Network


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

Hotspot


Dive into the research topics where Christopher T. Veldkamp is active.

Publication


Featured researches published by Christopher T. Veldkamp.


Science Signaling | 2008

Structural Basis of CXCR4 Sulfotyrosine Recognition by the Chemokine SDF-1/CXCL12

Christopher T. Veldkamp; C Seibert; Francis C. Peterson; N.B De la Cruz; J.C Haugner; H Basnet; Thomas P. Sakmar; Brian F. Volkman

Solving the structure of the chemokine SDF-1 bound to an extracellular domain of its receptor CXCR4 has illustrated the basis of receptor sulfotyrosine recognition by chemokines and led to the discovery of an inhibitor of leukocyte chemotaxis. Insights into Chemokine-Receptor Interactions The chemokine stromal cell–derived factor 1 (SDF-1, also known as CXCL12) is the ligand of the chemokine receptor CXCR4, a G protein–coupled receptor (GPCR). This ligand–receptor pair plays important roles in development, leukocyte migration, and metastasis. High-affinity interactions between SDF-1 and CXCR4 depend on the sulfation of three critical tyrosine residues in the extracellular N-terminal region of CXCR4, a posttranslational modification common to other chemokine receptors. Transitions between the monomeric and dimeric forms of SDF-1 have interfered with previous attempts to solve the structure of the SDF-1:CXCR4 complex, which prompted Veldkamp et al. to use a constitutively dimeric form of SDF-1 (SDF12) in their study. In addition to solving the nuclear magnetic resonance structure of the SDF12:CXCR4-N-domain complex and thus determining the structural basis of the recognition of receptor sulfotyrosine residues by the chemokine, the authors also found another use for the dimeric chemokine. Although SDF12 stimulated CXCR4-mediated intracellular Ca2+ mobilization, it was unable to stimulate chemotaxis. Moreover, it inhibited chemotaxis to monomeric SDF-1, which suggests that it may be a useful therapeutic means of targeting CXCR4 activity. Stem cell homing and breast cancer metastasis are orchestrated by the chemokine stromal cell–derived factor 1 (SDF-1) and its receptor CXCR4. Here, we report the nuclear magnetic resonance structure of a constitutively dimeric SDF-1 in complex with a CXCR4 fragment that contains three sulfotyrosine residues important for a high-affinity ligand-receptor interaction. CXCR4 bridged the SDF-1 dimer interface so that sulfotyrosines sTyr7 and sTyr12 of CXCR4 occupied positively charged clefts on opposing chemokine subunits. Dimeric SDF-1 induced intracellular Ca2+ mobilization but had no chemotactic activity; instead, it prevented native SDF-1–induced chemotaxis, suggesting that it acted as a potent partial agonist. Our work elucidates the structural basis for sulfotyrosine recognition in the chemokine-receptor interaction and suggests a strategy for CXCR4-targeted drug development.


Protein Science | 2005

The monomer–dimer equilibrium of stromal cell-derived factor-1 (CXCL 12) is altered by pH, phosphate, sulfate, and heparin

Christopher T. Veldkamp; Francis C. Peterson; Adam J. Pelzek; Brian F. Volkman

Chemokines, like stromal cell‐derived factor‐1 (SDF1/CXCL12), are small secreted proteins that signal cells to migrate. Because SDF1 and its receptor CXCR4 play important roles in embryonic development, cancer metastasis, and HIV/AIDS, this chemokine signaling system is the subject of intense study. However, it is not known whether the monomeric or dimeric structure of SDF1 is responsible for signaling in vivo. Previous structural studies portrayed the SDF1 structure as either strictly monomeric in solution or dimeric when crystallized. Here, we report two‐dimensional NMR, pulsed‐field gradient diffusion and fluorescence polarization measurements at various SDF1 concentrations, solution conditions, and pH. These results demonstrate that SDF1 can form a dimeric structure in solution, but only at nonacidic pH when stabilizing counterions are present. Thus, while the previous NMR structural studies were performed under acidic conditions that strongly promote the monomeric state, crystallographic studies used nonacidic buffer conditions that included divalent anions shown here to promote dimerization. This pH‐sensitive aggregation behavior is explained by a dense cluster of positively charged residues at the SDF1 dimer interface that includes a histidine side chain at its center. A heparin disaccharide shifts the SDF1 monomer–dimer equilibrium in the same manner as other stabilizing anions, suggesting that glycosaminoglycan binding may be coupled to SDF1 dimerization in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Monomeric and dimeric CXCL12 inhibit metastasis through distinct CXCR4 interactions and signaling pathways

Luke J. Drury; Joshua J. Ziarek; Stéphanie Gravel; Christopher T. Veldkamp; Tomonori Takekoshi; Samuel T. Hwang; Nikolaus Heveker; Brian F. Volkman; Michael B. Dwinell

Chemokines and chemokine receptors are extensively and broadly involved in cancer metastasis. Previously, we demonstrated that epigenetic silencing of the chemokine CXCL12 sensitizes breast and colon cancer cells to endocrine signaling and metastasis to distant tissues. Yet, the precise mechanism whereby CXCL12 production by tumor cells regulates dissemination remains unclear. Here, we show that administration of CXCL12 extended survival of tumor-bearing mice by potently limiting metastasis of colorectal carcinoma or murine melanoma. Because secreted CXCL12 is a mixture of monomeric and dimeric species in equilibrium, oligomeric variants that either promote (monomer) or halt (dimer) chemotaxis were used to dissect the mechanisms interrupting carcinoma metastasis. Monomeric CXCL12 mobilized intracellular calcium, inhibited cAMP signaling, recruited β-arrestin-2, and stimulated filamentous-actin accumulation and cell migration. Dimeric CXCL12 activated G-protein-dependent calcium flux, adenylyl cyclase inhibition, and the rapid activation of ERK1/2, but only weakly, if at all, recruited arrestin, stimulated actin polymerization, or promoted chemotaxis. NMR analyses illustrated that CXCL12 monomers made specific contacts with CXCR4 that were lost following dimerization. Our results establish the potential for inhibiting CXCR4-mediated metastasis by administration of CXCL12. Chemokine-mediated migration and β-arrestin responses did not dictate the antitumor effect of CXCL12. We conclude that cellular migration is tightly regulated by selective CXCR4 signaling evoked by unique interactions with distinct ligand quaternary structures.


Protein Science | 2009

Monomeric structure of the cardioprotective chemokine SDF-1/CXCL12

Christopher T. Veldkamp; Joshua J. Ziarek; Jidong Su; Harihar Basnet; Richard C. Lennertz; Joshua J. Weiner; Francis C. Peterson; John E. Baker; Brian F. Volkman

The chemokine stromal cell‐derived factor‐1 (SDF‐1/CXCL12) directs leukocyte migration, stem cell homing, and cancer metastasis through activation of CXCR4, which is also a coreceptor for T‐tropic HIV‐1. Recently, SDF‐1 was shown to play a protective role after myocardial infarction, and the protein is a candidate for development of new anti‐ischemic compounds. SDF‐1 is monomeric at nanomolar concentrations but binding partners promote self‐association at higher concentrations to form a typical CXC chemokine homodimer. Two NMR structures have been reported for the SDF‐1 monomer, but only one matches the conformation observed in a series of dimeric crystal structures. In the other model, the C‐terminal helix is tilted at an angle incompatible with SDF‐1 dimerization. Using a rat heart explant model for ischemia/reperfusion injury, we found that dimeric SDF‐1 exerts no cardioprotective effect, suggesting that the active species is monomeric. To resolve the discrepancy between existing models, we solved the NMR structure of the SDF‐1 monomer in different solution conditions. Irrespective of pH and buffer composition, the C‐terminal helix remains tilted at an angle with no evidence for the perpendicular arrangement. Furthermore, we find that phospholipid bicelles promote dimerization that necessarily shifts the helix to the perpendicular orientation, yielding dipolar couplings that are incompatible with the NOE distance constraints. We conclude that interactions with the alignment medium biased the previous structure, masking flexibility in the helix position that may be essential for the distinct functional properties of the SDF‐1 monomer.


Science | 2016

Polysialylation controls dendritic cell trafficking by regulating chemokine recognition.

Eva Kiermaier; Christine Moussion; Christopher T. Veldkamp; Rita Gerardy-Schahn; Ingrid de Vries; Larry G. Williams; Gary R. Chaffee; Andrew J. Phillips; Friedrich Freiberger; Richard Imre; Richard J. Payne; Asolina Braun; Reinhold Förster; Karl Mechtler; Martina Mühlenhoff; Brian F. Volkman; Michael Sixt

A chemokines sugary release As immune cells survey the body for pathogens, they circulate through the blood and migrate through the lymphatic system. The latter route allows for tissues and lymph nodes—the central hubs of the immune system—to communicate. Kiermaier et al. reveal the importance of the monosaccharide sialic acid in keeping immune cells in motion. Multiple sialic acids decorate the surface CCR7 on immune cells. CCR7 recognizes proteins called chemokines, which direct where cells move in the body. Sialic acids on CCR7 release one such chemokine present on lymph node endothelial cells from an inhibited state, allowing immune cells to enter lymph nodes. Science, this issue p. 186 For dendritic cells to find their way to lymph nodes, the chemokine receptor CCR7 needs to have polysialic acid on it. The addition of polysialic acid to N- and/or O-linked glycans, referred to as polysialylation, is a rare posttranslational modification that is mainly known to control the developmental plasticity of the nervous system. Here we show that CCR7, the central chemokine receptor controlling immune cell trafficking to secondary lymphatic organs, carries polysialic acid. This modification is essential for the recognition of the CCR7 ligand CCL21. As a consequence, dendritic cell trafficking is abrogated in polysialyltransferase-deficient mice, manifesting as disturbed lymph node homeostasis and unresponsiveness to inflammatory stimuli. Structure-function analysis of chemokine-receptor interactions reveals that CCL21 adopts an autoinhibited conformation, which is released upon interaction with polysialic acid. Thus, we describe a glycosylation-mediated immune cell trafficking disorder and its mechanistic basis.


Biochemistry | 2008

Sequential Tyrosine Sulfation of CXCR4 by Tyrosylprotein Sulfotransferases

Christoph Seibert; Christopher T. Veldkamp; Francis C. Peterson; Brian T. Chait; Brian F. Volkman; Thomas P. Sakmar

CXC-chemokine receptor 4 (CXCR4) is a G protein-coupled receptor for stromal cell-derived factor-1 (SDF-1/CXCL12). SDF-1-induced CXCR4 signaling is indispensable for embryonic development and crucial for immune cell homing and has been implicated in metastasis of numerous types of cancer. CXCR4 also serves as the major coreceptor for cellular entry of T-cell line-tropic (X4) HIV-1 strains. Tyrosine residues in the N-terminal tail of CXCR4, which are post-translationally sulfated, are implicated in the high-affinity binding of SDF-1 to CXCR4. However, the specific roles of three potential tyrosine sulfation sites are not well understood. We investigated the pattern and sequence of CXCR4 sulfation by using recombinant human tyrosylprotein sulfotransferases TPST-1 and TPST-2 to modify a peptide that corresponds to amino acids 1-38 of the receptor (CXCR4 1-38). We analyzed the reaction products with a combination of reversed-phase HPLC, proteolytic cleavage, and mass spectrometry. We found that CXCR4 1-38 is sulfated efficiently by both TPST enzymes, leading to a final product with three sulfotyrosine residues. Sulfates were added stepwise to the peptide, producing specific intermediates with one or two sulfotyrosines. The pattern of sulfation in these intermediates indicates that with both enzymes Tyr-21 is sulfated first, followed by Tyr-12 or Tyr-7. Using heteronuclear NMR spectroscopy, we demonstrated that the SDF-1 binding affinity of CXCR4 1-38 increases with the number of sulfotyrosines present, which suggests a potential physiological role for sulfation of all three sites in the N-terminus of CXCR4. These results provide a structural basis for understanding the role of post-translational tyrosine sulfation in SDF-1-induced CXCR4 signaling.


Journal of the American Chemical Society | 2010

Targeting SDF-1/CXCL12 with a ligand that prevents activation of CXCR4 through structure-based drug design.

Christopher T. Veldkamp; Joshua J. Ziarek; Francis C. Peterson; Yu Chen; Brian F. Volkman

CXCL12 is an attractive target for clinical therapy because of its involvement in autoimmune diseases, cancer growth, metastasis, and neovascularization. Tyrosine sulfation at three positions in the CXCR4 N-terminus is crucial for specific, high-affinity CXCL12 binding. An NMR structure of the complex between the CXCL12 dimer and a sulfotyrosine-containing CXCR4 fragment enabled high-throughput in silico screening for inhibitors of the chemokine-receptor interface. A total of 1.4 million compounds from the ZINC database were docked into a cleft on the CXCL12 surface normally occupied by sulfotyrosine 21 (sY21), and five were selected for experimental screening. NMR titrations with CXCL12 revealed that four of the compounds occupy the sY21 site, one of which binds with a K(d) of 64 microM. This compound selectively inhibits SDF1-induced CXCR4 signaling in THP1 cells. Our results suggest that sulfotyrosine recognition sites can be targeted for the development of novel chemokine inhibitors.


Journal of Biological Chemistry | 2013

Heparin Oligosaccharides Inhibit Chemokine (CXC Motif) Ligand 12 (CXCL12) Cardioprotection by Binding Orthogonal to the Dimerization Interface, Promoting Oligomerization, and Competing with the Chemokine (CXC Motif) Receptor 4 (CXCR4) N Terminus

Joshua J. Ziarek; Christopher T. Veldkamp; Fuming Zhang; Nathan J. Murray; Gabriella A. Kartz; Xinle Liang; Jidong Su; John E. Baker; Robert J. Linhardt; Brian F. Volkman

Background: GAG/CXCL12 interactions are critical for chemokine function but co-administration may abrogate their individual cardioprotective effects in a clinical setting. Results: Biophysical studies distinguish CXCL12 residues involved in dimerization from those likely to contact heparin directly. Conclusion: CXCL12 dimerization is required for high affinity heparin binding and protects N-terminal degradation. Significance: Knowledge of the GAG-binding site will enable future development of heparin-insensitive CXCL12 therapeutics. The ability to interact with cell surface glycosaminoglycans (GAGs) is essential to the cell migration properties of chemokines, but association with soluble GAGs induces the oligomerization of most chemokines including CXCL12. Monomeric CXCL12, but not dimeric CXCL12, is cardioprotective in a number of experimental models of cardiac ischemia. We found that co-administration of heparin, a common treatment for myocardial infarction, abrogated the protective effect of CXCL12 in an ex vivo rat heart model for myocardial infarction. The interaction between CXCL12 and heparin oligosaccharides has previously been analyzed through mutagenesis, in vitro binding assays, and molecular modeling. However, complications from heparin-induced CXCL12 oligomerization and studies using very short oligosaccharides have led to inconsistent conclusions as to the residues involved, the orientation of the binding site, and whether it overlaps with the CXCR4 N-terminal site. We used a constitutively dimeric variant to simplify the NMR analysis of CXCL12-binding heparin oligosaccharides of varying length. Biophysical and mutagenic analyses reveal a CXCL12/heparin interaction surface that lies perpendicular to the dimer interface, does not involve the chemokine N terminus, and partially overlaps with the CXCR4-binding site. We further demonstrate that heparin-mediated enzymatic protection results from the promotion of dimerization rather than direct heparin binding to the CXCL12 N terminus. These results clarify the structural basis for GAG recognition by CXCL12 and lend insight into the development of CXCL12-based therapeutics.


Biochemistry | 2012

Solution structure of CCL21 and identification of a putative CCR7 binding site.

Melissa Love; Jamie L. Sandberg; Joshua J. Ziarek; Kyle P. Gerarden; Renee R. Rode; Davin R. Jensen; Darrell R. McCaslin; Francis C. Peterson; Christopher T. Veldkamp

CCL21 is a human chemokine that recruits normal immune cells and metastasizing tumor cells to lymph nodes through activation of the G protein-coupled receptor CCR7. The CCL21 structure solved by NMR contains a conserved chemokine domain followed by an extended, unstructured C-terminus that is not typical of most other chemokines. A sedimentation equilibrium study showed CCL21 to be monomeric. Chemical shift mapping indicates that the CCR7 N-terminus binds to the N-loop and third β-strand of CCL21s chemokine domain. Details of CCL21-receptor recognition may enable structure-based drug discovery of novel antimetastatic agents.


Science Signaling | 2017

Structural basis for chemokine recognition by a G protein–coupled receptor and implications for receptor activation

Joshua J. Ziarek; Andrew B. Kleist; Nir London; Barak Raveh; Nicolas Montpas; Julien Bonneterre; Geneviève St-Onge; Crystal J. DiCosmo-Ponticello; Chad A. Koplinski; Ishan Roy; Bryan Stephens; Sylvia Thelen; Christopher T. Veldkamp; Frederick D. Coffman; Marion C. Cohen; Michael B. Dwinell; Marcus Thelen; Francis C. Peterson; Nikolaus Heveker; Brian F. Volkman

Structural analysis of the interactions between a receptor and monomeric or dimeric forms of its ligand may aid in drug design. How receptors view monomers versus dimers Chemokines are proteins that stimulate cell migration in processes such as development, immune responses, and metastasis. Monomeric, dimeric, and oligomeric forms of chemokines can engage their cognate G protein–coupled receptors. Both the G protein–dependent and β-arrestin–dependent signaling pathways downstream of chemokine receptors must be activated to induce cell migration. Previous studies showed that a locked dimeric form of CXCL12 (LD CXCL12) fails to activate β-arrestin–dependent signaling after binding to its receptor CXCR4. Ziarek et al. solved the NMR structure of CXCR4 bound to a locked monomeric form of CXCL12 (LM CXCL12). LM CXCL12 physically interacted with the receptor differently than did the dimeric chemokine, and it stimulated both CXCR4-dependent signaling pathways to induce migration. Analysis of a hybrid NMR- and x-ray–based structure provided insights into the conformational changes required for chemokine receptor signaling, which may aid in designing drugs to target the chemokine family. Chemokines orchestrate cell migration for development, immune surveillance, and disease by binding to cell surface heterotrimeric guanine nucleotide–binding protein (G protein)–coupled receptors (GPCRs). The array of interactions between the nearly 50 chemokines and their 20 GPCR targets generates an extensive signaling network to which promiscuity and biased agonism add further complexity. The receptor CXCR4 recognizes both monomeric and dimeric forms of the chemokine CXCL12, which is a distinct example of ligand bias in the chemokine family. We demonstrated that a constitutively monomeric CXCL12 variant reproduced the G protein–dependent and β-arrestin–dependent responses that are associated with normal CXCR4 signaling and lead to cell migration. In addition, monomeric CXCL12 made specific contacts with CXCR4 that are not present in the structure of the receptor in complex with a dimeric form of CXCL12, a biased agonist that stimulates only G protein–dependent signaling. We produced an experimentally validated model of an agonist-bound chemokine receptor that merged a nuclear magnetic resonance–based structure of monomeric CXCL12 bound to the amino terminus of CXCR4 with a crystal structure of the transmembrane domains of CXCR4. The large CXCL12:CXCR4 protein-protein interface revealed by this structure identified previously uncharacterized functional interactions that fall outside of the classical “two-site model” for chemokine-receptor recognition. Our model suggests a mechanistic hypothesis for how interactions on the extracellular face of the receptor may stimulate the conformational changes required for chemokine receptor–mediated signal transduction.

Collaboration


Dive into the Christopher T. Veldkamp's collaboration.

Top Co-Authors

Avatar

Brian F. Volkman

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Francis C. Peterson

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Joshua J. Ziarek

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Michael B. Dwinell

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Phillips

University of Wisconsin–Whitewater

View shared research outputs
Top Co-Authors

Avatar

Chad A. Koplinski

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Christoph Seibert

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Emily R. Lackner

University of Wisconsin–Whitewater

View shared research outputs
Top Co-Authors

Avatar

Gary R. Chaffee

University of Wisconsin–Whitewater

View shared research outputs
Top Co-Authors

Avatar

Scott J. Schoeller

University of Wisconsin–Whitewater

View shared research outputs
Researchain Logo
Decentralizing Knowledge