Network


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

Hotspot


Dive into the research topics where Kathleen N. Tomkinson is active.

Publication


Featured researches published by Kathleen N. Tomkinson.


Biochemical and Biophysical Research Communications | 2003

Inhibition of myostatin in adult mice increases skeletal muscle mass and strength

Lisa-Anne Whittemore; Kening Song; Xiangping Li; Jane Aghajanian; Monique V. Davies; Stefan Girgenrath; Jennifer J. Hill; Mary Jalenak; Pamela Kelley; Andrea Knight; Rich Maylor; Denise O’Hara; Adele A. Pearson; Amira Quazi; Stephanie Ryerson; Xiang-Yang Tan; Kathleen N. Tomkinson; Geertruida M. Veldman; Angela Widom; Jill F. Wright; Steve Wudyka; Liz Zhao; Neil M. Wolfman

A human therapeutic that specifically modulates skeletal muscle growth would potentially provide a benefit for a variety of conditions including sarcopenia, cachexia, and muscular dystrophy. Myostatin, a member of the TGF-beta family of growth factors, is a known negative regulator of muscle mass, as mice lacking the myostatin gene have increased muscle mass. Thus, an inhibitor of myostatin may be useful therapeutically as an anabolic agent for muscle. However, since myostatin is expressed in both developing and adult muscles, it is not clear whether it regulates muscle mass during development or in adults. In order to test the hypothesis that myostatin regulates muscle mass in adults, we generated an inhibitory antibody to myostatin and administered it to adult mice. Here we show that mice treated pharmacologically with an antibody to myostatin have increased skeletal muscle mass and increased grip strength. These data show for the first time that myostatin acts postnatally as a negative regulator of skeletal muscle growth and suggest that myostatin inhibitors could provide a therapeutic benefit in diseases for which muscle mass is limiting.


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

Activation of latent myostatin by the BMP-1/tolloid family of metalloproteinases.

Neil M. Wolfman; Alexandra C. McPherron; William N. Pappano; Monique V. Davies; Kening Song; Kathleen N. Tomkinson; Jill F. Wright; Liz Zhao; Suzanne Sebald; Daniel S. Greenspan; Se-Jin Lee

Myostatin is a transforming growth factor β family member that acts as a negative regulator of skeletal muscle growth. Myostatin circulates in the blood of adult mice in a noncovalently held complex with other proteins, including its propeptide, which maintain the C-terminal dimer in a latent, inactive state. This latent form of myostatin can be activated in vitro by treatment with acid; however, the mechanisms by which latent myostatin is activated in vivo are unknown. Here, we show that members of the bone morphogenetic protein-1/tolloid (BMP-1/TLD) family of metalloproteinases can cleave the myostatin propeptide in this complex and can thereby activate latent myostatin. Furthermore, we show that a mutant form of the propeptide resistant to cleavage by BMP-1/TLD proteinases can cause significant increases in muscle mass when injected into adult mice. These findings raise the possibility that members of the BMP-1/TLD family may be involved in activating latent myostatin in vivo and that molecules capable of inhibiting these proteinases may be effective agents for increasing muscle mass for both human therapeutic and agricultural applications.


Journal of Biological Chemistry | 2007

Identification of an Interleukin 17F/17A Heterodimer in Activated Human CD4+ T Cells

Jill F. Wright; Yongjing Guo; Amira Quazi; Deborah Luxenberg; Frann Bennett; John F. Ross; Yongchang Qiu; Matthew J. Whitters; Kathleen N. Tomkinson; Kyri Dunussi-Joannopoulos; Beatriz M. Carreno; Mary Collins; Neil M. Wolfman

IL-17F and IL-17A are members of the IL-17 pro-inflammatory cytokine family. IL-17A has been implicated in the pathogenesis of autoimmune diseases. IL-17F is a disulfide-linked dimer that contains a cysteine-knot motif. We hypothesized that IL-17F and IL-17A could form a heterodimer due to their sequence homology and overlapping pattern of expression. We evaluated the structure of recombinant IL-17F and IL-17A proteins, as well as that of natural IL-17F and IL-17A derived from activated human CD4+ T cells, by enzyme-linked immunosorbent assay, immunoprecipitation followed by Western blotting, and mass spectrometry. We find that both IL-17F and IL-17A can form both homodimeric and heterodimeric proteins when expressed in a recombinant system, and that all forms of the recombinant proteins have in vitro functional activity. Furthermore, we find that in addition to the homodimers of IL-17F and IL-17A, activated human CD4+ T cells also produce the IL-17F/IL-17A heterodimer. These data suggest that the IL-17F/IL-17A heterodimer may contribute to the T cell-mediated immune responses.


Journal of Immunology | 2008

The Human IL-17F/IL-17A Heterodimeric Cytokine Signals through the IL-17RA/IL-17RC Receptor Complex

Jill F. Wright; Frann Bennett; Bilian Li; Jonathan Brooks; Deborah Luxenberg; Matthew J. Whitters; Kathleen N. Tomkinson; Lori Fitz; Neil M. Wolfman; Mary Collins; Kyri Dunussi-Joannopoulos; Moitreyee Chatterjee-Kishore; Beatriz M. Carreno

IL-17A and IL-17F, produced by the Th17 CD4+ T cell lineage, have been linked to a variety of inflammatory and autoimmune conditions. We recently reported that activated human CD4+ T cells produce not only IL-17A and IL-17F homodimers but also an IL-17F/IL-17A heterodimeric cytokine. All three cytokines can induce chemokine secretion from bronchial epithelial cells, albeit with different potencies. In this study, we used small interfering RNA and Abs to IL-17RA and IL-17RC to demonstrate that heterodimeric IL-17F/IL-17A cytokine activity is dependent on the IL-17RA/IL-17RC receptor complex. Interestingly, surface plasmon resonance studies indicate that the three cytokines bind to IL-17RC with comparable affinities, whereas they bind to IL-17RA with different affinities. Thus, we evaluated the effect of the soluble receptors on cytokine activity and we find that soluble receptors exhibit preferential cytokine blockade. IL-17A activity is inhibited by IL-17RA, IL-17F is inhibited by IL-17RC, and a combination of soluble IL-17RA/IL-17RC receptors is required for inhibition of the IL-17F/IL-17A activity. Altogether, these results indicate that human IL-17F/IL-17A cytokine can bind and signal through the same receptor complex as human IL-17F and IL-17A. However, the distinct affinities of the receptor components for IL-17A, IL-17F, and IL-17F/IL-17A heterodimer can be exploited to differentially affect the activity of these cytokines.


Journal of Applied Physiology | 2010

Administration of a soluble activin type IIB receptor promotes skeletal muscle growth independent of fiber type

Samuel M. Cadena; Kathleen N. Tomkinson; Travis E. Monnell; Matthew Spaits; Ravindra Kumar; Kathryn W. Underwood; R. Scott Pearsall; Jennifer Lachey

This is the first report that inhibition of negative regulators of skeletal muscle by a soluble form of activin type IIB receptor (ACE-031) increases muscle mass independent of fiber-type expression. This finding is distinct from the effects of selective pharmacological inhibition of myostatin (GDF-8), which predominantly targets type II fibers. In our study 8-wk-old C57BL/6 mice were treated with ACE-031 or vehicle control for 28 days. By the end of treatment, mean body weight of the ACE-031 group was 16% greater than that of the control group, and wet weights of soleus, plantaris, gastrocnemius, and extensor digitorum longus muscles increased by 33, 44, 46 and 26%, respectively (P<0.05). Soleus fiber-type distribution was unchanged with ACE-031 administration, and mean fiber cross-sectional area increased by 22 and 28% (P<0.05) in type I and II fibers, respectively. In the plantaris, a predominantly type II fiber muscle, mean fiber cross-sectional area increased by 57% with ACE-031 treatment. Analysis of myosin heavy chain (MHC) isoform transcripts by real-time PCR indicated no change in transcript levels in the soleus, but a decline in MHC I and IIa in the plantaris. In contrast, electrophoretic separation of total soleus and plantaris protein indicated that there was no change in the proportion of MHC isoforms in either muscle. Thus these data provide optimism that ACE-031 may be a viable therapeutic in the treatment of musculoskeletal diseases. Future studies should be undertaken to confirm that the observed effects are not age dependent or due to the relatively short study duration.


Journal of Biological Chemistry | 2010

Characterization of the ligand binding functionality of the extracellular domain of activin receptor type IIB

Dianne Sako; Asya Grinberg; June Liu; Monique V. Davies; Roselyne Castonguay; Silas Maniatis; Amy Andreucci; Eileen Pobre; Kathleen N. Tomkinson; Travis E. Monnell; Jeffrey Ucran; Erik Martinez-Hackert; R. Scott Pearsall; Kathryn W. Underwood; Jasbir Seehra; Ravindra Kumar

The single transmembrane domain serine/threonine kinase activin receptor type IIB (ActRIIB) has been proposed to bind key regulators of skeletal muscle mass development, including the ligands GDF-8 (myostatin) and GDF-11 (BMP-11). Here we provide a detailed kinetic characterization of ActRIIB binding to several low and high affinity ligands using a soluble activin receptor type IIB-Fc chimera (ActRIIB.Fc). We show that both GDF-8 and GDF-11 bind the extracellular domain of ActRIIB with affinities comparable with those of activin A, a known high affinity ActRIIB ligand, whereas BMP-2 and BMP-7 affinities for ActRIIB are at least 100-fold lower. Using site-directed mutagenesis, we demonstrate that ActRIIB binds GDF-11 and activin A in different ways such as, for example, substitutions in ActRIIB Leu79 effectively abolish ActRIIB binding to activin A yet not to GDF-11. Native ActRIIB has four isoforms that differ in the length of the C-terminal portion of their extracellular domains. We demonstrate that the C terminus of the ActRIIB extracellular domain is crucial for maintaining biological activity of the ActRIIB.Fc receptor chimera. In addition, we show that glycosylation of ActRIIB is not required for binding to activin A or GDF-11. Together, our findings reveal binding specificity and activity determinants of the ActRIIB receptor that combine to effect specificity in the activation of distinct signaling pathways.


Endocrinology | 2010

A Soluble Activin Receptor Type IIB Prevents the Effects of Androgen Deprivation on Body Composition and Bone Health

Alan Koncarevic; Milton Cornwall-Brady; Abigail Pullen; Monique V. Davies; Dianne Sako; June Liu; Ravindra Kumar; Kathleen N. Tomkinson; Theresa Baker; Ben Umiker; Travis E. Monnell; Asya Grinberg; Katia Liharska; Kathryn W. Underwood; Jeffrey Ucran; Elizabeth Howard; Joseph Barberio; Matthew Spaits; Scott Pearsall; Jasbir Seehra; Jennifer Lachey

Androgen deprivation, a consequence of hypogonadism, certain cancer treatments, or normal aging in men, leads to loss of muscle mass, increased adiposity, and osteoporosis. In the present study, using a soluble chimeric form of activin receptor type IIB (ActRIIB) we sought to offset the adverse effects of androgen deprivation on muscle, adipose tissue, and bone. Castrated (ORX) or sham-operated (SHAM) mice received either TBS [vehicle-treated (VEH)] or systemic administration of ActRIIB-mFc, a soluble fusion protein comprised of a form of the extracellular domain of ActRIIB fused to a murine IgG2aFc subunit. In vivo body composition imaging demonstrated that ActRIIB-mFc treatment results in increased lean tissue mass of 23% in SHAM mice [19.02 +/- 0.42 g (VEH) versus 23.43 +/- 0.35 g (ActRIIB-mFc), P < 0.00001] and 26% in ORX mice [15.59 +/- 0.26 g (VEH) versus 19.78 +/- 0.26 g (ActRIIB-mFc), P < 0.00001]. Treatment also caused a decrease in adiposity of 30% in SHAM mice [5.03 +/- 0.48 g (VEH) versus 3.53 +/- 0.19 g (ActRIIB-mFc), NS] and 36% in ORX mice [7.12 +/- 0.53 g (VEH) versus 4.57 +/- 0.28 g (ActRIIB-mFc), P < 0.001]. These changes were also accompanied by altered serum levels of leptin, adiponectin, and insulin, as well as by prevention of steatosis (fatty liver) in ActRIIB-mFc-treated ORX mice. Finally, ActRIIB-mFc prevented loss of bone mass in ORX mice as assessed by whole body dual x-ray absorptiometry and micro-computed tomography of proximal tibias. The data demonstrate that treatment with ActRIIB-mFc restored muscle mass, adiposity, and bone quality to normal levels in a mouse model of androgen deprivation, thereby alleviating multiple adverse consequences of such therapy.


Archive | 2002

Modified and stabilized GDF propeptides and uses thereof

Neil M. Wolfman; Soo Peang Khor; Kathleen N. Tomkinson


Archive | 2004

Antibodies against interleukin-22 and uses therefor

Jing Li; Xiang-Yang Tan; Kathleen N. Tomkinson; Debra D. Pittman; Geertruida M. Veldman; Lynette A. Fouser


Archive | 1998

Rapid generation of stable mammalian cell lines producing high levels of recombinant proteins

Kathleen N. Tomkinson; Monique V. Davies; John Reading McCOY

Collaboration


Dive into the Kathleen N. Tomkinson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ravindra Kumar

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jasbir Seehra

University of Southampton

View shared research outputs
Top Co-Authors

Avatar

Beatriz M. Carreno

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge