Network


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

Hotspot


Dive into the research topics where Maya Arai is active.

Publication


Featured researches published by Maya Arai.


Journal of Biological Chemistry | 2006

Protein Kinase Cζ Is Up-regulated in Osteoarthritic Cartilage and Is Required for Activation of NF-κB by Tumor Necrosis Factor and Interleukin-1 in Articular Chondrocytes

Edward R. Lavallie; Priya S. Chockalingam; Lisa A. Collins-Racie; Bethany A. Freeman; Cristin Keohan; Michael Leitges; Andrew J. Dorner; Elisabeth A. Morris; Manas K. Majumdar; Maya Arai

Protein kinase Cζ (PKCζ) is an intracellular serine/threonine protein kinase that has been implicated in the signaling pathways for certain inflammatory cytokines, including interleukin-1 (IL-1) and tumor necrosis factor α (TNF-α), in some cell types. A study of gene expression in articular chondrocytes from osteoarthritis (OA) patients revealed that PKCζ is transcriptionally up-regulated in human OA articular cartilage clinical samples. This finding led to the hypothesis that PKCζ may be an important signaling component of cytokine-mediated cartilage matrix destruction in articular chondrocytes, believed to be an underlying factor in the pathophysiology of OA. IL-1 treatment of chondrocytes in culture resulted in rapidly increased phosphorylation of PKCζ, implicating PKCζ activation in the signaling pathway. Chondrocyte cell-based assays were used to evaluate the contribution of PKCζ activity in NF-κB activation and extracellular matrix degradation mediated by IL-1, TNF, or sphingomyelinase. In primary chondrocytes, IL-1 and TNF-α caused an increase in NF-κB activity resulting in induction of aggrecanase-1 and aggrecanase-2 expression, with consequent increased proteoglycan degradation. This effect was blocked by the pan-specific PKC inhibitors RO 31-8220 and bisindolylmaleimide I, partially blocked by Gö 6976, and was unaffected by the PKCζ-sparing inhibitor calphostin C. A cell-permeable PKCζ pseudosubstrate peptide inhibitor was capable of blocking TNFand IL-1-mediated NF-κB activation and proteoglycan degradation in chondrocyte pellet cultures. In addition, overexpression of a dominant negative PKCζ protein effectively prevented cytokine-mediated NF-κB activation in primary chondrocytes. These data implicate PKCζ as a necessary component of the IL-1 and TNF signaling pathways in chondrocytes that result in catabolic destruction of extracellular matrix proteins in osteoarthritic cartilage.


Journal of Biological Chemistry | 2012

An ultra-specific avian antibody to phosphorylated tau protein reveals a unique mechanism for phosphoepitope recognition.

Heather H. Shih; Chao Tu; Wei Cao; Anne S. Klein; Renee Ramsey; Brian J. Fennell; Matthew Lambert; Deirdre Ní Shúilleabháin; Bénédicte Autin; Eugenia Kouranova; Sri Laxmanan; Steven P. Braithwaite; Leeying Wu; Mostafa Ait-Zahra; Anthony J. Milici; Jo Ann Dumin; Edward R. LaVallie; Maya Arai; Christopher John Corcoran; Janet E. Paulsen; Davinder Gill; Orla Cunningham; Joel Bard; Lydia Mosyak; William J. J. Finlay

Background: Truly phosphospecific antibodies are difficult to generate and are poorly understood. Results: Avian single chain Fv library selections yielded fully phosphospecific anti-phospho-tau antibodies, enabling the generation of a 1.9 Å co-crystal structure. Conclusion: Phosphospecific antibodies were readily generated and can exhibit unique epitope recognition mechanisms. Significance: High-affinity antibody phosphoepitope recognition has been defined, at high resolution, for the first time. Highly specific antibodies to phosphoepitopes are valuable tools to study phosphorylation in disease states, but their discovery is largely empirical, and the molecular mechanisms mediating phosphospecific binding are poorly understood. Here, we report the generation and characterization of extremely specific recombinant chicken antibodies to three phosphoepitopes on the Alzheimer disease-associated protein tau. Each antibody shows full specificity for a single phosphopeptide. The chimeric IgG pT231/pS235_1 exhibits a KD of 0.35 nm in 1:1 binding to its cognate phosphopeptide. This IgG is murine ortholog-cross-reactive, specifically recognizing the pathological form of tau in brain samples from Alzheimer patients and a mouse model of tauopathy. To better understand the underlying binding mechanisms allowing such remarkable specificity, we determined the structure of pT231/pS235_1 Fab in complex with its cognate phosphopeptide at 1.9 Å resolution. The Fab fragment exhibits novel complementarity determining region (CDR) structures with a “bowl-like” conformation in CDR-H2 that tightly and specifically interacts with the phospho-Thr-231 phosphate group, as well as a long, disulfide-constrained CDR-H3 that mediates peptide recognition. This binding mechanism differs distinctly from either peptide- or hapten-specific antibodies described to date. Surface plasmon resonance analyses showed that pT231/pS235_1 binds a truly compound epitope, as neither phosphorylated Ser-235 nor free peptide shows any measurable binding affinity.


Shock | 2011

A monoclonal antibody against RAGE alters gene expression and is protective in experimental models of sepsis and pneumococcal pneumonia.

Eirini Christaki; Steven M. Opal; James C. Keith; Nubar Kessimian; John E. Palardy; Nicolas A. Parejo; Xiang Yang Tan; Nicole Piche-Nicholas; Lioudmila Tchistiakova; George P. Vlasuk; Kathleen M. Shields; Jeffrey L. Feldman; Edward R. Lavallie; Maya Arai; William Mounts; Debra D. Pittman

The RAGE (receptor for advanced glycation end products) is believed to play a role in sepsis by perpetuating inflammation. The interaction of RAGE with a variety of host-derived ligands that accumulate during stress and inflammation further induces the expression of RAGE. It was previously shown that a rat anti-RAGE monoclonal antibody protected mice from lethality in a cecal ligation and puncture model. We studied the effects of a humanized anti-RAGE monoclonal antibody in the murine pneumococcal pneumonia model of sepsis. Moreover, a gene expression analysis was performed in lung tissue of animals that underwent cecal ligation and puncture and treated with the rat anti-RAGE monoclonal antibody, compared with controls. Administration of humanized anti-RAGE mAb 6 h after intratracheal infection with Streptococcus pneumoniae improved mortality in BALB/c mice whether a 7.5 mg/kg (P < 0.01) or a 15 mg/kg dose (P < 0.01) was administered in combination with antibiotics. Gene expression analysis showed that many of the genes modulated by treatment with the anti-RAGE antibody were those that play an important role in regulating inflammation. Anti-RAGE monoclonal antibody offered a survival advantage to septic mice. This protective role in treated animals is supported by the observed gene expression profile changes of genes involved in sepsis and inflammation.


Osteoarthritis and Cartilage | 2009

Global analysis of nuclear receptor expression and dysregulation in human osteoarthritic articular cartilage: Reduced LXR signaling contributes to catabolic metabolism typical of osteoarthritis

Lisa A. Collins-Racie; Z. Yang; Maya Arai; N. Li; M.K. Majumdar; S. Nagpal; W.M. Mounts; A.J. Dorner; Elisabeth A. Morris; Edward R. Lavallie

OBJECTIVE Compare the expression and regulation of nuclear receptors (NRs) in osteoarthritic and normal human articular cartilage. METHOD The transcriptional levels of 48 NRs and additional related proteins were measured in mRNA from human articular cartilage from subjects with osteoarthritis (OA) and compared to samples from subjects without OA, using microarrays, individual quantitative reverse transcriptase polymerase chain reaction assays, and a custom human NR TaqMan Low Density Array (TLDA). The functional effect of liver X receptor (LXR) activity in cartilage was studied by measuring proteoglycan (PG) synthesis and degradation in articular cartilage explant cultures following treatment with the synthetic LXR agonist T0901317. RESULTS Thirty-one of 48 NRs analyzed by TLDA were found to be measurably expressed in human articular cartilage; 23 of these 31 NRs showed significantly altered expression in OA vs unaffected cartilage. Among these, LXRalpha and LXRbeta, and their heterodimeric partners retinoid X receptor (RXR)alpha and RXRbeta were all expressed at significantly lower levels in OA cartilage, as were LXR target genes ABCG1 and apolipoproteins D and E. Addition of LXR agonist to human OA articular chondrocytes and to cartilage explant cultures resulted in activation of LXR-mediated transcription and significant reduction of both basal and interleukin (IL)-1-mediated PG degradation. CONCLUSIONS Articular cartilage expresses a substantial number of NRs, and a large proportion of the expressed NRs are dysregulated in OA. In particular, LXR signaling in OA articular cartilage is impaired, and stimulation of LXR transcriptional activity can counteract the catabolic effects of IL-1. We conclude that LXR agonism may be a possible therapeutic option for OA.


Journal of Translational Medicine | 2008

Discovery and implementation of transcriptional biomarkers of synthetic LXR agonists in peripheral blood cells

Elizabeth DiBlasio-Smith; Maya Arai; Elaine Quinet; Mark J Evans; Tad Kornaga; Michael D Basso; Liang Chen; Irene Feingold; Anita R Halpern; Qiang-Yuan Liu; Ponnal Nambi; Dawn Savio; Shuguang Wang; William Mounts; Jennifer A. Isler; Anna M. Slager; Michael E. Burczynski; Andrew J. Dorner; Edward R. Lavallie

BackgroundLXRs (Liver X Receptor α and β) are nuclear receptors that act as ligand-activated transcription factors. LXR activation causes upregulation of genes involved in reverse cholesterol transport (RCT), including ABCA1 and ABCG1 transporters, in macrophage and intestine. Anti-atherosclerotic effects of synthetic LXR agonists in murine models suggest clinical utility for such compounds.ObjectiveBlood markers of LXR agonist exposure/activity were sought to support clinical development of novel synthetic LXR modulators.MethodsTranscript levels of LXR target genes ABCA1 and ABCG1 were measured using quantitative reverse transcriptase/polymerase chain reaction assays (qRT-PCR) in peripheral blood from mice and rats (following a single oral dose) and monkeys (following 7 daily oral doses) of synthetic LXR agonists. LXRα, LXRβ, ABCA1, and ABCG1 mRNA were measured by qRT-PCR in human peripheral blood mononuclear cells (PBMC), monocytes, T- and B-cells treated ex vivo with WAY-252623 (LXR-623), and protein levels in human PBMC were measured by Western blotting. ABCA1/G1 transcript levels in whole-blood RNA were measured using analytically validated assays in human subjects participating in a Phase 1 SAD (Single Ascending Dose) clinical study of LXR-623.ResultsA single oral dose of LXR agonists induced ABCA1 and ABCG1 transcription in rodent peripheral blood in a dose- and time-dependent manner. Induction of gene expression in rat peripheral blood correlated with spleen expression, suggesting LXR gene regulation in blood has the potential to function as a marker of tissue gene regulation. Transcriptional response to LXR agonist was confirmed in primates, where peripheral blood ABCA1 and ABCG1 levels increased in a dose-dependent manner following oral treatment with LXR-623. Human PBMC, monocytes, T- and B cells all expressed both LXRα and LXRβ, and all cell types significantly increased ABCA1 and ABCG1 expression upon ex vivo LXR-623 treatment. Peripheral blood from a representative human subject receiving a single oral dose of LXR-623 showed significant time-dependent increases in ABCA1 and ABCG1 transcription.ConclusionPeripheral blood cells express LXRα and LXRβ, and respond to LXR agonist treatment by time- and dose-dependently inducing LXR target genes. Transcript levels of LXR target genes in peripheral blood are relevant and useful biological indicators for clinical development of synthetic LXR modulators.


Antibodies | 2016

Development of PF-06671008, a Highly Potent Anti-P-cadherin/Anti-CD3 Bispecific DART Molecule with Extended Half-Life for the Treatment of Cancer

Adam R. Root; Wei Cao; Bilian Li; Peter LaPan; Caryl Meade; Jocelyn Sanford; Macy Jin; Cliona O’Sullivan; Emma Cummins; Matthew Lambert; Alfredo Darmanin Sheehan; Weijun Ma; Scott Gatto; Kelvin M. Kerns; Khetemenee Lam; Aaron M. D’Antona; Lily Zhu; William A. Brady; Susan Benard; Amy King; Tao He; Lisa A. Racie; Maya Arai; Dianah Barrett; Wayne Stochaj; Edward R. Lavallie; James R. Apgar; Kristine Svenson; Lidia Mosyak; Yinhua Yang

Bispecific antibodies offer a promising approach for the treatment of cancer but can be challenging to engineer and manufacture. Here we report the development of PF-06671008, an extended-half-life dual-affinity re-targeting (DART®) bispecific molecule against P-cadherin and CD3 that demonstrates antibody-like properties. Using phage display, we identified anti-P-cadherin single chain Fv (scFv) that were subsequently affinity-optimized to picomolar affinity using stringent phage selection strategies, resulting in low picomolar potency in cytotoxic T lymphocyte (CTL) killing assays in the DART format. The crystal structure of this disulfide-constrained diabody shows that it forms a novel compact structure with the two antigen binding sites separated from each other by approximately 30 Å and facing approximately 90° apart. We show here that introduction of the human Fc domain in PF-06671008 has produced a molecule with an extended half-life (~4.4 days in human FcRn knock-in mice), high stability (Tm1 > 68 °C), high expression (>1 g/L), and robust purification properties (highly pure heterodimer), all with minimal impact on potency. Finally, we demonstrate in vivo anti-tumor efficacy in a human colorectal/human peripheral blood mononuclear cell (PBMC) co-mix xenograft mouse model. These results suggest PF-06671008 is a promising new bispecific for the treatment of patients with solid tumors expressing P-cadherin.


Journal of Translational Medicine | 2010

Development and application of a biomarker assay for determining the pharmacodynamic activity of an antagonist candidate biotherapeutic antibody to IL21R in whole blood.

Maya Arai; Sadhana Jain; Amy A Weaver; Andrew A Hill; Yongjing Guo; Andrea G Bree; Michael F Smith; Scott W Allen; Edward R. LaVallie; Deborah Young; Laird Bloom; Karissa Adkins; Margot O'Toole

BackgroundIn preparation for potential clinical development of Ab-01, an antagonistic antibody directed against the IL21R, studies were undertaken to address translational medicine needs that fall into four categories: 1) development of a pharmacodynamic biomarker assay suitable for use in the clinic, 2) demonstration that Ab-01 has the desired biological activity in vitro and in vivo in cynomolgus monkeys, the preferred safety study species, 3) pre-clinical in vivo proof-of-concept that the assay can be used to detect Ab-01 pharmacodynamic (PD) activity in treated subjects, and 4) comprehensive assessment of the agonistic potential of Ab-01 when cross-linked. This report and a recently published companion report address the first three of these needs. The fourth has been addressed in a separate study.MethodsGenes that change RNA expression upon ex vivo rhIL21 stimulation of whole blood were identified in human and cynomolgus monkey. The inhibitory effects of exogenously added Ab-01 were measured ex vivo in human and monkey, and the in vivo inhibitory effects of Ab-01 treatment were measured in monkey.ResultsStimulation of whole human blood for 2 hours with rhIL21 induced robust increases in RNA expression of 6 genes. This response was blocked by Ab-01, indicating that the assay is suitable for measuring Ab-01 activity in blood. rhIL21 induced expression of a similar set of genes in cynomolgus monkey blood. This response was blocked with Ab-01, thus demonstrating that Ab-01 has the desired activity in the species, and that safety studies done in cynomolgus monkeys are relevant. Proof -of-concept for using this assay system to detect PD activity in vivo was generated by measuring the response in monkey blood to ex vivo rhIL21 stimulation before and 5 minutes following in vivo Ab-01 administration.ConclusionsA robust PD biomarker assay suitable for clinical use has been developed in human whole blood. The successful adaptation of the assay to cynomolgus monkeys has enabled the demonstration of Ab-01 activity both in vitro and in vivo in monkey, thus validating the use of this species in safety studies and establishing proof-of-concept for using this PD assay system to aid in dose selection in clinical studies.


Matrix Biology | 2004

ADAMTS-8 exhibits aggrecanase activity and is expressed in human articular cartilage

Lisa A. Collins-Racie; Carl R. Flannery; Weilan Zeng; Christopher John Corcoran; Bethany Annis-Freeman; Michael J. Agostino; Maya Arai; Elizabeth DiBlasio-Smith; Andrew J. Dorner; Katy E. Georgiadis; Macy Jin; Xiang-Yang Tan; Elisabeth A. Morris; Edward R. Lavallie


Osteoarthritis and Cartilage | 2004

Effect of adenovirus-mediated overexpression of bovine ADAMTS-4 and human ADAMTS-5 in primary bovine articular chondrocyte pellet culture system

Maya Arai; Dina Anderson; Yahya Kurdi; Bethany Annis-Freeman; Kathleen M. Shields; Lisa A. Collins-Racie; Christopher John Corcoran; Elizabeth DiBlasio-Smith; Debra D. Pittman; Andrew J. Dorner; Elisabeth A. Morris; Edward R. Lavallie


Archive | 2004

PROTEIN KINASE C ZETA AS A DRUG TARGET FOR ARTHRITIS AND OTHER INFLAMMATORY DISEASES

Edward R. Lavallie; Lisa A. Collins-Racie; Maya Arai

Collaboration


Dive into the Maya Arai's collaboration.

Top Co-Authors

Avatar

Edward R. Lavallie

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge