Network


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

Hotspot


Dive into the research topics where Ed Croze is active.

Publication


Featured researches published by Ed Croze.


Journal of Biological Chemistry | 1999

Catalytically Active TYK2 Is Essential for Interferon-β-mediated Phosphorylation of STAT3 and Interferon-α Receptor-1 (IFNAR-1) but Not for Activation of Phosphoinositol 3-Kinase

M.R. Sandhya Rani; Douglas W. Leaman; Yulong Han; Stewart Leung; Ed Croze; Eleanor N. Fish; Alan Wolfman; Richard M. Ransohoff

TYK2, a Janus kinase, plays both structural and catalytic roles in type I interferon (IFN) signaling. We recently reported (Rani, M. R. S., Gauzzi, C., Pellegrini, S., Fish, E., Wei, T., and Ransohoff, R. M. (1999) J. Biol. Chem. 274, 1891–1897) that catalytically active TYK2 was necessary for IFN-β to induce the β-R1 gene. We now report IFN-β-mediated activation of STATs and other components in U1 (TYK2-null) cell lines that were complemented with kinase-negative (U1.KR930) or wild-type TYK2 (U1.wt). We found that IFN-β induced phosphorylation on tyrosine of STAT3 in U1.wt cells but not in U1.KR930 cells, whereas STAT1 and STAT2 were activated in both cell lines. Additionally, IFN-β-mediated phosphorylation of interferon-α receptor-1 (IFNAR-1) was defective in IFN-β treated U1.KR930 cells, but evident in U1.wt cells. In U1A-derived cells, the p85/p110 phosphoinositol 3-kinase isoform was associated with IFNAR-1 but not STAT3, and the association was ligand-independent. Further, IFN-β treatment stimulated IFNAR-1-associated phosphoinositol kinase activity equally in either U1.wt or U1.KR930 cells. Our results indicate that catalytically active TYK2 is required for IFN-β-mediated tyrosine phosphorylation of STAT3 and IFNAR-1 in intact cells.


Inflammatory Bowel Diseases | 2010

Granulocyte‐macrophage colony‐stimulating factor elicits bone marrow‐derived cells that promote efficient colonic mucosal healing

Eric Bernasconi; Laurent Favre; Michel H. Maillard; Daniel Bachmann; Catherine Pythoud; Hanifa Bouzourene; Ed Croze; Sharlene Velichko; John Parkinson; Pierre Michetti; Dominique Velin

Background: Granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) therapy is effective in treating some Crohns disease (CD) patients and protects mice from colitis induced by dextran sulfate sodium (DSS) administration. However, its mechanisms of action remain elusive. We hypothesized that GM‐CSF affects intestinal mucosal repair. Methods: DSS colitic mice were treated with daily pegylated GM‐CSF or saline and clinical, histological, and inflammatory parameters were kinetically evaluated. Further, the role of bone marrow‐derived cells in the impact of GM‐CSF therapy on DSS colitis was addressed using cell transfers. Results: GM‐CSF therapy reduced clinical signs of colitis and the release of inflammatory mediators. GM‐CSF therapy improved mucosal repair, with faster ulcer reepithelialization, accelerated hyperproliferative response of epithelial cells in ulcer‐adjacent crypts, and lower colonoscopic ulceration scores in GM‐CSF‐administered mice relative to untreated mice. We observed that GM‐CSF‐induced promotion of mucosal repair is timely associated with a reduction in neutrophil numbers and increased accumulation of CD11b+ monocytic cells in colon tissues. Importantly, transfer of splenic GM‐CSF‐induced CD11b+ myeloid cells into DSS‐exposed mice improved colitis, and lethally irradiated GM‐CSF receptor‐deficient mice reconstituted with wildtype bone marrow cells were protected from DSS‐induced colitis upon GM‐CSF therapy. Lastly, GM‐CSF‐induced CD11b+ myeloid cells were shown to promote in vitro wound repair. Conclusions: Our study shows that GM‐CSF‐dependent stimulation of bone marrow‐derived cells during DSS‐induced colitis accelerates colonic tissue repair. These data provide a putative mechanism for the observed beneficial effects of GM‐CSF therapy in Crohns disease. (Inflamm Bowel Dis 2010;)


Journal of Biological Chemistry | 1997

The Short Form of the Interferon α/β Receptor Chain 2 Acts as a Dominant Negative for Type I Interferon Action

Lawrence M. Pfeffer; Leela Basu; Susan R. Pfeffer; Chuan He Yang; Aruna Murti; Dean Russell-Harde; Ed Croze

We have characterized the functional properties of the short form of the human interferon α/β receptor chain 2 (IFNAR2), denoted IFNAR2.1. IFNAR2.1 contains a shortened cytoplasmic domain when compared with the recently cloned full-length IFNAR2 chain (IFNAR2.2). We show that IFNα8 and IFNβ1b induce antiviral and antiproliferative activity in mouse cell transfectants expressing the human IFNAR1 chain of the receptor and induce the formation of STAT1/STAT2 dimers in IFN-stimulated response element (ISRE)-dependent gel shift assays. In contrast, coexpression of IFNAR2.1 with IFNAR1 reduces the IFN-induced antiviral, antiproliferative and ISRE-dependent gel shift binding activity conferred by IFNAR1 alone. No antiviral or antiproliferative response to IFN, nor IFN-induced ISRE-dependent gel shift binding activity, was observed when IFNAR2.1 was expressed alone in murine cells. Therefore, IFNAR2.1 acts as a dominant negative for these IFN-induced activities. Our results suggest that IFNAR2.1 represents a nonfunctional version of the full-length chain (IFNAR2.2).


International Journal of Cancer | 2004

Interferon receptor expression regulates the antiproliferative effects of interferons on cancer cells and solid tumors

T. Charis Wagner; Sharlene Velichko; Steven K. Chesney; Sandra L. Biroc; Dean Harde; David Vogel; Ed Croze

In addition to antiviral effects, Type I interferons (IFN) have potent antiproliferative and immunomodulatory activities. Because of these properties IFNs have been evaluated as therapeutics for the treatment of a number of human diseases, including cancer. Currently, IFNs have been shown to be efficacious for the treatment of only a select number of cancers. The reason for this is unclear. Recent evidence has demonstrated that some cancer cell types seem to be defective in their ability to respond to IFN. It has been suggested that defects in IFN signaling is one mechanism by which cancer cells escape responsiveness to Type I IFNs and growth control in general. We report that transfection and enhanced expression of the Type I IFN receptor chain (IFNAR2c) in 3 different human cancer cell lines markedly increases the sensitivity of these cells to the antiproliferative effects of IFNs. In cancer cells transfected with IFNAR2c, dose response curves demonstrate a significant decrease in the concentrations of IFN required to achieve maximum cell death. Furthermore, in these transfected cells, we observe a significant increase in the number of cells undergoing apoptosis, as measured by DNA fragmentation and Caspase 3 activation. In addition, using an in vivo xenograft tumor model we show an increase in the effectiveness of systemically delivered Betaseron™ in decreasing tumor burden in animals in which solid tumors were generated from IFNAR2c transfected cells. These data show that specific regulation of IFN receptor expression can play a major role in determining the clinical outcome of IFN‐based cancer therapeutics by regulating the relative sensitivity of cancer cells to IFN‐dependent growth control.


Journal of Interferon and Cytokine Research | 2008

IFN-β1b Induces Transient and Variable Gene Expression in Relapsing-Remitting Multiple Sclerosis Patients Independent of Neutralizing Antibodies or Changes in IFN Receptor RNA Expression

Anthony T. Reder; Sharlene Velichko; Ken D. Yamaguchi; Kemal Hamamcioglu; Karin Ku; Johanna Beekman; T. Charis Wagner; H. Daniel Perez; Hugh Salamon; Ed Croze

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS). Interferon-beta (IFN-beta) therapy for MS is hypothesized to cause short-term and long-term changes in gene expression that shift the inflammation from Th1 to Th2. In vivo gene induction to define kinetics of response to IFN-beta therapy in a large cohort of MS patients is described. Differential gene expression in peripheral blood mononuclear cells (PBMCs) obtained from relapsing-remitting MS patients (RRMS) was assessed using high content microarrays. Rapid onset of gene expression appeared within 4 h of subcutaneous IFN-beta administration, returning to baseline levels at 42 h in clinically stable RRMS. IFN-beta therapy in vivo rapidly but transiently induced strong upregulation of genes mediating immune modulation, IFN signaling, and antiviral responses. RT-PCR showed significant patient-to-patient variation in the magnitude of expression of multiple genes, especially for IFN-beta-inducible genes, such as MxA, IRF7, and CCL8, a Th1 product. Variation among patients in IFN-beta-induced RNA transcription was not explained by neutralizing antibodies or IFN receptor expression. Surprisingly, genes regulated in vivo by IFN-beta therapy do not support a simple Th1 to Th2 shift. A complex interplay between both proinflammatory and anti-inflammatory immune regulatory genes is likely to act in concert in the treatment of RRMS.


The Journal of Urology | 1996

Human Renal Cancers Resistant to IFN's Antiproliferative Action Exhibit Sensitivity to IFN's Gene-Inducing and Antiviral Actions

Lawrence M. Pfeffer; Chiang Wang; Stefan N. Constantinescu; Ed Croze; Lawrence M. Blatt; Anthony P. Albino; David M. Nanus

PURPOSE Although treatment with interferon-alpha (IFN alpha) results in tumor regression in a subset (< 20%) of patients with renal cell carcinoma, the underlying mechanisms for the resistance of renal cancer (RC) cells to IFN alpha is unknown. MATERIALS AND METHODS We examined 5 RC lines resistant and 5 RC lines sensitive to the antiproliferative effects of IFN alpha for differences in: 1) the number of IFN binding sites, 2) the number of signal-transducing IFNAR-1 chains of the IFN alpha receptor, 3) IFN alpha receptor structure, 4) IFN-stimulated gene (ISG) expression and 5) IFN alpha sensitivity in antiviral assays. RESULTS No structural alterations in the IFN alpha receptor were detected in any RC line examined, although varying numbers of ligand binding sites and IFNAR-1 signal transducer chains were present. All 5 IFN-sensitive, and 4 of 5 IFN-resistant RC lines were sensitive to the antiviral and gene-inducing actions of IFN alpha. CONCLUSIONS The resistance of RC lines to IFNs antiproliferative action is not due to defects in ligand binding or in IFN-receptor structure. Our results indicate that the defective antiproliferative response in most RC cells is not due to their failure to induce the gene-inducing and antiviral effects of IFN alpha.


Journal of Biological Chemistry | 2002

STAT3 Activation by Type I Interferons Is Dependent on Specific Tyrosines Located in the Cytoplasmic Domain of Interferon Receptor Chain 2c ACTIVATION OF MULTIPLE STATS PROCEEDS THROUGH THE REDUNDANT USAGE OF TWO TYROSINE RESIDUES

Sharlene Velichko; T. Charis Wagner; James Turkson; Richard Jove; Ed Croze

Human type I interferons (IFNs) play an important role in the regulation of antiviral defense mechanisms, immunomodulatory activities, and growth control. Recent efforts have demonstrated the importance of IFNs in the activation ofsignal transducers and activators of transcription (STATs). The role of STAT1 and STAT2 in IFN-dependent JAK-STAT signaling is well established; however, the role of STAT3 and its activation by IFNs remains unclear. Understanding the IFN-dependent regulation of STAT3 is of increasing interest because recent studies have demonstrated that STAT3 may play a role in cancer. Studies have revealed that STAT3 is constitutively active in a number of cancer cell lines and that overexpression of an active form of STAT3 transforms normal fibroblasts. Therefore, STAT3 exhibits properties indicative of known oncogenes. In this report, we define the role of the type I IFN receptor in STAT3 activation and identify for the first time tyrosine residues present in the cytoplasmic domain of IFNAR2c that are critical for STAT3 activation. The regulation of STAT3 activation by IFNs was measured in a human lung fibrosarcoma cell line lacking IFNAR2c but stably expressing various IFNAR2c tyrosine mutants. We show here that in addition to IFN-dependent tyrosine phosphorylation of STAT3, activation using a STAT3-dependent electrophoretic mobility shift assay and a STAT3-specific reporter can also be demonstrated. Furthermore, we demonstrate that type I IFN-dependent activation of STAT3 proceeds through a novel mechanism that is dependent on two tyrosines, Tyr337 and Tyr512, present in IFNAR2c and contained within a conserved six-amino acid residue motif, GxGYxM. Surprisingly, both tyrosines were previously shown to be required for type I IFN-dependent STAT1 and STAT2 activation. Our results reveal that type I IFNs activate multiple STATs via the overlapping usage of two tyrosine residues located in the cytoplasmic domain of IFNAR2c.


International Journal of Cell Biology | 2010

Interferonβ-1b Induces the Expression of RGS1 a Negative Regulator of G-Protein Signaling

Tiffany Tran; Pedro Paz; Sharlene Velichko; Jill Cifrese; Praveen Belur; Ken D. Yamaguchi; Karin Ku; Parham Mirshahpanah; Anthony T. Reder; Ed Croze

We present evidence of a link between interferonβ-1b (IFN-β) and G-protein signaling by demonstrating that IFN-β can induce the expression of the negative regulator of G-protein signaling 1 (RGS1). RGS1 reduces G-protein activation and immune cell migration by interacting with heterotrimeric G-proteins and enhancing their intrinsic GTPase activity. In this study, IFN-β treatment resulted in the induction of RGS1 in peripheral blood mononuclear cells (PBMCs), monocytes, T cells, and B cells. Induction of RGS1 by IFN-β was concentration dependent and observed at both the RNA and protein level. Other members of the RGS family were not induced by IFN-β, and induction of RGS1 required the activation of the IFN receptor. In addition, RGS1 induction was observed in PBMCs obtained from IFN-β-treated multiple sclerosis patients suggesting a possible, as yet unexplored, involvement of G-protein regulation in disease treatment. The upregulation of RGS1 by IFN-β has not been previously reported.


Journal of Interferon and Cytokine Research | 2010

Differential gene expression and translational approaches to identify biomarkers of interferon beta activity in multiple sclerosis.

Ed Croze

More than 16 years ago human interferon-β-1b (IFN-β-1β) was shown to be effective in the treatment of the relapsing-remitting form of multiple sclerosis (MS). Over time, IFN-β has been demonstrated to be both a safe and effective treatment. However, the mechanism of action of IFN-β in MS remains unknown. To better understand the mechanism of action of IFN-β, considerable effort has been made in transcriptional profiling of peripheral blood mononuclear cells collected from MS patients. IFN-β is known to induce a large number of genes that play an important role in regulating responses to viral infection, immune modulation, and cell proliferation. Identifying differentially induced genes that are linked to the beneficial effects observed during treatment is under active investigation. IFN biomarkers in MS patients have been proposed but have not been clearly confirmed in independent studies or consistently correlated with clinical measures of disease progression. Organizing single genes or gene signatures grouped according to molecular mechanisms meaningful in MS may help to link IFN activity measurements to clinical outcomes. In this review, IFN activity measurements will be discussed with a specific emphasis on what is known about differential gene expression and treatment effects in MS.


Journal of Biological Chemistry | 1996

Direct Association of STAT3 with the IFNAR-1 Chain of the Human Type I Interferon Receptor

Chuanhe Yang; Wei Shi; Leela Basu; Aruna Murti; Stefan N. Constantinescu; Lawrence M. Blatt; Ed Croze; Jerald E. Mullersman; Lawrence M. Pfeffer

Collaboration


Dive into the Ed Croze's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aruna Murti

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Lawrence M. Pfeffer

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Leela Basu

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jerald E. Mullersman

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Stefan N. Constantinescu

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge