Network


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

Hotspot


Dive into the research topics where Nessa Hawkins is active.

Publication


Featured researches published by Nessa Hawkins.


Cell | 1998

Osteoprotegerin Ligand Is a Cytokine that Regulates Osteoclast Differentiation and Activation

David L. Lacey; E Timms; Hong-Lin Tan; Michael Kelley; Colin R. Dunstan; Teresa L. Burgess; Robin Elliott; Anne Colombero; Gary Elliott; Sheila Scully; Hailing Hsu; J Sullivan; Nessa Hawkins; E Davy; Casey Capparelli; A Eli; Y.-X Qian; Stephen Kaufman; Ildiko Sarosi; V Shalhoub; Giorgio Senaldi; J Guo; J Delaney; William J. Boyle

The ligand for osteoprotegerin has been identified, and it is a TNF-related cytokine that replaces the requirement for stromal cells, vitamin D3, and glucocorticoids in the coculture model of in vitro osteoclastogenesis. OPG ligand (OPGL) binds to a unique hematopoeitic progenitor cell that is committed to the osteoclast lineage and stimulates the rapid induction of genes that typify osteoclast development. OPGL directly activates isolated mature osteoclasts in vitro, and short-term administration into normal adult mice results in osteoclast activation associated with systemic hypercalcemia. These data suggest that OPGL is an osteoclast differentiation and activation factor. The effects of OPGL are blocked in vitro and in vivo by OPG, suggesting that OPGL and OPG are key extracellular regulators of osteoclast development.


Nature Immunology | 2000

APRIL and TALL-1 and receptors BCMA and TACI: system for regulating humoralimmunity

Gang Yu; Tom Boone; John M. Delaney; Nessa Hawkins; Michael Kelley; Meena Ramakrishnan; Susan McCabe; Wanrong Qiu; Masayo Kornuc; Xing-Zhong Xia; Jane Guo; Marina Stolina; William J. Boyle; Ildiko Sarosi; Hailing Hsu; Giorgio Senaldi; Lars Eyde Theill

We report that the tumor neurosis factor homolog APRIL (a proliferation-inducing ligand) stimulates in vitro proliferation of primary B and T cells and increases spleen weight due to accumulation of B cells in vivo. APRIL functions via binding to BCMA (B cell maturation antigen) and TACI (transmembrane activator and CAML-interactor) and competes with TALL-1 (also called BLyS or BAFF) for receptor binding. Soluble BCMA and TACI specifically prevent binding of APRIL and block APRIL-stimulated proliferation of primary B cells. BCMA-Fc also inhibits production of antibodies against keyhole limpet hemocyanin and Pneumovax in mice, indicating that APRIL and/or TALL-1 signaling via BCMA and/or TACI are required for generation of humoral immunity. Thus, APRIL–TALL-1 and BCMA-TACI form a two ligands–two receptors pathway involved in stimulation of B and T cell function.


Journal of Clinical Investigation | 2012

FGF23 neutralization improves chronic kidney disease–associated hyperparathyroidism yet increases mortality

Victoria Shalhoub; Edward Shatzen; Sabrina Ward; James O. Davis; Jennitte Stevens; Vivian Bi; Lisa Renshaw; Nessa Hawkins; Wei Wang; Ching Chen; Mei-Mei Tsai; Russell C. Cattley; Thomas J. Wronski; Xuechen Xia; Xiaodong Li; Charles Henley; Michael Eschenberg; William G. Richards

Chronic kidney disease-mineral and bone disorder (CKD-MBD) is associated with secondary hyperparathyroidism (HPT) and serum elevations in the phosphaturic hormone FGF23, which may be maladaptive and lead to increased morbidity and mortality. To determine the role of FGF23 in the pathogenesis of CKD-MBD and development of secondary HPT, we developed a monoclonal FGF23 antibody to evaluate the impact of chronic FGF23 neutralization on CKD-MBD, secondary HPT, and associated comorbidities in a rat model of CKD-MBD. CKD-MBD rats fed a high-phosphate diet were treated with low or high doses of FGF23-Ab or an isotype control antibody. Neutralization of FGF23 led to sustained reductions in secondary HPT, including decreased parathyroid hormone, increased vitamin D, increased serum calcium, and normalization of bone markers such as cancellous bone volume, trabecular number, osteoblast surface, osteoid surface, and bone-formation rate. In addition, we observed dose-dependent increases in serum phosphate and aortic calcification associated with increased risk of mortality in CKD-MBD rats treated with FGF23-Ab. Thus, mineral disturbances caused by neutralization of FGF23 limited the efficacy of FGF23-Ab and likely contributed to the increased mortality observed in this CKD-MBD rat model.


FEBS Letters | 2009

FGF21 N- and C-termini play different roles in receptor interaction and activation.

Junming Yie; Randy Ira Hecht; Jennifer Patel; Jennitte Stevens; Wei Wang; Nessa Hawkins; Shirley Steavenson; Steve Smith; Dwight Winters; Seth Fisher; Ling Cai; Ed Belouski; Ching Chen; Mark Leo Michaels; Yue-Sheng Li; Richard Lindberg; Minghan Wang; Murielle M. Véniant; Jing Xu

MINT‐6799907, MINT‐6799922: FGF21 (uniprotkb: Q9NSA1) binds (MI:0407) to β‐Klotho (uniprotkb: Q86Z14) by surface plasmon resonance (MI:0107)


Journal of Biological Chemistry | 2007

Co-receptor Requirements for Fibroblast Growth Factor-19 Signaling

Xinle Wu; Hongfei Ge; Jamila Gupte; Jennifer Weiszmann; Grant Shimamoto; Jennitte Stevens; Nessa Hawkins; Bryan Lemon; Wenyan Shen; Jing Xu; Murielle M. Véniant; Yue-Sheng Li; Richard Lindberg; Jin-Long Chen; Hui Tian; Yang Li

FGF19 is a unique member of the fibroblast growth factor (FGF) family of secreted proteins that regulates bile acid homeostasis and metabolic state in an endocrine fashion. Here we investigate the cell surface receptors required for signaling by FGF19. We show that βKlotho, a single-pass transmembrane protein highly expressed in liver and fat, induced ERK1/2 phosphorylation in response to FGF19 treatment and significantly increased the interactions between FGF19 and FGFR4. Interestingly, our results show that αKlotho, another Klotho family protein related to βKlotho, also induced ERK1/2 phosphorylation in response to FGF19 treatment and increased FGF19-FGFR4 interactions in vitro, similar to the effects of βKlotho. In addition, heparin further enhanced the effects of both αKlotho and βKlotho in FGF19 signaling and interaction experiments. These results suggest that a functional FGF19 receptor may consist of FGF receptor (FGFR) and heparan sulfate complexed with either αKlotho or βKlotho.


Journal of Biological Chemistry | 2008

C-terminal Tail of FGF19 Determines Its Specificity toward Klotho Co-receptors

Xinle Wu; Bryan Lemon; Xiaofan Li; Jamila Gupte; Jennifer Weiszmann; Jennitte Stevens; Nessa Hawkins; Wenyan Shen; Richard Lindberg; Jin-Long Chen; Hui Tian; Yang Li

FGF19 subfamily proteins (FGF19, FGF21, and FGF23) are unique members of fibroblast growth factors (FGFs) that regulate energy, bile acid, glucose, lipid, phosphate, and vitamin D homeostasis in an endocrine fashion. Their activities require the presence of α or βKlotho, two related single-pass transmembrane proteins, as co-receptors in relevant target tissues. We previously showed that FGF19 can bind to both α and βKlotho, whereas FGF21 and FGF23 can bind only to either βKlotho or αKlotho, respectively in vitro. To determine the mechanism regulating the binding and specificity among FGF19 subfamily members to Klotho family proteins, chimeric proteins between FGF19 subfamily members or chimeric proteins between Klotho family members were constructed to probe the interaction between those two families. Our results showed that a chimera of FGF19 with the FGF21 C-terminal tail interacts only with βKlotho and a chimera with the FGF23 C-terminal tail interacts only with αKlotho. FGF signaling assays also reflected the change of specificity we observed for the chimeras. These results identified the C-terminal tail of FGF19 as a region necessary for its recognition of Klotho family proteins. In addition, chimeras between α and βKlotho were also generated to probe the regions in Klotho proteins that are important for signaling by this FGF subfamily. Both FGF23 and FGF21 require intact α or βKlotho for signaling, respectively, whereas FGF19 can signal through a Klotho chimera consisting of the N terminus of αKlotho and the C terminus of βKlotho. Our results provide the first glimpse of the regions that regulate the binding specificity between this unique family of FGFs and their co-receptors.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Selective activation of FGFR4 by an FGF19 variant does not improve glucose metabolism in ob/ob mice

Xinle Wu; Hongfei Ge; Bryan Lemon; Jennifer Weiszmann; Jamila Gupte; Nessa Hawkins; Xiaofan Li; Jie Tang; Richard Lindberg; Yang Li

FGF19 is a hormone that regulates bile acid and glucose homeostasis. Progress has been made in identifying cofactors for receptor activation. However, several functions of FGF19 have not yet been fully defined, including the actions of FGF19 on target tissues, its FGF receptor specificity, and the contributions of other cofactors, such as heparin. Here, we explore the requirements for FGF19-FGFR/co-receptor interactions and signaling in detail. We show that βKlotho was essential for FGF19 interaction with FGFRs 1c, 2c, and 3c, but FGF19 was able to interact directly with FGFR4 in the absence of βKlotho in a heparin-dependent manner. Further, FGF19 activated FGFR4 signaling in the presence or absence of βKlotho, but activation of FGFRs 1c, 2c, or 3c was completely βKlotho dependent. We then generated an FGF19 molecule, FGF19dCTD, which has a deletion of the C-terminal region responsible for βKlotho interaction. We determined that βKlotho-dependent FGFR1c, 2c, and 3c interactions and activation were abolished, and βKlotho-independent FGFR4 activation was preserved; therefore, FGF19dCTD is an FGFR4-specific activator. This unique FGF19 molecule specifically activated FGFR4-dependent signaling in liver and suppressed CYP7A1 expression in vivo, but was unable to activate signaling in adipose where FGFR4 expression is very low. Interestingly, unlike FGF19, treatment of ob/ob mice with FGF19dCTD failed to improve glucose levels and insulin sensitivity. These results suggest that FGF19-regulated liver bile acid metabolism could be independent of its glucose-lowering effect, and direct FGFR activation in adipose tissue may play an important role in the regulation of glucose homeostasis.


Journal of Molecular Biology | 2010

Generation of novel long-acting globular adiponectin molecules.

Hongfei Ge; Yumei Xiong; Bryan Lemon; Ki Jeong Lee; Jay Tang; Ping Wang; Jennifer Weiszmann; Nessa Hawkins; John Laudemann; Xiaoshan Min; David Penny; Thomas Gerard Wolfe; Qiang Liu; Richard Zhang; Weh-Chen Yeh; Wenyan Shen; Richard Lindberg; Zhulun Wang; Jackie Zeqi Sheng; Yang Li

Adiponectin is an adipocyte-derived hormone that has been shown to play important roles in the regulation of glucose and energy homeostasis. It exists as homotrimers or complexes containing multiple homotrimer units in plasma. The recombinant adiponectin proteins have been difficult to produce, making it challenging for both research as well as potential therapeutic development. Here, we show a novel approach for the generation of globular adiponectin that involves linking three monomer sequences together in tandem to generate one continuous polypeptide, which we have termed single-chain globular adiponectin (sc-gAd). To improve the pharmacokinetic properties of sc-gAd further, we fused it to an Fc fragment. The combined effects of single-chain and Fc fusion improved the plasma half-life from less than 2 h to close to 2 weeks. Using adeno-associated virus as a delivery method, we demonstrate that Fc-sc-gAd improved both fasting glucose levels and the tolerance to a glucose challenge in ob/ob mice without changes in body weight. Therefore, our results demonstrated the feasibility of generating globular adiponectin trimers from a single polypeptide and a long-acting globular adiponectin that could serve as a starting point for adiponectin-based therapeutics. This novel approach could also be applied to other complement factor C1q family members; in particular, this opens the possibility to study the biological functions of precisely defined heterotrimers of various family members that had not been previously possible.


Journal of Molecular Biology | 2012

A unique FGF23 with the ability to activate FGFR signaling through both αKlotho and βKlotho.

Xinle Wu; Jennifer Weiszmann; Hongfei Ge; Helene Baribault; Jennitte Stevens; Nessa Hawkins; Steven Vonderfecht; Jonitha Gardner; Jamila Gupte; Jackie Zeqi Sheng; Minghan Wang; Yang Li

Three fibroblast growth factor (FGF) molecules, FGF19, FGF21, and FGF23, form a unique subfamily that functions as endocrine hormones. FGF19 and FGF21 can regulate glucose, lipid, and energy metabolism, while FGF23 regulates phosphate homeostasis. The FGF receptors and co-receptors for these three FGF molecules have been identified, and domains important for receptor interaction and specificity determination are beginning to be elucidated. However, a number of questions remain unanswered, such as the identification of fibroblast growth factor receptor responsible for glucose regulation. Here, we have generated a variant of FGF23: FGF23-21c, where the C-terminal domain of FGF23 was replaced with the corresponding regions from FGF21. FGF23-21c showed a number of interesting and unexpected properties in vitro. In contrast to wild-type FGF23, FGF23-21c gained the ability to activate FGFR1c and FGFR2c in the presence of βKlotho and was able to stimulate glucose uptake into adipocytes in vitro and lower glucose levels in ob/ob diabetic mice model to similar extent as FGF21 in vivo. These results suggest that βKlotho/FGFR1c or FGFR2c receptor complexes are sufficient for glucose regulation. Interestingly, without the FGF23 C-terminal domain, FGF23-21c was still able to activate fibroblast growth factor receptors in the presence of αKlotho. This suggests not only that sequences outside of the C-terminal region may also contribute to the interaction with co-receptors but also that FGF23-21c may be able to regulate both glucose and phosphate metabolisms. This raises an interesting concept of designing an FGF molecule that may be able to address multiple diseases simultaneously. Further understanding of FGF/receptor interactions may allow the development of exciting opportunities for novel therapeutic discovery.


FEBS Letters | 1993

Crystallization and preliminary X-ray analysis of leukemia inhibitory factor

Christian Betzel; Marcia Visanji; Zbigniew Dauter; Roger Fourme; Wolfgang A. Weber; Ulf Marnitz; Thomas C. Boone; Joseph Pope; James A. Miller; Nessa Hawkins; Babru Samal

Leukemia inhibitory factor (LIF) is a polyfunctional molecule with significant and diverse biological activities. LIF is a glycoprotein secreted by a number of different cell types in vitro. It is induced in fibroblasts, lymphocytes, monocytes and astrocytes by various inducers such as serum, TNF, interleukin‐IP and EGF. Due to extensive and variable glycosylation the molecular weight can range from 38 to 67 kDA. The biological functions of LIF are mediated through a receptor and a signal transducer, gp130, which is also used by factors like interleukm‐6 (IL‐6), cilliary neurotropic factor (CNTF), and oncostatin M (OSM). Here, we report the crystallization of the non‐glycosylated human‐like LIF expressed in E. coli. The present crystals diffract to 2.0 Å using synchrotron radiation. They belong to the monoclinic space group C2, and the cell dimensions are a = 61.5 Å, b = 45.3 Å , c =77.7 Å and β = 112.3°.

Collaboration


Dive into the Nessa Hawkins's collaboration.

Researchain Logo
Decentralizing Knowledge