Network


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

Hotspot


Dive into the research topics where Patricia M. Davis is active.

Publication


Featured researches published by Patricia M. Davis.


International Immunopharmacology | 2001

Stimulation of inflammatory responses in vitro and in vivo by lipophilic HMG-CoA reductase inhibitors

Peter A. Kiener; Patricia M. Davis; Judy L Murray; Sonia Youssef; Bruce M. Rankin; Mark C. Kowala

The enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase catalyses the rate limiting step in cholesterol biosynthesis and is markedly inhibited by the statin family of drugs. The effect of statins on lipid lowering is clearly defined, but the ability of the drugs to directly regulate inflammatory functions has not been well explored. In this report, we show that there are differences among the statins in their capacity to induce proinflammatory responses both in human monocytes in vitro, and in leukocytes in mice in vivo. Treatment of human monocytes with lipophilic statins alone stimulated the production of MCP-1, IL-8, TNF-alpha and IL-1 beta and markedly sensitized the cells to subsequent challenge with inflammatory agents. Lipophilic statins also increased the production of reactive oxygen species in monocytes. In contrast, pretreatment of cells with the hydrophilic pravastatin did not induce these heightened inflammatory responses. Furthermore, treatment of mice with lipophilic statins caused a markedly higher influx of leukocytes into the inflamed peritoneal cavity following challenge with thioglycollate. Overall, these results demonstrate that the lipophilic statins influence a regulatory pathway in monocytes that controls cytokine production and that the statins induce different pro-inflammatory responses both in vitro and in vivo.


Transplantation | 2009

Assessment of belatacept-mediated costimulation blockade through evaluation of CD80/86-receptor saturation.

Robert Latek; Catherine A. Fleener; Vahideh Lamian; Edward Kulbokas; Patricia M. Davis; Suzanne J. Suchard; Mark E. Curran; Flavio Vincenti; Robert Townsend

Background. The selective inhibitor of T-cell costimulation, belatacept, blocks CD28-mediated T-cell activation by binding CD80 and CD86 on antigen-presenting cells. Understanding the extent to which belatacept binds to its targets in patients may enable correlation of belatacept exposure to receptor saturation as a pharmacodynamic measure of costimulation blockade. Methods. Flow cytometry-based receptor competition assays were developed to monitor concentration-dependent occupancy of CD80 and CD86 receptors in whole blood and dendritic cell cultures in vitro. Receptor occupancy was correlated with inhibition of mixed leukocyte reactions and clinical validation was obtained by comparing receptor saturation in whole blood from healthy volunteers and in de novo renal transplant recipients participating in studies comparing cyclosporine and belatacept-based immunosuppression. Results. Belatacept saturated CD80 and CD86 receptors in whole blood and dendritic cell cultures, although the belatacept concentrations required for CD86-receptor saturation were approximately 10-fold higher than those required for CD80 saturation (IC50=0.102 &mgr;g/mL vs. 0.009 &mgr;g/mL). Primary alloresponses were inhibited at the belatacept concentration required for CD86-receptor saturation, but not at the lower concentration needed to saturate CD80. Whole blood from belatacept-treated patients had significantly lower levels of free CD86 receptors versus pretransplant levels, healthy volunteers, or cyclosporine-treated patients. CD86-receptor saturation correlated with belatacept dose/dose frequency and remained consistently more than 80%. Conclusions. These results suggest that belatacept-mediated inhibition of alloresponses involved in transplant rejection correlates with CD86 saturation, indicating that CD86-receptor occupancy may be a valid pharmacodynamic measure of costimulation blockade and provide the first direct clinical evidence that belatacept binds to one of its targets.


Journal of Immunology | 2013

A Monovalent Anti-Human CD28 Domain Antibody Antagonist: Preclinical Efficacy and Safety

Suzanne J. Suchard; Patricia M. Davis; Selena Kansal; Dawn K. Stetsko; Ruth Brosius; James Tamura; Lumelle A. Schneeweis; James W. Bryson; Theodora W. Salcedo; Haiqing Wang; Zheng Yang; Catherine A. Fleener; Olga Ignatovich; Christopher Plummer; Steven Grant; Steven G. Nadler

Targeting the CD28-CD80/86 pathway with an anti-CD28 antagonist is a promising alternative to current therapies for autoimmunity. However, attempts at generating conventional anti-CD28 mAbs lacking stimulatory activity has been challenging. In this study, we describe anti-human CD28 receptor antagonist domain Abs (dAbs) that are specific for human CD28. These dAbs are potent inhibitors of T cell activation, with an EC50 of 35 ± 14 ng/ml for inhibition of proliferation. The EC50 of 53 ± 11 ng/ml in an ex vivo CD28 receptor occupancy assay corresponds with in vitro functional activity, suggesting a direct correlation. The anti-CD28 dAb is equipotent in the inhibition of CD80- and CD86-mediated T cell proliferation and does not interfere with CTLA-4–mediated downmodulation of CD86 expression on APCs. The anti-CD28 dAbs are monomeric and do not demonstrate any evidence of agonism or costimulatory activity. In cynomolgus monkeys, the anti-CD28 dAb demonstrated pharmacodynamic activity, as measured by the inhibition of a T cell–dependent Ab response, without evidence of T cell depletion or cytokine release. Furthermore, there was a strong correlation between systemic exposure, duration, and extent of CD28 receptor occupancy, and pharmacodynamic activity. Taken together, these data support clinical evaluation of this novel anti-CD28 dAb for the treatment of autoimmune diseases.


Journal of Immunology | 2010

An LFA-1 (αLβ2) Small-Molecule Antagonist Reduces Inflammation and Joint Destruction in Murine Models of Arthritis

Suzanne J. Suchard; Dawn K. Stetsko; Patricia M. Davis; Stacey Skala; Dominique Potin; Michele Launay; T. G. Murali Dhar; Joel C. Barrish; Vojkan Susulic; David J. Shuster; Kim W. McIntyre; Murray McKinnon; Luisa Salter-Cid

LFA-1 appears to play a central role in normal immune responses to foreign Ags. In autoimmune or inflammatory diseases, there is increased expression of LFA-1 and/or its counterligand, ICAM-1. Others have demonstrated that the targeted disruption of LFA-1:ICAM interactions, either by gene deletion or Ab treatment in mice, results in reduced leukocyte trafficking, inflammatory responses, and inhibition of inflammatory arthritis in the K/BxN serum transfer model. However, there has been little success in finding a small-molecule LFA-1 antagonist that can similarly impact rodent models of arthritis. In this paper, we present the first reported example of an LFA-1 small-molecule antagonist, BMS-587101, that is efficacious in preclinical disease models. In vitro, BMS-587101 inhibited LFA-1–mediated adhesion of T cells to endothelial cells, T cell proliferation, and Th1 cytokine production. Because BMS-587101 exhibits in vitro potency, cross-reactivity, and oral bioavailability in rodents, we evaluated the impact of oral administration of this compound in two different models of arthritis: Ab-induced arthritis and collagen-induced arthritis. Significant impact of BMS-587101 on clinical score in both models was observed, with inhibition comparable or better than anti-mouse LFA-1 Ab. In addition, BMS-587101 significantly reduced cytokine mRNA levels in the joints of Ab-induced arthritis animals as compared with those receiving vehicle alone. In paws taken from the collagen-induced arthritis study, the bones of vehicle-treated mice had extensive inflammation and bone destruction, whereas treatment with BMS-587101 resulted in marked protection. These findings support the potential use of an LFA-1 small-molecule antagonist in rheumatoid arthritis, with the capacity for disease modification.


JCI insight | 2016

B cells from African American lupus patients exhibit an activated phenotype

Laurence C. Menard; Sium Habte; Waldemar Gonsiorek; Deborah Lee; Dana Banas; Deborah A. Holloway; Nataly Manjarrez-Orduño; Mark D. Cunningham; Dawn K. Stetsko; Francesca Casano; Selena Kansal; Patricia M. Davis; Julie Carman; Clarence K. Zhang; Ferva Abidi; Richard A. Furie; Steven G. Nadler; Suzanne J. Suchard

Systemic lupus erythematosus (SLE) is a complex systemic autoimmune disease driven by both innate and adaptive immune cells. African Americans tend to present with more severe disease at an earlier age compared with patients of European ancestry. In order to better understand the immunological differences between African American and European American patients, we analyzed the frequencies of B cell subsets and the expression of B cell activation markers from a total of 68 SLE patients and 69 normal healthy volunteers. We found that B cells expressing the activation markers CD86, CD80, PD1, and CD40L, as well as CD19+CD27-IgD- double-negative B cells, were enriched in African American patients vs. patients of European ancestry. In addition to increased expression of CD40L, surface levels of CD40 on B cells were lower, suggesting the engagement of the CD40 pathway. In vitro experiments confirmed that CD40L expressed by B cells could lead to CD40 activation and internalization on adjacent B cells. To conclude, these results indicate that, compared with European American patients, African American SLE patients present with a particularly active B cell component, possibly via the activation of the CD40/CD40L pathway. These data may help guide the development of novel therapies.


Journal of Experimental Medicine | 1997

Differential Induction of Apoptosis by Fas–Fas Ligand Interactions in Human Monocytes and Macrophages

Peter A. Kiener; Patricia M. Davis; Gary C. Starling; Christopher Mehlin; Seymour J. Klebanoff; Jeffrey A. Ledbetter; W. Conrad Liles


Journal of Immunology | 1997

Human monocytic cells contain high levels of intracellular Fas ligand: rapid release following cellular activation.

Peter A. Kiener; Patricia M. Davis; Bruce M. Rankin; Seymour J. Klebanoff; Jeffrey A. Ledbetter; Gary C. Starling; W C Liles


The Journal of Rheumatology | 2007

Abatacept binds to the Fc receptor CD64 but does not mediate complement-dependent cytotoxicity or antibody-dependent cellular cytotoxicity.

Patricia M. Davis; Ralph Abraham; Linda Xu; Steven G. Nadler; Suzanne J. Suchard


Clinical Immunology | 2008

Abatacept modulates human dendritic cell-stimulated T-cell proliferation and effector function independent of IDO induction

Patricia M. Davis; Steven G. Nadler; Dawn K. Stetsko; Suzanne J. Suchard


Arteriosclerosis, Thrombosis, and Vascular Biology | 1995

Immune Complexes of LDL Induce Atherogenic Responses in Human Monocytic Cells

Peter A. Kiener; Bruce M. Rankin; Patricia M. Davis; Sue A. Yocum; Glenn Warr; Robert I. Grove

Collaboration


Dive into the Patricia M. Davis'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge