Network


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

Hotspot


Dive into the research topics where Asne R. Bauskin is active.

Publication


Featured researches published by Asne R. Bauskin.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Large-scale delineation of secreted protein biomarkers overexpressed in cancer tissue and serum

John B. Welsh; Lisa M. Sapinoso; Suzanne Kern; David A. Brown; Tao Liu; Asne R. Bauskin; Robyn L. Ward; Nicholas J. Hawkins; David I. Quinn; Pamela J. Russell; Robert L. Sutherland; Samuel N. Breit; Christopher A. Moskaluk; Henry F. Frierson; Garret M. Hampton

Genetic alterations in tumor cells often lead to the emergence of growth-stimulatory autocrine and paracrine signals, involving overexpression of secreted peptide growth factors, cytokines, and hormones. Increased levels of these soluble proteins may be exploited for cancer diagnosis and management or as points of therapeutic intervention. Here, we combined the use of controlled vocabulary terms and sequence-based algorithms to predict genes encoding secreted proteins from among ≈12,500 sequences represented on oligonucleotide microarrays. Expression of these genes was queried in 150 carcinomas from 10 anatomic sites of origin and compared with 46 normal tissues derived from the corresponding sites of tumor origin and other body tissues and organs. Of 74 different genes identified as overexpressed in cancer tissues, several encode proteins with demonstrated clinical diagnostic application, such as α-fetoprotein in liver carcinoma, and kallikreins 6 and 10 in ovarian cancer, or therapeutic utility, such as gastrin-releasing peptide/bombesin in lung carcinomas. We show that several of the other candidate genes encode proteins with high levels of tumor-associated expression by immunohistochemistry on tissue microarrays and further demonstrate significantly elevated levels of another novel candidate protein, macrophage inhibitory cytokine 1, a distant member of the tranforming growth factor-β superfamily, in the serum of patients with metastatic prostate, breast, and colorectal carcinomas. Our results suggest that the combination of annotation/protein sequence analysis, transcript profiling, immunohistochemistry, and immunoassay is a powerful approach for delineating candidate biomarkers with potential clinical significance and may be broadly applicable to other human diseases.


Circulation Research | 2006

GDF15/MIC-1 Functions As a Protective and Antihypertrophic Factor Released From the Myocardium in Association With SMAD Protein Activation

Jian Xu; Thomas R. Kimball; John N. Lorenz; David A. Brown; Asne R. Bauskin; Raisa Klevitsky; Timothy E. Hewett; Samuel N. Breit; Jeffery D. Molkentin

Here we identified growth-differentiation factor 15 (GDF15) (also known as MIC-1), a secreted member of the transforming growth factor (TGF)-&bgr; superfamily, as a novel antihypertrophic regulatory factor in the heart. GDF15 is not expressed in the normal adult heart but is induced in response to conditions that promote hypertrophy and dilated cardiomyopathy. To elucidate the function of GDF15 in the heart, we generated transgenic mice with cardiac-specific overexpression. GDF15 transgenic mice were normal but were partially resistant to pressure overload-induced hypertrophy. Expression of GDF15 in neonatal cardiomyocyte cultures by adenoviral-mediated gene transfer antagonized agonist-induced hypertrophy in vitro. Transient expression of GDF15 outside the heart by intravenous adenoviral delivery, or by direct injection of recombinant GDF15 protein, attenuated ventricular dilation and heart failure in muscle lim protein gene–targeted mice through an endocrine effect. Conversely, examination of Gdf15 gene-targeted mice showed enhanced cardiac hypertrophic growth following pressure overload stimulation. Gdf15 gene-targeted mice also demonstrated a pronounced loss in ventricular performance following only 2 weeks of pressure overload stimulation, whereas wild-type controls maintained function. Mechanistically, GDF15 stimulation promoted activation of SMAD2/3 in cultured neonatal cardiomyocytes. Overexpression of SMAD2 attenuated cardiomyocyte hypertrophy similar to GDF15 treatment, whereas overexpression of the inhibitory SMAD proteins, SMAD6/7, reversed the antihypertrophic effects of GDF15. These results identify GDF15 as a novel autocrine/endocrine factor that antagonizes the hypertrophic response and loss of ventricular performance, possibly through a mechanism involving SMAD proteins.


Nature Medicine | 2007

Tumor-induced anorexia and weight loss are mediated by the TGF-beta superfamily cytokine MIC-1.

Heiko Johnen; Shu Lin; Tamara Kuffner; David A. Brown; Vicky Wang-Wei Tsai; Asne R. Bauskin; Liyun Wu; Greg J. Pankhurst; Lele Jiang; Simon Junankar; Mark Hunter; W. Douglas Fairlie; Nicola J. Lee; Ronaldo F. Enriquez; Paul A. Baldock; Eva Corey; Fred S. Apple; MaryAnn M. Murakami; En Ju Lin; Chuansong Wang; Matthew J. During; Amanda Sainsbury; Herbert Herzog; Samuel N. Breit

Anorexia and weight loss are part of the wasting syndrome of late-stage cancer, are a major cause of morbidity and mortality in cancer, and are thought to be cytokine mediated. Macrophage inhibitory cytokine-1 (MIC-1) is produced by many cancers. Examination of sera from individuals with advanced prostate cancer showed a direct relationship between MIC-1 abundance and cancer-associated weight loss. In mice with xenografted prostate tumors, elevated MIC-1 levels were also associated with marked weight, fat and lean tissue loss that was mediated by decreased food intake and was reversed by administration of antibody to MIC-1. Additionally, normal mice given systemic MIC-1 and transgenic mice overexpressing MIC-1 showed hypophagia and reduced body weight. MIC-1 mediates its effects by central mechanisms that implicate the hypothalamic transforming growth factor-β receptor II, extracellular signal–regulated kinases 1 and 2, signal transducer and activator of transcription-3, neuropeptide Y and pro-opiomelanocortin. Thus, MIC-1 is a newly defined central regulator of appetite and a potential target for the treatment of both cancer anorexia and weight loss, as well as of obesity.


The Lancet | 2002

Concentration in plasma of macrophage inhibitory cytokine-1 and risk of cardiovascular events in women: a nested case-control study.

David A. Brown; Samuel N. Breit; Julie E. Buring; W. Douglas Fairlie; Asne R. Bauskin; Tao Liu; Paul M. Ridker

BACKGROUND Macrophage inhibitory cytokine-1 (MIC-1) is part of the TGF-beta superfamily. Raised concentrations of MIC-1 in serum arise in several disease states, can be detected in normal individuals, and can partly be genetically determined. We aimed to investigate whether MIC-1 has a role in atherothrombosis. METHODS We did a prospective, nested, case-control study in 27628 initially healthy women. Of these women, we established baseline concentrations of MIC-1 in 257 who subsequently had myocardial infarction, stroke, or died from a cardiovascular event (cases) and in 257 matched for age and smoking status, who did not report cardiovascular disease during 4-year follow-up (controls). We also assessed polymorphisms in the MIC-1 gene (MIC-1 H and MIC-1 D) in all 514 women. FINDINGS MIC-1 concentrations were higher at baseline in women who subsequently had cardiovascular events than in those who did not (618 vs 538 pg/mL, p=0.0002). Concentrations above the 90th percentile (>856 pg/mL) were associated with a 2.7-fold increase in risk (95% CI 1.6-4.9, p=0.001). This effect was independent of traditional cardiovascular risk factors and at least additive to that of C-reactive protein. There was no significant association between MIC-1 polymorphism and vascular events. INTERPRETATION MIC-1 could be a novel target for cardiovascular disease prevention.


Cancer Research | 2006

Role of Macrophage Inhibitory Cytokine-1 in Tumorigenesis and Diagnosis of Cancer

Asne R. Bauskin; David A. Brown; Tamara Kuffner; Heiko Johnen; X. Wei Luo; Mark Hunter; Samuel N. Breit

Macrophage inhibitory cytokine-1 (MIC-1), a transforming growth factor-beta superfamily cytokine, is involved in tumor pathogenesis, and its measurement can be used as a clinical tool for the diagnosis and management of a wide range of cancers. Although generally considered to be part of the cells antitumorigenic repertoire, MIC-1 secretion, processing, and latent storage suggest a complex, dynamic variability in MIC-1 bioavailability in the tumor microenvironment, potentially modulating tumor progression and invasiveness.


Journal of Leukocyte Biology | 1999

MIC‐1 is a novel TGF‐β superfamily cytokine associated with macrophage activation

W.D Fairlie; Anthony G. Moore; Asne R. Bauskin; Patricia K. Russell; Hong-Ping Zhang; Samuel N. Breit

As part of a study to identify novel genes associated with macrophage activation, we have cloned a new member of the transforming growth factor β (TGF‐β) superfamily designated macrophage inhibitory cytokine 1 (MIC‐1). MIC‐1 is synthesized as a 62‐kDa intracellular protein, which, after cleavage by a furin like protease, is secreted as a 25‐kDa disulfide‐linked dimeric protein. Sequence analysis indicates that it does not cluster within any existing TGF‐β families, suggesting it may be the first member of a new grouping within the TGF‐β superfamily. Tissue Northern blots show that MIC‐1 transcripts are only found abundantly in placenta, although smaller amounts are seen in a limited number of other adult and fetal tissues. MIC‐1 is not expressed in resting macrophages but is induced by a number of different activation agents, including phorbol myristate acetate, interleukin 1, tumor necrosis factor α, and macrophage colony‐stimulating factor but not by lipopolysaccharide or interferon‐γ. We have hypothesized that it may be an autocrine inhibitor of macrophage activation but its major biological role is still uncertain. J. Leukoc. Biol. 65: 2–5; 1999.


Journal of Biological Chemistry | 2004

The Intracellular Chloride Ion Channel Protein CLIC1 Undergoes a Redox-controlled Structural Transition*

Dene R. Littler; Stephen J. Harrop; W. Douglas Fairlie; Louise J. Brown; Greg J. Pankhurst; Susan Pankhurst; Matthew Z. DeMaere; Terence J. Campbell; Asne R. Bauskin; Raffaella Tonini; Michele Mazzanti; Samuel N. Breit; Paul M. G. Curmi

Most proteins adopt a well defined three-dimensional structure; however, it is increasingly recognized that some proteins can exist with at least two stable conformations. Recently, a class of intracellular chloride ion channel proteins (CLICs) has been shown to exist in both soluble and integral membrane forms. The structure of the soluble form of CLIC1 is typical of a soluble glutathione S-transferase superfamily protein but contains a glutaredoxin-like active site. In this study we show that on oxidation CLIC1 undergoes a reversible transition from a monomeric to a non-covalent dimeric state due to the formation of an intramolecular disulfide bond (Cys-24–Cys-59). We have determined the crystal structure of this oxidized state and show that a major structural transition has occurred, exposing a large hydrophobic surface, which forms the dimer interface. The oxidized CLIC1 dimer maintains its ability to form chloride ion channels in artificial bilayers and vesicles, whereas a reducing environment prevents the formation of ion channels by CLIC1. Mutational studies show that both Cys-24 and Cys-59 are required for channel activity.


The EMBO Journal | 2000

The propeptide of macrophage inhibitory cytokine (MIC‐1), a TGF‐β superfamily member, acts as a quality control determinant for correctly folded MIC‐1

Asne R. Bauskin; Hong-Ping Zhang; W. Douglas Fairlie; Xiao Yan He; Patricia K. Russell; Anthony G. Moore; David A. Brown; Keith K. Stanley; Samuel N. Breit

Macrophage inhibitory cytokine (MIC‐1), a divergent member of the transforming growth factor‐β (TGF‐β) superfamily and activation associated cytokine, is secreted as a 28 kDa dimer. To understand its secretion, we examined its processing in MIC‐1‐transfected Chinese hamster ovary cells. Mature MIC‐1 dimer arises post‐endoplasmic reticulum (ER) by proteolytic cleavage of dimeric pro‐MIC‐1 precursor at a furin‐like site. Unlike previously characterized TGF‐β superfamily members, MIC‐1 dimers are also secreted in constructs lacking the propeptide. A clue to the function of the propeptide came from the observation that a range of proteasome inhibitors, including lactacystin and MG132, cause major increases in levels of undimerized pro‐MIC‐1 precursor. There was no effect of proteasome inhibitors on cells expressing mature MIC‐1 without the propeptide, suggesting that the propeptide can signal misfolding of MIC‐1, leading to proteasomal degradation. Deletion mutagenesis showed the N‐terminal 28 amino acids of the propeptide are necessary for proteasomal degradation. This is the first demonstration, to our knowledge, of a quality control function in a propeptide domain of a secretory protein and represents an additional mechanism to ensure correct folding of proteins leaving the ER.


Cancer Research | 2005

The Propeptide Mediates Formation of Stromal Stores of PROMIC-1: Role in Determining Prostate Cancer Outcome

Asne R. Bauskin; David A. Brown; Simon Junankar; K. Rasiah; Sarah A. Eggleton; Mark Hunter; Tao Liu; Dave Smith; Tamara Kuffner; Greg J. Pankhurst; Heiko Johnen; Pamela J. Russell; Wade Barret; John J. Grygiel; James G. Kench; Susan M. Henshall; Robert L. Sutherland; Samuel N. Breit

The extracellular matrix (ECM) is a reservoir of cellular binding proteins and growth factors that are critical for normal cell behavior, and aberrations in the ECM invariably accompany malignancies such as prostate cancer. Carcinomas commonly overexpress macrophage inhibitory cytokine 1 (MIC-1), a proapoptotic and antitumorigenic transforming growth factor-beta superfamily cytokine. Here we show that MIC-1 is often secreted in an unprocessed propeptide containing form. It is variably processed intracellularly, with unprocessed forms being secreted from several tumor lines, including prostate carcinoma lines, PC-3 and LNCaP. Once secreted, only unprocessed proMIC-1 binds ECM, demonstrating for the first time the occurrence of extracellular stores of MIC-1. The propeptide mediates this association via its COOH-terminal 89 amino acids. Xenograft models bearing tumors secreting various engineered forms of MIC-1 show that the propeptide regulates the balance between ECM stores and circulating serum levels of mature MIC-1 in vivo. The absence of propeptide results in approximately 20-fold increase in serum MIC-1 levels. The significance of stromal MIC-1 stores was evaluated in prostate cancer tissue cores, which show major variation in stromal levels of MIC-1. Stromal MIC-1 levels are linked to prostate cancer outcome following radical prostatectomy, with decreasing stromal levels providing an important independent predictor of disease relapse. In low-grade localized prostate cancer (Gleason sum score < or = 6), the level of MIC-1 stromal stores was the best predictor of future relapse when compared with all other clinicopathologic variables. The secretion and ECM association of unprocessed proMIC-1 is likely to play a central role in modulating local bioavailability of MIC-1 which can affect patient outcome in prostate cancer and other epithelial tumors.


Clinical Cancer Research | 2006

Measurement of Serum Levels of Macrophage Inhibitory Cytokine 1 Combined with Prostate-Specific Antigen Improves Prostate Cancer Diagnosis

David A. Brown; Carsten Stephan; Robyn L. Ward; Mathew Law; Mark Hunter; Asne R. Bauskin; Janaki Amin; Klaus Jung; Eleftherios P. Diamandis; Garret M. Hampton; Pamela J. Russell; Graham G. Giles; Samuel N. Breit

Purpose: Current serum testing for the detection of prostate cancer (PCa) lacks specificity. On diagnosis, the optimal therapeutic pathway is not clear and tools for adequate risk assessment of localized PCa progression are not available. This leads to a significant number of men having unnecessary diagnostic biopsies and surgery. A search for novel tumor markers identified macrophage inhibitory cytokine 1 (MIC-1) as a potentially useful marker. Follow-up studies revealed MIC-1 overexpression in local and metastatic PCa whereas peritumoral interstitial staining for MIC-1 identified lower-grade tumors destined for recurrence. Consequently, we sought to assess serum MIC-1 measurement as a diagnostic tool. Experimental Design: Using immunoassay determination of serum MIC-1 concentration in 1,000 men, 538 of whom had PCa, we defined the relationship of MIC-1 to disease variables. A diagnostic algorithm (MIC-PSA score) based on serum levels of MIC-1, total serum prostate-specific antigen, and percentage of free prostate-specific antigen was developed. Results: Serum MIC-1 was found to be an independent predictor of the presence of PCa and tumors with a Gleason sum ≥7. We validated the MIC-PSA score in a separate population and showed an improved specificity for diagnostic blood testing for PCa over percentage of free prostate-specific antigen, potentially reducing unnecessary biopsies by 27%. Conclusions: Serum MIC-1 is an independent marker of the presence of PCa and tumors with a Gleason sum of ≥7. The use of serum MIC-1 significantly increases diagnostic specificity and may be a future tool in the management of PCa.

Collaboration


Dive into the Asne R. Bauskin's collaboration.

Top Co-Authors

Avatar

Samuel N. Breit

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

David A. Brown

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

W. Douglas Fairlie

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Hong-Ping Zhang

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Patricia K. Russell

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Tao Liu

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Anthony G. Moore

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Pamela J. Russell

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Tamara Kuffner

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Terence J. Campbell

Victor Chang Cardiac Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge