OM integrity, and for class A colicins and filamentous phages to enter cells, its precise role has remained unclear. We show that all five components accumulate at constriction sites in Escherichia coli and that mutants lacking an intact system suffer delayed OM invagination and contain large OM blebs at constriction sites and cell poles. We propose that Tol–Pal constitutes a dynamic subcomplex of the division apparatus in Gram‐negative bacteria that consumes energy to establish transient trans‐envelope connections at/near the septal ring to draw the OM onto the invaginating PG and IM layers during constriction.


Journal of Immunology | 2006

Mycobacterium tuberculosis LprA is a lipoprotein agonist of TLR2 that regulates innate immunity and APC function

Nicole D. Pecora; Adam J. Gehring; David H. Canaday; W. Henry Boom; Clifford V. Harding

TLR2 recognizes components of Mycobacterium tuberculosis (Mtb) and initiates responses by APCs that influence both innate and adaptive immunity. Mtb lipoproteins are an important class of TLR2 ligand, but only two, LpqH and LprG, have been characterized to date. In this study, we characterize a third Mtb lipoprotein, LprA, and determine its effects on host macrophages and dendritic cells. LprA is a cell wall-associated lipoprotein with no homologs outside the slow-growing mycobacteria. Using Mycobacterium smegmatis as an expression host, we purified 6× His-tagged LprA both with and without its acyl modifications. Acylated LprA had agonist activity for both human and murine TLR2 and induced expression of TNF-α, IL-10, and IL-12. LprA also induced dendritic cell maturation as shown by increased expression of CD40, CD80, and class II MHC (MHC-II). In macrophages, prolonged (24 h) incubation with LprA decreased IFN-γ-induced MHC-II Ag processing and presentation, consistent with an observed decrease in MHC-II expression (macrophage viability was not affected and apoptosis was not induced by LprA). Reduced MHC-II Ag presentation may represent a negative feedback mechanism for control of inflammation that may be subverted by Mtb for immune evasion. Thus, Mtb LprA is a TLR2 agonist that induces cytokine responses and regulates APC function.


Cellular Immunology | 2009

TLR2 and its co-receptors determine responses of macrophages and dendritic cells to lipoproteins of Mycobacterium tuberculosis

Michael G. Drage; Nicole D. Pecora; Amy G. Hise; Maria Febbraio; Roy L. Silverstein; Douglas T. Golenbock; W. Henry Boom; Clifford V. Harding

Mycobacterium tuberculosis (Mtb) signals through Toll-like receptor 2 (TLR2) to regulate antigen presenting cells (APCs). Mtb lipoproteins, including LpqH, LprA, LprG and PhoS1, are TLR2 agonists, but their co-receptor requirements are unknown. We studied Mtb lipoprotein-induced responses in TLR2(-/-), TLR1(-/-), TLR6(-/-), CD14(-/-) and CD36(-/-) macrophages. Responses to LprA, LprG, LpqH and PhoS1 were completely dependent on TLR2. LprG, LpqH, and PhoS1 were dependent on TLR1, but LprA did not require TLR1. None of the lipoproteins required TLR6, although a redundant contribution by TLR6 cannot be excluded. CD14 contributed to detection of LprA, LprG and LpqH, whereas CD36 contributed only to detection of LprA. Studies of lung APC subsets revealed lower TLR2 expression by CD11b(high)/CD11c(low) lung macrophages than CD11b(low)/CD11c(high) alveolar macrophages, which correlated with hyporesponsiveness of lung macrophages to LpqH. Thus, lung APC subsets differ in TLR expression, which may determine differences in responses to Mtb.


Nature Structural & Molecular Biology | 2010

Mycobacterium tuberculosis lipoprotein LprG (Rv1411c) binds triacylated glycolipid agonists of Toll-like receptor 2

Michael G. Drage; Han Chun Tsai; Nicole D. Pecora; Tan Yun Cheng; Ahmad R. Arida; Supriya Shukla; Roxana E. Rojas; Chetan Seshadri; D. Branch Moody; W. Henry Boom; James C. Sacchettini; Clifford V. Harding

Knockout of lprG results in decreased virulence of Mycobacterium tuberculosis (MTB) in mice. MTB lipoprotein LprG has TLR2 agonist activity, which is thought to be dependent on its N-terminal triacylation. Unexpectedly, here we find that nonacylated LprG retains TLR2 activity. Moreover, we show LprG association with triacylated glycolipid TLR2 agonists lipoarabinomannan, lipomannan and phosphatidylinositol mannosides (which share core structures). Binding of triacylated species was specific to LprG (not LprA) and increased LprG TLR2 agonist activity; conversely, association of glycolipids with LprG enhanced their recognition by TLR2. The crystal structure of LprG in complex with phosphatidylinositol mannoside revealed a hydrophobic pocket that accommodates the three alkyl chains of the ligand. In conclusion, we demonstrate a glycolipid binding function of LprG that enhances recognition of triacylated MTB glycolipids by TLR2 and may affect glycolipid assembly or transport for bacterial cell wall biogenesis.


Cellular Immunology | 2009

Mycobacterium bovis BCG decreases MHC-II expression in vivo on murine lung macrophages and dendritic cells during aerosol infection

Nicole D. Pecora; Scott A. Fulton; Scott M. Reba; Michael G. Drage; Daimon P. Simmons; Nancy Urankar-Nagy; W. Henry Boom; Clifford V. Harding

Mycobacterium tuberculosis and M. bovis BCG infect APCs. In vitro, mycobacteria inhibit IFN-gamma-induced MHC-II expression by macrophages, but the effects of mycobacteria on lung APCs in vivo remain unclear. To assess MHC-II expression on APCs infected in vivo, mice were aerosol-infected with GFP-expressing BCG. At 28 d, approximately 1% of lung APCs were GFP+ by flow cytometry and CFU data. Most GFP+ cells were CD11b(high)/CD11c(neg-mid) lung macrophages (58-68%) or CD11b(high)/CD11c(high) DCs (28-31%). Lung APC MHC-II expression was higher in infected mice than naïve mice. Within infected lungs, however, MHC-II expression was lower in GFP+ cells than GFP- cells for both macrophages and DCs. MHC-II expression was also inhibited on purified lung macrophages and DCs that were infected with BCG in vitro. Thus, lung APCs that harbor mycobacteria in vivo have decreased MHC-II expression relative to uninfected APCs from the same lung, possibly contributing to evasion of T cell responses.


Infection and Immunity | 2011

Mycobacterium tuberculosis Lipoproteins Directly Regulate Human Memory CD4 T Cell Activation via Toll-Like Receptors 1 and 2

Christina Lancioni; Qing Li; Jeremy J. Thomas; Xue Dong Ding; Bonnie Thiel; Michael G. Drage; Nicole D. Pecora; Assem G. Ziady; Samuel Shank; Clifford V. Harding; W. Henry Boom; Roxana E. Rojas

ABSTRACT The success of Mycobacterium tuberculosis as a pathogen relies on its ability to regulate the host immune response. M. tuberculosis can manipulate adaptive T cell responses indirectly by modulating antigen-presenting cell (APC) function or by directly interacting with T cells. Little is known about the role of M. tuberculosis molecules in direct regulation of T cell function. Using a biochemical approach, we identified lipoproteins LprG and LpqH as major molecules in M. tuberculosis lysate responsible for costimulation of primary human CD4+ T cells. In the absence of APCs, activation of memory CD4+ T cells with LprG or LpqH in combination with anti-CD3 antibody induces Th1 cytokine secretion and cellular proliferation. Lipoprotein-induced T cell costimulation was inhibited by blocking antibodies to Toll-like receptor 2 (TLR2) and TLR1, indicating that human CD4+ T cells can use TLR2/TLR1 heterodimers to directly respond to M. tuberculosis products. M. tuberculosis lipoproteins induced NF-κB activation in CD4+ T cells in the absence of TCR co-engagement. Thus, TLR2/TLR1 engagement alone by M. tuberculosis lipoprotein triggered intracellular signaling, but upregulation of cytokine production and proliferation required co-engagement of the TCR. In conclusion, our results demonstrate that M. tuberculosis lipoproteins LprG and LpqH participate in the regulation of adaptive immunity not only by inducing cytokine secretion and costimulatory molecules in innate immune cells but also through directly regulating the activation of memory T lymphocytes.


Proceedings of the National Academy of Sciences of the United States of America | 2001

CD5 expression by B lymphocytes and its regulation upon Epstein-Barr virus transformation.

David L. Kaplan; Dawn Smith; Howard Meyerson; Nicole D. Pecora; Kristine Lewandowska

Dim expression of CD5 on human B lymphocytes has been used to delineate B1 and B2 subsets. Nevertheless, others have suggested that the molecule is an activation marker and does not predicate a subset distinction. We have used enzymatic amplification staining, a technology that enhances the resolution of flow cytometric analysis of cell surface molecules by as much as 100-fold, to determine that essentially all human B cells express CD5. Furthermore, we show that this expression is regulated during Epstein–Barr virus transformation.


Journal of Virology | 2010

Mycobacterium tuberculosis Promotes HIV trans-Infection and Suppresses Major Histocompatibility Complex Class II Antigen Processing by Dendritic Cells

Morgan A. Reuter; Nicole D. Pecora; Clifford V. Harding; David H. Canaday; David McDonald

ABSTRACT Mycobacterium tuberculosis is a leading killer of HIV-infected individuals worldwide, particularly in sub-Saharan Africa, where it is responsible for up to 50% of HIV-related deaths. Infection by HIV predisposes individuals to M. tuberculosis infection, and coinfection accelerates the progression of both diseases. In contrast to most other opportunistic infections associated with HIV, an increased risk of M. tuberculosis infection occurs during early-stage HIV disease, long before CD4 T cell counts fall below critical levels. We hypothesized that M. tuberculosis infection contributes to HIV pathogenesis by interfering with dendritic cell (DC)-mediated immune control. DCs carry pathogens like M. tuberculosis and HIV from sites of infection into lymphoid tissues, where they process and present antigenic peptides to CD4 T cells. Paradoxically, DCs can also deliver infectious HIV to T cells without first becoming infected, a process known as trans-infection. Lipopolysaccharide (LPS)-activated DCs sequester HIV in pocketlike membrane invaginations that remain open to the cell surface, and individual virions are delivered from the pocket into T cells at the site of contact during trans-infection. Here we report that M. tuberculosis exposure increases HIV trans-infection and induces viral sequestration within surface-accessible compartments identical to those seen in LPS-stimulated DCs. At the same time, M. tuberculosis dramatically decreases the degradative processing and major histocompatibility complex class II (MHC-II) presentation of HIV antigens to CD4 T cells. Our data suggest that M. tuberculosis infection promotes a shift in the dynamic balance between antigen processing and intact virion presentation, favoring DC-mediated amplification of HIV infections.


Journal of Leukocyte Biology | 2012

Rv2468c, a novel Mycobacterium tuberculosis protein that costimulates human CD4+ T cells through VLA-5.

Qing Li; Xuedong Ding; Jeremy J. Thomas; Clifford V. Harding; Nicole D. Pecora; Assem G. Ziady; Samuel Shank; W. Henry Boom; Christina L. Lancioni; Roxana E. Rojas

Mtb regulates many aspects of the host immune response, including CD4+ T lymphocyte responses that are essential for protective immunity to Mtb, and Mtb effects on the immune system are paradoxical, having the capacity to inhibit (immune evasion) and to activate (adjuvant effect) immune cells. Mtb regulates CD4+ T cells indirectly (e.g., by manipulation of APC function) and directly, via integrins and TLRs expressed on T cells. We now report that previously uncharacterized Mtb protein Rv2468c/MT2543 can directly regulate human CD4+ T cell activation by delivering costimulatory signals. When combined with TCR stimulation (e.g., anti‐CD3), Rv2468c functioned as a direct costimulator for CD4+ T cells, inducing IFN‐γ secretion and T cell proliferation. Studies with blocking antibodies and soluble RGD motifs demonstrated that Rv2468c engaged integrin VLA‐5 (α5β1) on CD4+ T cells through its FN‐like RGD motif. Costimulation by Rv2468c induced phosphorylation of FAKs and Pyk2. These results reveal that by expressing molecules that mimic host protein motifs, Mtb can directly engage receptors on CD4+ T cells and regulate their function. Rv2468c‐induced costimulation of CD4+ T cells could have implications for TB immune pathogenesis and Mtb adjuvant effect.


Fems Microbiology Letters | 2002

Isolation of lacZ fusions to Proteus mirabilis genes regulated by intercellular signaling: potential role for the sugar phosphotransferase (Pts) system in regulation

Gwen Sturgill; Soofia Siddiqui; Xuedong Ding; Nicole D. Pecora; Philip N. Rather

Collaboration


Dive into the Nicole D. Pecora's collaboration.

Top Co-Authors

Avatar

Clifford V. Harding

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Michael G. Drage

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

W. Henry Boom

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Roxana E. Rojas

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Jeremy J. Thomas

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Xuedong Ding

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Assem G. Ziady

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Christina Lancioni

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

David H. Canaday

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Qing Li

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge