Network


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

Hotspot


Dive into the research topics where Lori Fitz is active.

Publication


Featured researches published by Lori Fitz.


FEBS Letters | 2004

PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3ζ signalosome and downstream signaling to PKCθ

Kelly-Ann Sheppard; Lori Fitz; Julie M. Lee; Christina Benander; Judith A. George; Joe Wooters; Yongchang Qiu; Jason Jussif; Laura Carter; Clive Wood; Divya Chaudhary

Engagement of the immunoinhibitory receptor, programmed death‐1 (PD‐1) attenuates T‐cell receptor (TCR)‐mediated activation of IL‐2 production and T‐cell proliferation. Here, we demonstrate that PD‐1 modulation of T‐cell function involves inhibition of TCR‐mediated phosphorylation of ZAP70 and association with CD3ζ. In addition, PD‐1 signaling attenuates PKCθ activation loop phosphorylation in a cognate TCR signal. PKCθ has been shown to be required for T‐cell IL‐2 production. A phosphorylated PD‐1 peptide, corresponding to the C‐terminal immunoreceptor tyrosine‐switch motif (ITSM), acts as a docking site in vitro for both SHP‐2 and SHP‐1, while the phosphorylated peptide containing the N‐terminal PD‐1 immunoreceptor tyrosine based inhibitory motif (ITIM) associates only with SHP‐2.


Journal of Immunology | 2008

The Human IL-17F/IL-17A Heterodimeric Cytokine Signals through the IL-17RA/IL-17RC Receptor Complex

Jill F. Wright; Frann Bennett; Bilian Li; Jonathan Brooks; Deborah Luxenberg; Matthew J. Whitters; Kathleen N. Tomkinson; Lori Fitz; Neil M. Wolfman; Mary Collins; Kyri Dunussi-Joannopoulos; Moitreyee Chatterjee-Kishore; Beatriz M. Carreno

IL-17A and IL-17F, produced by the Th17 CD4+ T cell lineage, have been linked to a variety of inflammatory and autoimmune conditions. We recently reported that activated human CD4+ T cells produce not only IL-17A and IL-17F homodimers but also an IL-17F/IL-17A heterodimeric cytokine. All three cytokines can induce chemokine secretion from bronchial epithelial cells, albeit with different potencies. In this study, we used small interfering RNA and Abs to IL-17RA and IL-17RC to demonstrate that heterodimeric IL-17F/IL-17A cytokine activity is dependent on the IL-17RA/IL-17RC receptor complex. Interestingly, surface plasmon resonance studies indicate that the three cytokines bind to IL-17RC with comparable affinities, whereas they bind to IL-17RA with different affinities. Thus, we evaluated the effect of the soluble receptors on cytokine activity and we find that soluble receptors exhibit preferential cytokine blockade. IL-17A activity is inhibited by IL-17RA, IL-17F is inhibited by IL-17RC, and a combination of soluble IL-17RA/IL-17RC receptors is required for inhibition of the IL-17F/IL-17A activity. Altogether, these results indicate that human IL-17F/IL-17A cytokine can bind and signal through the same receptor complex as human IL-17F and IL-17A. However, the distinct affinities of the receptor components for IL-17A, IL-17F, and IL-17F/IL-17A heterodimer can be exploited to differentially affect the activity of these cytokines.


Journal of Biological Chemistry | 2004

Catalytic domain crystal structure of protein kinase C-theta (PKCtheta)

Zhangbao Xu; Divya Chaudhary; Stephane Olland; Scott Wolfrom; Robert M. Czerwinski; Karl Malakian; Laura Lin; Mark L. Stahl; Diane Joseph-McCarthy; Christina Benander; Lori Fitz; Rita Greco; William S. Somers; Lidia Mosyak

A member of the novel protein kinase C (PKC) subfamily, PKCθ, is an essential component of the T cell synapse and is required for optimal T cell activation and interleukin-2 production. Selective involvement of PKCθ in TCR signaling makes this enzyme an attractive therapeutic target in T cell-mediated disease processes. In this report we describe the crystal structure of the catalytic domain of PKCθ at 2.0-Å resolution. Human recombinant PKCθ kinase domain was expressed in bacteria as catalytically active phosphorylated enzyme and co-crystallized with its subnanomolar, ATP site inhibitor staurosporine. The structure follows the classic bilobal kinase fold and shows the enzyme in its active conformation and phosphorylated state. Inhibitory interactions between conserved features of staurosporine and the ATP-binding cleft are accompanied by closing of the glycine-rich loop, which also maintains an inhibitory arrangement by blocking the phosphate recognition subsite. The two major phosphorylation sites, Thr-538 in the activation loop and Ser-695 in the hydrophobic motif, are both occupied in the structure, playing key roles in stabilizing active conformation of the enzyme and indicative of PKCθ autocatalytic phosphorylation and activation during bacterial expression. The PKCθ-staurosporine complex represents the first kinase domain crystal structure of any PKC isotypes to be determined and as such should provide valuable insight into PKC specificity and into rational drug design strategies for PKCθ selective leads.


Oncogene | 1997

Characterization of murine Flt4 ligand/VEGF-C.

Lori Fitz; Morris Jc; Paul Towler; Andrew J. Long; Paul Burgess; Rita Greco; Jack Wang; Rob Gassaway; Elliott Nickbarg; Sharlotte Kovacic; Agnes B. Ciarletta; Joann Giannotti; Heather Finnerty; Richard Zollner; David R. Beier; Lee V. Leak; Katherine Turner; Clive Wood

Flt4 is a receptor protein tyrosine kinase that is expressed in the adult lymphatic endothelium and high endothelial venules. We have used a BIAcore assay to identify rodent and human cell conditioned media containing the ligand of Flt4 (Flt4-L). Receptor-based affinity chromatography was used to purify this growth factor, followed by amino acid sequencing and molecular cloning of the murine cDNA, the orthologue of human vascular endothelial growth factor-C and vascular endothelial growth factor related protein. The murine flt4-L gene was localized to chromosome 8 and demonstrated to be widely expressed. Flt4-L was found to have a hydrophobic signal sequence and a pro-peptide-like sequence that is removed to generate the mature N-terminus. In addition, the C-terminal region of Flt4-L has four repeats of a cysteine-rich motif that is presumably also proteolytically processed to generate the 21 000 Mr polypeptide subunit of the Flt4-L homodimer. Recombinant Flt4-L activated Flt4 as judged by induction of tyrosyl phosphorylation, and induced mitogenesis in vitro of lymphatic endothelial cells.


Journal of Immunology | 2010

Complement C3a, CpG Oligos, and DNA/C3a Complex Stimulate IFN-α Production in a Receptor for Advanced Glycation End Product-Dependent Manner

Benfang Helen Ruan; Xin Li; Aaron Winkler; Kristina Cunningham; Jun Kuai; Rita Greco; Karl Nocka; Lori Fitz; Jill F. Wright; Debra D. Pittman; Xiang-Yang Tan; Janet E. Paulsen; Lih Ling Lin; David Winkler

The receptor for advanced glycation end products (RAGE) is a multiligand transmembrane receptor implicated in a number of diseases including autoimmune diseases. To further understand the pathogenic mechanism of RAGE in these diseases, we searched for additional ligands. We discovered that C3a bound to RAGE with an EC50 of 1.9 nM in an ELISA, and the binding was increased both in magnitude (by >2-fold) and in affinity (EC50 70 pM) in the presence of human stimulatory unmethylated cytosine-guanine-rich DNA A (hCpGAs). Surface plasmon resonance and fluorescence anisotropy analyses demonstrated that hCpGAs could bind directly to RAGE and C3a and form a ternary complex. In human PBMCs, C3a increased IFN-α production in response to low levels of hCpGAs, and this synergy was blocked by soluble RAGE or by an Ab directed against RAGE. IFN-α production was reduced in response to mouse CpGAs and C3a in RAGE−/− mouse bone marrow cells compared wild-type mice. Taken together, these data demonstrate that RAGE is a receptor for C3a and CpGA. Through direct interaction, C3a and CpGA synergize to increase IFN-α production in a RAGE-dependent manner and stimulate an innate immune response. These findings indicate a potential role of RAGE in autoimmune diseases that show accumulation of immunostimulatory DNA and C3a.


Journal of Pharmacology and Experimental Therapeutics | 2008

Interleukin-13 Neutralization by Two Distinct Receptor Blocking Mechanisms Reduces Immunoglobulin E Responses and Lung Inflammation in Cynomolgus Monkeys

Marion T. Kasaian; Xiang-Yang Tan; Macy Jin; Lori Fitz; Kimberly Marquette; Nancy Wood; Timothy A. Cook; Julie Lee; Angela Widom; Rita Agostinelli; Andrea Bree; Franklin J. Schlerman; Stephane Olland; Michael Wadanoli; Joseph P. Sypek; Davinder Gill; Samuel J. Goldman; Lioudmila Tchistiakova

Interleukin (IL)-13 is a key cytokine driving allergic and asthmatic responses and contributes to airway inflammation in cynomolgus monkeys after segmental challenge with Ascaris suum antigen. IL-13 bioactivity is mediated by a heterodimeric receptor (IL-13Rα1/IL-4Rα) and can be inhibited in vitro by targeting IL-13 interaction with either chain. However, in cytokine systems, in vitro neutralization activity may not always predict inhibitory function in vivo. To address the efficacy of two different IL-13 neutralization mechanisms in a primate model of atopic disease, two humanized monoclonal antibodies to IL-13 were generated, with highly homologous properties, differing in epitope recognition. Ab01 blocks IL-13 interaction with IL-4Rα, and Ab02 blocks IL-13 interaction with IL-13Rα1. In a cynomolgus monkey model of IgE responses to A. suum antigen, both Ab01 and Ab02 effectively reduced serum titers of Ascaris-specific IgE and diminished ex vivo Ascaris-triggered basophil histamine release, assayed 8 weeks after a single administration of antibody. The two antibodies also produced comparable reductions in pulmonary inflammation after lung segmental challenge with Ascaris antigen. Increased serum levels of IL-13, lacking demonstrable biological activity, were seen postchallenge in animals given either anti-IL-13 antibody but not in control animals given human IgG of irrelevant specificity. These findings demonstrate a potent effect of IL-13 neutralization on IgE-mediated atopic responses in a primate system and show that IL-13 can be efficiently neutralized by targeting either the IL-4Rα-binding epitope or the IL-13Rα1-binding epitope.


Journal of Medicinal Chemistry | 2008

Identification, Characterization and Initial Hit-to-Lead Optimization of a Series of 4-Arylamino-3-Pyridinecarbonitrile as Protein Kinase C theta (PKCθ) Inhibitors

Derek Cecil Cole; Magda Asselin; Agnes Brennan; Robert M. Czerwinski; John W. Ellingboe; Lori Fitz; Rita Greco; Xinyi Huang; Diane Joseph-McCarthy; Michael F. Kelly; Matthew Kirisits; Julie Lee; Yuanhong Li; Paul Morgan; Joseph Raymond Stock; Désirée H. H. Tsao; Allan Wissner; Xiaoke Yang; Divya Chaudhary

The protein kinase C (PKC) family of serine/threonine kinases is implicated in a wide variety of cellular processes. The PKC theta (PKCtheta) isoform is involved in TCR signal transduction and T cell activation and regulates T cell mediated diseases, including lung inflammation and airway hyperresponsiveness. Thus inhibition of PKCtheta enzyme activity by a small molecule represents an attractive strategy for the treatment of asthma. A PKCtheta high-throughput screening (HTS) campaign led to the identification of 4-(3-bromophenylamino)-5-(3,4-dimethoxyphenyl)-3-pyridinecarbonitrile 4a, a low microM ATP competitive PKCtheta inhibitor. Structure based hit-to-lead optimization led to the identification of 5-(3,4-dimethoxyphenyl)-4-(1H-indol-5-ylamino)-3-pyridinecarbonitrile 4p, a 70 nM PKCtheta inhibitor. Compound 4p was selective for inhibition of novel PKC isoforms over a panel of 21 serine/threonine, tyrosine, and phosphoinositol kinases, in addition to the conventional and atypical PKCs, PKCbeta, and PKCzeta, respectively. Compound 4p also inhibited IL-2 production in antiCD3/anti-CD28 activated T cells enriched from splenocytes.


Protein Science | 2009

Triad of polar residues implicated in pH specificity of acidic mammalian chitinase

Andrea Olland; James Strand; Eleonora Presman; Robert M. Czerwinski; Diane Joseph-McCarthy; Rustem Krykbaev; Gerhard Schlingmann; Rajiv Chopra; Laura Lin; Margaret Fleming; Ron Kriz; Mark Stahl; William Stuart Somers; Lori Fitz; Lidia Mosyak

Acidic mammalian chitinase (AMCase) is a mammalian chitinase that has been implicated in allergic asthma. One of only two active mammalian chinases, AMCase, is distinguished from other chitinases by several unique features. Here, we present the novel structure of the AMCase catalytic domain, both in the apo form and in complex with the inhibitor methylallosamidin, determined to high resolution by X‐ray crystallography. These results provide a structural basis for understanding some of the unique characteristics of this enzyme, including the low pH optimum and the preference for the β‐anomer of the substrate. A triad of polar residues in the second‐shell is found to modulate the highly conserved chitinase active site. As a novel target for asthma therapy, structural details of AMCase activity will help guide the future design of specific and potent AMCase inhibitors.


PLOS Pathogens | 2012

Chitinase Dependent Control of Protozoan Cyst Burden in the Brain

J. Philip Nance; Kevin M. Vannella; Danielle Worth; Clément N. David; David Carter; Shahani Noor; Cedric Hubeau; Lori Fitz; Thomas E. Lane; Thomas A. Wynn; Emma H. Wilson

Chronic infections represent a continuous battle between the hosts immune system and pathogen replication. Many protozoan parasites have evolved a cyst lifecycle stage that provides it with increased protection from environmental degradation as well as endogenous host mechanisms of attack. In the case of Toxoplasma gondii, these cysts are predominantly found in the immune protected brain making clearance of the parasite more difficult and resulting in a lifelong infection. Currently, little is known about the nature of the immune response stimulated by the presence of these cysts or how they are able to propagate. Here we establish a novel chitinase-dependent mechanism of cyst control in the infected brain. Despite a dominant Th1 immune response during Toxoplasma infection there exists a population of alternatively activated macrophages (AAMØ) in the infected CNS. These cells are capable of cyst lysis via the production of AMCase as revealed by live imaging, and this chitinase is necessary for protective immunity within the CNS. These data demonstrate chitinase activity in the brain in response to a protozoan pathogen and provide a novel mechanism to facilitate cyst clearance during chronic infections.


Analytical Biochemistry | 2010

A fluorescent assay suitable for inhibitor screening and vanin tissue quantification.

Benfang H. Ruan; Derek C. Cole; Paul Wu; Amira Quazi; Karen Page; Jill F. Wright; Nelson Huang; Joseph Stock; Karl Nocka; Ann Aulabaugh; Rustem Krykbaev; Lori Fitz; Neil M. Wolfman; Margaret Fleming

Vanin-1 is a pantetheinase that catalyzes the hydrolysis of pantetheine to produce pantothenic acid (vitamin B5) and cysteamine. Reported here is a highly sensitive fluorescent assay using a novel fluorescently labeled pantothenate derivative. The assay has been used for characterization of a soluble version of human vanin-1 recombinant protein, identification and characterization of hits from high-throughput screening (HTS), and quantification of vanin pantothenase activity in cell lines and tissues. Under optimized assay conditions, we quantified vanin pantothenase activity in tissue lysate and found low activity in lung and liver but high activity in kidney. We demonstrated that the purified recombinant vanin-1 consisting of the extracellular portion without the glycosylphosphatidylinositol (GPI) linker was highly active with an apparent K(m) of 28 microM for pantothenate-7-amino-4-methylcoumarin (pantothenate-AMC), which was converted to pantothenic acid and AMC based on liquid chromatography-mass spectrometry (LC-MS) analysis. The assay also performed well in a 384-well microplate format under initial rate conditions (10% conversion) with a signal-to-background ratio (S/B) of 7 and a Z factor of 0.75. Preliminary screening of a library of 1280 pharmaceutically active compounds identified inhibitors with novel chemical scaffolds. This assay will be a powerful tool for target validation and drug lead identification and characterization.

Collaboration


Dive into the Lori Fitz's collaboration.

Top Co-Authors

Avatar

Debra D. Donaldson

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tamlyn Neben

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge