Network


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

Hotspot


Dive into the research topics where Richard Zollner is active.

Publication


Featured researches published by Richard Zollner.


Journal of Immunology | 2000

Cutting Edge: Identification of GL50, a Novel B7-Like Protein That Functionally Binds to ICOS Receptor

Vincent Ling; Paul W. Wu; Heather Finnerty; Kevin M. Bean; Vicki Spaulding; Lynette A. Fouser; John P. Leonard; Sharon E. Hunter; Richard Zollner; Jenifer L. Thomas; Joy S. Miyashiro; Kenneth A. Jacobs; Mary Collins

By the genetic selection of mouse cDNAs encoding secreted proteins, a B7-like cDNA clone termed mouse GL50 (mGL50) was isolated encoding a 322-aa polypeptide identical with B7h. Isolation of the human ortholog of this cDNA (hGL50) revealed a coding sequence of 309 aa residues with 42% sequence identity with mGL50. Northern analysis indicated GL50 to be present in many tissues including lymphoid, embryonic yolk sac, and fetal liver samples. Of the CD28, CTLA4, and ICOS fusion constructs tested, flow cytometric analysis demonstrated only mouse ICOS-IgG binding to mGL50 cell transfectants. Subsequent phenotyping demonstrated high levels of ICOS ligand staining on splenic CD19+ B cells and low levels on CD3+ T cells. These results indicate that GL50 is a specific ligand for the ICOS receptor and suggest that the GL50-ICOS interaction functions in lymphocyte costimulation.


Arthritis & Rheumatism | 2009

Prevention of Cartilage Degeneration in a Rat Model of Osteoarthritis by Intraarticular Treatment With Recombinant Lubricin

Carl R. Flannery; Richard Zollner; Chris Corcoran; Aled R.C. Jones; Adam Root; Moisés A. Rivera-Bermúdez; Tracey Blanchet; Jason P. Gleghorn; Lawrence J. Bonassar; Alison Bendele; E.A. Morris; S.S. Glasson

OBJECTIVE Lubricin, also referred to as superficial zone protein and PRG4, is a synovial glycoprotein that supplies a friction-resistant, antiadhesive coating to the surfaces of articular cartilage, thereby protecting against arthritis-associated tissue wear and degradation. This study was undertaken to generate and characterize a novel recombinant lubricin protein construct, LUB:1, and to evaluate its therapeutic efficacy following intraarticular delivery in a rat model of osteoarthritis (OA). METHODS Binding and localization of LUB:1 to cartilage surfaces was assessed by immunohistochemistry. The cartilage-lubricating properties of LUB:1 were determined using a custom friction testing apparatus. A cell-binding assay was performed to quantify the ability of LUB:1 to prevent cell adhesion. Efficacy studies were conducted in a rat meniscal tear model of OA. One week after the surgical induction of OA, LUB:1 or phosphate buffered saline vehicle was administered by intraarticular injection for 4 weeks, with dosing intervals of either once per week or 3 times per week. OA pathology scores were determined by histologic analysis. RESULTS LUB:1 was shown to bind effectively to cartilage surfaces, and facilitated both cartilage boundary lubrication and inhibition of synovial cell adhesion. Treatment of rat knee joints with LUB:1 resulted in significant disease-modifying, chondroprotective effects during the progression of OA, by markedly reducing cartilage degeneration and structural damage. CONCLUSION Our findings demonstrate the potential use of recombinant lubricin molecules in novel biotherapeutic approaches to the treatment of OA and associated cartilage abnormalities.


Protein Science | 2008

Crystal structures of the two major aggrecan degrading enzymes, ADAMTS4 and ADAMTS5

Lidia Mosyak; Katy E. Georgiadis; Tania Shane; Kristine Svenson; Tracy Hebert; Thomas McDonagh; Stewart Mackie; Stephane Olland; Laura Lin; Xiaotian Zhong; Ronald W. Kriz; Erica Reifenberg; Lisa A. Collins-Racie; Christopher John Corcoran; Bethany A. Freeman; Richard Zollner; Tod Marvell; Matthew Vera; Phaik-Eng Sum; Edward R. Lavallie; Mark Stahl; William Stuart Somers

Aggrecanases are now believed to be the principal proteinases responsible for aggrecan degradation in osteoarthritis. Given their potential as a drug target, we solved crystal structures of the two most active human aggrecanase isoforms, ADAMTS4 and ADAMTS5, each in complex with bound inhibitor and one wherein the enzyme is in apo form. These structures show that the unliganded and inhibitor‐bound enzymes exhibit two essentially different catalytic‐site configurations: an autoinhibited, nonbinding, closed form and an open, binding form. On this basis, we propose that mature aggrecanases exist as an ensemble of at least two isomers, only one of which is proteolytically active.


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.


The Journal of Infectious Diseases | 2013

Staphylococcal Enterotoxin B–Specific Monoclonal Antibody 20B1 Successfully Treats Diverse Staphylococcus aureus Infections

Avanish K. Varshney; Xiaobo Wang; Matthew D. Scharff; Jennifer MacIntyre; Richard Zollner; Oleg V. Kovalenko; Luis R. Martinez; Fergus R. Byrne; Bettina C. Fries

BACKGROUND Methicillin-resistant Staphylococcus aureus (MRSA) has become a major health threat in the United States. Staphylococcal enterotoxin B (SEB) is a potent superantigen that contributes to its virulence. High mortality and frequent failure of therapy despite available antibiotics have stimulated research efforts to develop adjunctive therapies. METHODS Treatment benefits of SEB-specific monoclonal antibody (mAb) 20B1 were investigated in mice in sepsis, superficial skin, and deep-tissue infection models. RESULTS Mice challenged with a SEB-producing MRSA strain developed fatal sepsis, extensive tissue skin infection, and abscess-forming deep-seeded thigh muscle infection. Animals preimmunized against SEB or treated passively with mAb 20B1 exhibited enhanced survival in the sepsis model, whereas decrease of bacterial burden was observed in the superficial skin and deep-tissue models. mAb 20B1 bound to SEB in the infected tissue and decreased abscess formation and proinflammatory cytokine levels, lymphocyte proliferation, and neutrophil recruitment. CONCLUSIONS mAb 20B1, an SEB-neutralizing mAb, is effective against MRSA infection. mAb 20B1 protects against lethal sepsis and reduces skin tissue invasion and deep-abscess formation. The mAb penetrates well into the abscess and binds to SEB. It affects the outcome of S. aureus infection by modulating the hosts proinflammatory immune response.


FEBS Journal | 2009

Oligomerization is required for the activity of recombinant soluble LOX‐1

Wei Cao; Valerie Calabro; Adam R. Root; Grace Yan; Khetemenee Lam; Stephane Olland; Jocelyn Sanford; Angela Robak; Richard Zollner; Zhijian Lu; Mostafa Ait-Zahra; Rita Agostinelli; Lioudmila Tchistiakova; Davinder Gill; Douglas Harnish; Janet E. Paulsen; Heather H. Shih

LOX‐1 is a scavenger receptor that functions as the primary receptor for oxidized low‐density lipoprotein (OxLDL) in endothelial cells. The binding of OxLDL to LOX‐1 is believed to lead to endothelial activation, dysfunction, and injury, which constitute early atherogenic events. Because of its potential pathological role in atherosclerosis, LOX‐1 has been proposed as a therapeutic target for the treatment of this disease. In order to antagonize the ligand‐binding function of cell surface LOX‐1, we generated a series of recombinant human LOX‐1–crystallizable fragment (Fc) fusion proteins and subsequently characterized their biochemical properties and ligand‐binding activities in vitro. Consistent with the notion that oligomerization of cell surface LOX‐1 is required for high‐avidity binding of ligands, we found that LOX‐1–Fc fusion protein containing four ligand‐binding domains per Fc dimer, but not the one containing two ligand‐binding domains, exhibited ligand‐binding activity. Optimal ligand‐binding activity could be achieved via crosslinking of LOX‐1–Fc fusion proteins with a polyclonal antibody against Fc. The crosslinked LOX‐1–Fc protein also effectively inhibited the binding and internalization of OxLDL by cell surface LOX‐1. These findings demonstrate that functional oligomerization is required for recombinant LOX‐1–Fc to function as an effective antagonist.


The Journal of Infectious Diseases | 2014

Humanized Staphylococcal Enterotoxin B (SEB)–Specific Monoclonal Antibodies Protect From SEB Intoxication and Staphylococcus aureus Infections Alone or as Adjunctive Therapy With Vancomycin

Avanish K. Varshney; Xiaobo Wang; Jennifer MacIntyre; Richard Zollner; Kerry S. Kelleher; Oleg V. Kovalenko; Ximo Pechuan; Fergus R. Byrne; Bettina C. Fries

BACKGROUND Staphylococcal enterotoxin B (SEB), a potential biological warfare agent, is a potent superantigen that contributes to the virulence of methicillin-resistant Staphylococcus aureus (MRSA), which is a major health threat in the United States. Efforts to develop toxin-neutralizing antibodies as adjunctive therapies are justified, given the high mortality and frequent failure of therapy despite available antibiotics. METHODS Murine SEB-specific mAb 20B1 was humanized, and treatment benefits of Hu-1.6/1.1 and Hu-1.4/1.1 variants were investigated in mice in an SEB intoxication model, as well as in sepsis and deep-tissue infection models. RESULTS Hu-1.6/1.1 and Hu-1.4/1.1 protected mice against SEB-induced lethal shock. Hu-1.6/1.1 also enhanced survival of mice that developed fatal sepsis after challenge with a SEB-producing MRSA strain. Combined treatment of Hu-1.6/1.1 with vancomycin further increased survival and altered cytokine responses, compared with monotherapy with either monoclonal antibody or vancomycin alone. Efficacy was also demonstrated in the deep-tissue infection model, where Hu-1.4/1.1 bound to SEB in vivo and decreased abscess formation, as well as proinflammatory cytokine levels. CONCLUSIONS SEB-neutralizing mAb 20B1 was successfully humanized. The mAb affects outcome by modulating the proinflammatory host response in both the sepsis and the intoxication models, which justifies further development.


Rheumatology | 2011

Distinct in vitro binding properties of the anti-CD20 small modular immunopharmaceutical 2LM20-4 result in profound and sustained in vivo potency in cynomolgus monkeys

Cheryl Nickerson-Nutter; Lioudmila Tchistiakova; Nilufer Seth; Marion Kasaian; Barbara Sibley; Stephane Olland; Richard Zollner; William A. Brady; Kendall M. Mohler; Peter Robert Baum; Alan Wahl; Deborah Herber; Yulia Vugmeyster; David Wensel; Neil M. Wolfman; Davinder Gill; Mary Collins; Kyri Dunussi-Joannopoulos

Objectives. To characterize the in vitro binding and effector function properties of CD20-directed small modular immunopharmaceutical (SMIP) 2LM20-4, and to compare its in vivo B-cell depletion activity with the mutated 2LM20-4 P331S [no in vitro complement-dependent cytotoxicity (CDC)] and rituximab in cynomolgus monkeys. Methods. Direct binding is examined in flow cytometry, confocal microscopy, scatchard and lipid raft assays. Effector function assays include CDC and Fc-mediated cellular toxicity. In the 6-month-long in vivo B-cell depletion study, single i.v. dosages of 1 or 10 mg/kg of anti-CD20 proteins were administered to monkeys and B-cell counts were monitored in peripheral blood, bone marrow and lymph nodes. Results. 2LM20-4 has lower saturation binding to human primary B cells and recruits fewer CD20 molecules into lipid rafts compared with rituximab; however, it induces higher in vitro CDC. In competitive binding, 2LM20-4 only partially displaces rituximab, suggesting that it binds to a fraction of CD20 molecules within certain locations of the plasma membrane as compared with rituximab. In monkeys, 2LM20-4 had more sustained B-cell depletion activity than rituximab in peripheral blood and had significantly more profound and sustained activity than 2LM20-4 P331S and rituximab in the lymph nodes. Conclusions. SMIP 2LM20-4, which binds to a fraction of CD20 molecules as compared with rituximab, has more potent in vitro CDC, and more potent and sustained B-cell depletion activity in cynomolgus monkeys. Our work has considerable clinical relevance since it provides novel insights related to the emerging B-cell depletion therapies in autoimmune diseases.


Journal of Biotechnology | 2017

Mechanistic understanding of the cysteine capping modifications of antibodies enables selective chemical engineering in live mammalian cells

Xiaotian Zhong; Tao He; Amar S. Prashad; Wenge Wang; Justin Cohen; Darren Ferguson; Amy Tam; Eric Sousa; Laura Lin; Lioudmila Tchistiakova; Scott Gatto; Aaron M. D’Antona; Yen-Tung Luan; Weijun Ma; Richard Zollner; Jing Zhou; Bo Arve; Will Somers; Ronald Kriz

Protein modifications by intricate cellular machineries often redesign the structure and function of existing proteins to impact biological networks. Disulfide bond formation between cysteine (Cys) pairs is one of the most common modifications found in extracellularly-destined proteins, key to maintaining protein structure. Unpaired surface cysteines on secreted mammalian proteins are also frequently found disulfide-bonded with free Cys or glutathione (GSH) in circulation or culture, the mechanism for which remains unknown. Here we report that these so-called Cys-capping modifications take place outside mammalian cells, not in the endoplasmic reticulum (ER) where oxidoreductase-mediated protein disulfide formation occurs. Unpaired surface cysteines of extracellularly-arrived proteins such as antibodies are uncapped upon secretion before undergoing disulfide exchange with cystine or oxidized GSH in culture medium. This observation has led to a feasible way to selectively modify the nucleophilic thiol side-chain of cell-surface or extracellular proteins in live mammalian cells, by applying electrophiles with a chemical handle directly into culture medium. These findings provide potentially an effective approach for improving therapeutic conjugates and probing biological systems.


International Immunopharmacology | 2015

Optimization of experimental conditions for functional in vitro characterization of humanized antibodies specific for staphylococcal enterotoxin B.

Jennifer MacIntyre; Avanish K. Varshney; Xiaobo Wang; Scott Gatto; Clarence Friedman; Yan Liu; Kelvin M. Kerns; Oleg V. Kovalenko; Karissa Adkins; Richard Zollner; Bettina C. Fries; Fergus R. Byrne

Staphylococcus aureus is a common nosocomial infection and its resistance to penicillin and methicillin antibiotics is a growing clinical problem. We previously described the development of a humanized anti-Staphylococcus enterotoxin B (SEB) antibody derived from the mouse antibody made by the 20B1 hybridoma. This antibody was humanized and characterized kinetically by surface plasmon resonance demonstrating that the humanized clones retained binding to SEB. Clones were then functionally characterized in an in vitro assay demonstrating that the murine 20B1, chimeric and humanized antibodies potently inhibited SEB-induced murine splenocyte proliferation assay. Here, we describe a human cell-based screening assay, optimized by varying multiple experimental parameters that resulted in an assay that was used to demonstrate full and potent neutralization by the parental, chimeric and humanized antibodies. The replacement of fetal bovine serum (FBS) with normal human serum (NHS) was found to be a crucial factor in the performance of the human cell based screening assay that enabled the calculation of mAb efficacy and potency. In addition, we found that anti-SEB antibodies showed similar efficacy and potency with a triple mutant Fc region (designed to be effector function null) or a wild-type Fc region, which is in contrast to previously described studies.

Collaboration


Dive into the Richard Zollner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Avanish K. Varshney

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Bettina C. Fries

Albert Einstein College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge