Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Obes Surg ; 33(6): 1944-1948, 2023 06.
Article in English | MEDLINE | ID: mdl-37058265

ABSTRACT

BACKGROUND: Obesity is associated with derangement of cardiac metabolism and the development of subclinical cardiovascular disease. This prospective study examined the impact of bariatric surgery on cardiac function and metabolism. METHODS: Subjects with obesity underwent cardiac magnetic resonance imaging (CMR) at Massachusetts General Hospital before and after bariatric surgery between 2019 and 2021. The imaging protocol included Cine for global cardiac function assessment and creatine chemical exchange saturation transfer (CEST) CMR for myocardial creatine mapping. RESULTS: Thirteen subjects were enrolled, and 6 subjects [mean BMI 40.5 ± 2.6] had completed the second CMR (i.e. post-surgery), with a median follow-up of 10 months. The median age was 46.5 years, 67% were female, and 16.67% had diabetes. Bariatric surgery led to significant weight loss, with achieved mean BMI of 31.0 ± 2.0. Additionally, bariatric surgery resulted in significant reduction in left ventricular (LV) mass, LV mass index, and epicardial adipose tissue (EAT) volume. This was accompanied by slight improvement in LV ejection fraction compared to baseline. Following bariatric surgery, there was a significant increase in creatine CEST contrast. Subjects with obesity had significantly lower CEST contrast compared to subjects with normal BMI (n = 10), but this contrast was normalized after the surgery, and statistically similar to non-obese cohort, indicating an improvement in myocardial energetics. CONCLUSIONS: CEST-CMR has the ability to identify and characterize myocardial metabolism in vivo non-invasively. These results demonstrate that in addition to reducing BMI, bariatric surgery may favorably affect cardiac function and metabolism.


Subject(s)
Bariatric Surgery , Obesity, Morbid , Humans , Female , Middle Aged , Male , Creatine/metabolism , Prospective Studies , Obesity, Morbid/surgery , Obesity/complications , Magnetic Resonance Imaging/methods , Ventricular Function, Left
2.
Magn Reson Med ; 90(1): 222-230, 2023 07.
Article in English | MEDLINE | ID: mdl-36864561

ABSTRACT

PURPOSE: To investigate the feasibility of combining simultaneous multislice (SMS) and region-optimized virtual coils (ROVir) for single breath-hold CINE imaging. METHOD: ROVir is a recent virtual coil approach that allows reduced-field of view (FOV) imaging by localizing the signal from a region-of-interest (ROI) and/or suppressing the signal from unwanted spatial regions. In this work, ROVir is used for reduced-FOV SMS bSSFP CINE imaging, which enables whole heart CINE with a single breath-hold acquisition. RESULTS: Reduced-FOV CINE with either SMS-only or ROVir-only resulted in significant aliasing, with severely reduced image quality when compared to the full FOV reference CINE, while the visual appearance of aliasing was substantially reduced with the proposed SMS+ROVir. The end diastolic volume, end systolic volume, and ejection fraction obtained using the proposed approach were similar to the clinical reference (correlations of 0.92, 0.94, and 0.88, respectively with p < 0 . 05 $$ p<0.05 $$ in each case, and biases of 0.1, 1.6 mL, and - 0 . 6 % $$ -0.6\% $$ , respectively). No statistically significant differences for these parameters were found with a Wilcoxon rank test (p = 0.96, 0.20, and 0.40, respectively). CONCLUSION: We demonstrated that reduced-FOV CINE imaging with SMS+ROVir enables single breath-hold whole-heart imaging without compromising visual image quality or quantitative cardiac function parameters.


Subject(s)
Breath Holding , Magnetic Resonance Imaging, Cine , Magnetic Resonance Imaging, Cine/methods , Reproducibility of Results , Image Interpretation, Computer-Assisted/methods
3.
J Mol Biol ; 427(10): 1934-48, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25765764

ABSTRACT

We have expressed and purified three soluble fragments of the human LRIG1-ECD (extracellular domain): the LRIG1-LRR (leucine-rich repeat) domain, the LRIG1-3Ig (immunoglobulin-like) domain, and the LRIG1-LRR-1Ig fragment using baculovirus vectors in insect cells. The two LRIG1 domains crystallised so that we have been able to determine the three-dimensional structures at 2.3Å resolution. We developed a three-dimensional structure for the LRIG1-ECD using homology modelling based on the LINGO-1 structure. The LRIG1-LRR domain and the LRIG1-LRR-1Ig fragment are monomers in solution, whereas the LRIG1-3Ig domain appears to be dimeric. We could not detect any binding of the LRIG1 domains or the LRIG1-LRR-1Ig fragment to the EGF receptor (EGFR), either in solution using biosensor analysis or when the EGFR was expressed on the cell surface. The FLAG-tagged LRIG1-LRR-1Ig fragment binds weakly to colon cancer cells regardless of the presence of EGFRs. Similarly, neither the soluble LRIG1-LRR nor the LRIG1-3Ig domains nor the full-length LRIG1 co-expressed in HEK293 cells inhibited ligand-stimulated activation of cell-surface EGFR.


Subject(s)
ErbB Receptors/chemistry , ErbB Receptors/metabolism , Extracellular Matrix/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Biosensing Techniques , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Crystallography, X-Ray , HEK293 Cells , Humans , Leucine-Rich Repeat Proteins , Ligands , Microscopy, Fluorescence , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism , Structure-Activity Relationship , Surface Plasmon Resonance , Tumor Cells, Cultured
4.
Biochem J ; 451(2): 165-75, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23384096

ABSTRACT

Gene deletion studies in mice have revealed critical roles for IL (interleukin)-4 and -13 in asthma development, with the latter controlling lung airways resistance and mucus secretion. We have now developed human neutralizing monoclonal antibodies against human IL-13Rα1 (IL-13 receptor α1) subunit that prevent activation of the receptor complex by both IL-4 and IL-13. We describe the crystal structures of the Fab fragment of antibody 10G5H6 alone and in complex with D3 (ectodomain 3) of IL-13Rα1. Although the structure showed significant domain swapping within a D3 dimer, we showed that Arg(230), Phe(233), Tyr(250), Gln(252) and Leu(293) in each D3 monomer and Ser(32), Asn(102) and Trp(103) in 10G5H6 Fab are the key interacting residues at the interface of the 10G5H6 Fab-D3 complex. One of the most striking contacts is the insertion of the ligand-contacting residue Leu(293) of D3 into a deep pocket on the surface of 10G5H6 Fab, and this appears to be a central determinant of the high binding affinity and neutralizing activity of the antibody.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Epitopes , Interleukin-13 Receptor alpha1 Subunit/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Binding Sites/immunology , Crystallography, X-Ray , Dimerization , Humans , Immunoglobulin Fab Fragments/chemistry , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-13 Receptor alpha1 Subunit/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Leucine/metabolism , Mice , Mice, Transgenic , Protein Structure, Tertiary
5.
Growth Factors ; 30(6): 394-409, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23163584

ABSTRACT

The activation of the epidermal growth factor receptor (EGFR) kinase requires ligand binding to the extracellular domain (ECD). Previous reports demonstrate that the EGFR-ECD can be crystallized in two conformations - a tethered monomer or, in the presence of ligand, an untethered back-to-back dimer. We use Biosensor analysis to demonstrate that even in the monomeric state different C-terminal extensions of both truncated (EGFR(1-501))-ECD and full-length EGFR(1-621)-ECD can change the conformation of the ligand-binding site. The binding of a monoclonal antibody mAb806, which recognizes the dimer interface, to the truncated EGFR(1-501)-Fc fusion protein is reduced in the presence of ligand, consistent with a change in conformation. On the cell surface, the presence of erythroblastosis B2 (erbB2) increases the binding of mAb806 to the EGFR. The conformation of the erbB2: EGFR heterodimer interface changes when the cells are treated with epidermal growth factor (EGF). We propose that ligand induces kinase-inactive, pre-formed EGFR dimers and heterodimers to change conformation leading to kinase-active tetramers, where kinase activation occurs via an asymmetric interaction between EGFR dimers.


Subject(s)
ErbB Receptors/chemistry , Ligands , Animals , Antibodies, Monoclonal/chemistry , Biosensing Techniques , Cell Line , Dimerization , Epitopes/chemistry , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Kinetics , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Structure, Tertiary
6.
J Biol Chem ; 285(28): 21214-8, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20489211

ABSTRACT

gp130 is the shared signal-transducing receptor subunit for the large and important family of interleukin 6-like cytokines. Previous x-ray structures of ligand-receptor complexes of this family lack the three membrane-proximal domains that are essential for signal transduction. Here we report the crystal structure of the entire extracellular portion of human gp130 (domains 1-6, D1-D6) at 3.6 A resolution, in an unliganded form, as well as a higher resolution structure of the membrane-proximal fibronectin type III domains (D4-D6) at 1.9 A. This represents the first atomic resolution structure of the complete ectodomain of any "tall" cytokine receptor. These structures show that other than a reorientation of the D1 domain, there is little structural change in gp130 upon ligand binding. They also reveal that the interface between the D4 and D5 domains forms an acute bend in the gp130 structure. Key residues at this interface are highly conserved across the entire tall receptor family, suggesting that this acute bend may be a common feature of these receptors. Importantly, this geometry positions the C termini of the membrane-proximal fibronectin type III domains of the tall cytokine receptors in close proximity within the transmembrane complex, favorable for receptor-associated Janus kinases to trans-phosphorylate and activate each other.


Subject(s)
Interleukin-6/chemistry , Neural Cell Adhesion Molecules/chemistry , Contactins , Crystallography, X-Ray/methods , Cytokines/metabolism , Dimerization , Fibronectins/chemistry , Humans , Ligands , Molecular Conformation , Phosphorylation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Signal Transduction , Structure-Activity Relationship
7.
Proc Natl Acad Sci U S A ; 106(13): 5082-7, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19289842

ABSTRACT

Epidermal Growth Factor Receptor (EGFR) is involved in stimulating the growth of many human tumors, but the success of therapeutic agents has been limited in part by interference from the EGFR on normal tissues. Previously, we reported an antibody (mab806) against a truncated form of EGFR found commonly in gliomas. Remarkably, it also recognizes full-length EGFR on tumor cells but not on normal cells. However, the mechanism for this activity was unclear. Crystallographic structures for Fab:EGFR(287-302) complexes of mAb806 (and a second, related antibody, mAb175) show that this peptide epitope adopts conformations similar to those found in the wtEGFR. However, in both conformations observed for wtEGFR, tethered and untethered, antibody binding would be prohibited by significant steric clashes with the CR1 domain. Thus, these antibodies must recognize a cryptic epitope in EGFR. Structurally, it appeared that breaking the disulfide bond preceding the epitope might allow the CR1 domain to open up sufficiently for antibody binding. The EGFR(C271A/C283A) mutant not only binds mAb806, but binds with 1:1 stoichiometry, which is significantly greater than wtEGFR binding. Although mAb806 and mAb175 decrease tumor growth in xenografts displaying mutant, overexpressed, or autocrine stimulated EGFR, neither antibody inhibits the in vitro growth of cells expressing wtEGFR. In contrast, mAb806 completely inhibits the ligand-associated stimulation of cells expressing EGFR(C271A/C283A). Clearly, the binding of mAb806 and mAb175 to the wtEGFR requires the epitope to be exposed either during receptor activation, mutation, or overexpression. This mechanism suggests the possibility of generating antibodies to target other wild-type receptors on tumor cells.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , ErbB Receptors/immunology , Neoplasm Proteins/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antigen-Antibody Complex/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Crystallography, X-Ray , Epitopes , Humans , Mice , Mice, Nude , Protein Conformation , Protein Denaturation/immunology , Xenograft Model Antitumor Assays
8.
J Mol Biol ; 386(3): 662-74, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19154741

ABSTRACT

The four mammalian SPRY (a sequence repeat in dual-specificity kinase splA and ryanodine receptors) domain-containing suppressor of cytokine signalling (SOCS) box proteins (SSB-1 to -4) are characterised by a C-terminal SOCS box and a central SPRY domain. The latter is a protein interaction module found in over 1600 proteins, with more than 70 encoded in the human genome. Here we report the crystal structure of the SPRY domain of murine SSB-2 and compare it with the SSB-2 solution structure and crystal structures of other B30.2/SPRY domain-containing family proteins. The structure is a bent beta-sandwich, consisting of two seven-stranded beta-sheets wrapped around a long loop that extends from the centre strands of the inner or concave beta-sheet; it closely matches those of GUSTAVUS and SSB-4. The structure is also similar to those of two recently determined Neuralized homology repeat (NHR) domains (also known as NEUZ domains), with detailed comparisons, suggesting that the NEUZ/NHR domains form a subclass of SPRY domains. The binding site on SSB-2 for the prostate apoptosis response-4 (Par-4) protein has been mapped in finer detail using mutational analyses. Moreover, SSB-1 was shown to have a Par-4 binding surface similar to that identified for SSB-2. Structural perturbations of SSB-2 induced by mutations affecting its interaction with Par-4 and/or c-Met have been characterised by NMR. These comparisons, in conjunction with previously published dynamics data from NMR relaxation studies and coarse-grained dynamics simulation using normal mode analysis, further refine our understanding of the structural basis for protein recognition of SPRY domain-containing proteins.


Subject(s)
DNA-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , DNA Mutational Analysis , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Receptors, Proteinase-Activated/metabolism , Sequence Alignment
9.
Proc Natl Acad Sci U S A ; 104(31): 12737-42, 2007 Jul 31.
Article in English | MEDLINE | ID: mdl-17652170

ABSTRACT

Leukemia inhibitory factor (LIF) receptor is a cell surface receptor that mediates the actions of LIF and other IL-6 type cytokines through the formation of high-affinity signaling complexes with gp130. Here we present the crystal structure of a complex of mouse LIF receptor with human LIF at 4.0 A resolution. The structure is, to date, the largest cytokine receptor fragment determined by x-ray crystallography. The binding of LIF to its receptor via the central Ig-like domain is unlike other cytokine receptor complexes that bind ligand predominantly through their cytokine-binding modules. This structure, in combination with previous crystallographic studies, also provides a structural template to understand the formation and orientation of the high-affinity signaling complex between LIF, LIF receptor, and gp130.


Subject(s)
Immunoglobulins/chemistry , Immunoglobulins/metabolism , Leukemia Inhibitory Factor/chemistry , Leukemia Inhibitory Factor/metabolism , Receptors, OSM-LIF/chemistry , Receptors, OSM-LIF/metabolism , Animals , Crystallography, X-Ray , Cytokine Receptor gp130/chemistry , Cytokine Receptor gp130/metabolism , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Interleukin-6/chemistry , Interleukin-6/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/immunology , Ligands , Mice , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, OSM-LIF/genetics , Receptors, OSM-LIF/immunology , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 103(33): 12429-34, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16894147

ABSTRACT

The insulin receptor (IR) and the type-1 insulin-like growth factor receptor (IGF1R) are homologous multidomain proteins that bind insulin and IGF with differing specificity. Here we report the crystal structure of the first three domains (L1-CR-L2) of human IR at 2.3 A resolution and compare it with the previously determined structure of the corresponding fragment of IGF1R. The most important differences seen between the two receptors are in the two regions governing ligand specificity. The first is at the corner of the ligand-binding surface of the L1 domain, where the side chain of F39 in IR forms part of the ligand binding surface involving the second (central) beta-sheet. This is very different to the location of its counterpart in IGF1R, S35, which is not involved in ligand binding. The second major difference is in the sixth module of the CR domain, where IR contains a larger loop that protrudes further into the ligand-binding pocket. This module, which governs IGF1-binding specificity, shows negligible sequence identity, significantly more alpha-helix, an additional disulfide bond, and opposite electrostatic potential compared to that of the IGF1R.


Subject(s)
Insulin-Like Growth Factor I/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, Insulin/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Crystallography, X-Ray , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Ligands , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Sequence Alignment
11.
Acta Crystallogr D Biol Crystallogr ; 62(Pt 9): 947-52, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16929094

ABSTRACT

The X-ray structure of influenza virus neuraminidase (NA) isolated from whale, subtype N9, has been determined at 2.2 A resolution and contains a tetrameric protein in the asymmetric unit. In structures of NA determined previously, a calcium ion is observed to coordinate amino acids near the substrate-binding site. In three of the NA monomers determined here this calcium is absent, resulting in structural alterations near the substrate-binding site. These changes affect the conformation of residues that participate in several key interactions between the enzyme and substrate and provide at a molecular level the basis of the structural and functional role of calcium in substrate and inhibitor binding. Several sulfate ions were identified in complex with the protein. These are located in the active site, occupying the space reserved for the substrate (sialic acid) carboxylate, and in positions leading away from the substrate-binding site. These sites offer a new opportunity for the design of inhibitors of influenza virus NA.


Subject(s)
Calcium/chemistry , Neuraminidase/chemistry , Orthomyxoviridae/enzymology , Animals , Binding Sites , Crystallography, X-Ray , Ions , Models, Chemical , Models, Molecular , Protein Binding , Protein Conformation , Sulfates/chemistry , Whales/virology , X-Rays
12.
Curr Opin Drug Discov Devel ; 7(5): 630-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15503865

ABSTRACT

The insulin receptor (IR) and epidermal growth factor receptor (EGFR) families are closely related members of the receptor tyrosine kinase superfamily, and are among the more intensively studied proteins in biology. Deregulated signaling by the type-1 insulin-like growth factor receptor (IGF-1R) or members of the EGFR family has been implicated in the progression of a variety of cancers. These receptors have thus emerged as validated therapeutic targets for the development of antitumor agents. In this review, recent progress in the elucidation of the three-dimensional structures of the extracellular domains of both receptor families is discussed. While the IGF-1R provided the first description of the extracellular domains of these two receptor families, it is the EGFR family in which greatest progress has been achieved. Over the past year, the field has progressed from having a complete absence of X-ray crystal structures to having eight such structures; ErbB-2 alone or complexed with the two monoclonal antibodies pertuzumab (Genentech Inc/Roche Holdings AG/Chugai Pharmaceutical Co Ltd) and trastuzamab, ErbB-3 without a ligand, EGFR with a ligand bound in an unactivated monomeric conformation, and EGFR with either epidermal growth factor (EGF) or transforming growth factor-alpha (TGFalpha) in a 2:2 dimeric complex. This review will discuss these developments and the opportunities they provide for the design of new therapeutic agents targeting their extracellular domains.


Subject(s)
ErbB Receptors/chemistry , Protein Conformation , Receptor, Insulin/chemistry , Drug Design , ErbB Receptors/antagonists & inhibitors , Humans , Models, Molecular , Receptor, Insulin/antagonists & inhibitors
13.
J Biol Chem ; 279(21): 22387-98, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15016810

ABSTRACT

Recent crystallographic data on the isolated extracellular domain of the epidermal growth factor receptor (EGFR) have suggested a model for its activation by ligand. We have tested this model in the context of the full-length EGFR displayed at the cell surface, by introducing mutations in two regions (CR1 and CR2) of the extracellular domain thought to be critical for regulation of receptor activation. Mutations in the CR1 and CR2 domains have opposing effects on ligand binding affinity, receptor dimerization, tyrosine kinase activation, and signaling competence. Tyr(246) is a critical residue in the CR1 loop, which is implicated in the positioning and stabilization of the receptor dimer interface after ligand binding; mutations of Tyr(246) impair or abolish receptor function. Mutations in CR2, which weaken the interaction that restricts the receptor to the tethered (inactive) state, enhance responsiveness to EGF by increasing affinity for the ligand. However, weakening of the CR1/CR2 interaction does not result in spontaneous activation of the receptors' kinase. We have used an antibody (mAb 806), which recognizes a transition state of the EGF receptor between the negatively constrained, tethered state and the fully active back-to-back dimer conformation, to follow conformational changes in the wild-type and mutant EGF receptors after ligand binding. Our results suggest that EGFR on the cell surface can be untethered, but this form is inactive; thus, untethering of the receptor is not sufficient for activation, and ligand binding is essential for the correct positioning of the two receptor subunits to achieve kinase activation.


Subject(s)
Cell Membrane/metabolism , ErbB Receptors/physiology , Animals , Cell Line , Cross-Linking Reagents/pharmacology , Dimerization , Enzyme Activation , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Kinetics , Ligands , MAP Kinase Signaling System , Mice , Models, Biological , Models, Molecular , Mutation , Phosphorylation , Point Mutation , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Transfection , Tyrosine/chemistry , Tyrosine/metabolism
14.
Mol Cell ; 12(3): 541-52, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14527402

ABSTRACT

Recent crystallographic studies have provided significant new insight into how receptor tyrosine kinases from the EGF receptor or ErbB family are regulated by their growth factor ligands. EGF receptor dimerization is mediated by a unique dimerization arm, which becomes exposed only after a dramatic domain rearrangement is promoted by growth factor binding. ErbB2, a family member that has no ligand, has its dimerization arm constitutively exposed, and this explains several of its unique properties. We outline a mechanistic view of ErbB receptor homo- and heterodimerization, which suggests new approaches for interfering with these processes when they are implicated in human cancers.


Subject(s)
ErbB Receptors/metabolism , Eukaryotic Cells/metabolism , Growth Substances/metabolism , Oncogene Proteins v-erbB/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Humans , Ligands , Models, Molecular , Molecular Conformation , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Structure, Tertiary
15.
Leuk Lymphoma ; 44(3): 477-81, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12688318

ABSTRACT

As both fludarabine and rituximab are active against indolent lymphoproliferative disorders, we have studied the combination of fludarabine and rituximab in patients with low-grade lymphoma and chronic lymphocytic leukemia (CLL) in phase I/II fashion. Of 33 patients enrolled, 21(63.6%) had low-grade lymphoma and 12 (36.4%) had CLL. They received fludarabine 30 mg/m2 on days 1-4 and rituximab 125, 250 or 375 mg/m2 on day 5 at intervals of 28 days to a maximum of 8 cycles. Three patients were removed from the study because of rituximab-associated anaphylaxis and four because of prolonged hematopoietic toxicity. Toxicity and responsiveness did not differ at the different dose levels of rituximab. For 29 evaluable patients, responses were seen in 82.8% and complete responses in 34.5%. Of 7 responding patients not referred for stem cell transplantation, 6 remain in complete remission at a median follow-up of 16 months (range 4-30 months). Of 13 previously untreated patients, all responded and 46.2% had a complete response. Of 16 previously treated patients, 68.5% responded and 25% had a complete response. The combination of fludarabine and rituximab has major activity and acceptable toxicity in patients with low-grade lymphoma and CLL.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antimetabolites, Antineoplastic/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Lymphoma, Non-Hodgkin/therapy , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use , Adult , Aged , Aged, 80 and over , Anaphylaxis/etiology , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Murine-Derived , Antimetabolites, Antineoplastic/adverse effects , Combined Modality Therapy , Female , Follow-Up Studies , Hematologic Diseases/chemically induced , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Male , Middle Aged , Peripheral Blood Stem Cell Transplantation , Remission Induction , Rituximab , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/adverse effects
16.
Exp Cell Res ; 284(1): 31-53, 2003 Mar 10.
Article in English | MEDLINE | ID: mdl-12648464

ABSTRACT

The epidermal growth factor (EGF) receptor (EGFR) is one of four homologous transmembrane proteins that mediate the actions of a family of growth factors including EGF, transforming growth factor-alpha, and the neuregulins. We review the structure and function of the EGFR, from ligand binding to the initiation of intracellular signalling pathways that lead to changes in the biochemical state of the cell. The recent crystal structures of different domains from several members of the EGFR family have challenged our concepts of these processes.


Subject(s)
ErbB Receptors/metabolism , Signal Transduction , Animals , Cell Movement , Humans , Models, Molecular , Protein Conformation , Transforming Growth Factors/metabolism
17.
Mol Cell ; 11(2): 495-505, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12620236

ABSTRACT

ErbB2 does not bind ligand, yet appears to be the major signaling partner for other ErbB receptors by forming heteromeric complexes with ErbB1, ErbB3, or ErbB4. The crystal structure of residues 1-509 of ErbB2 at 2.5 A resolution reveals an activated conformation similar to that of the EGFR when complexed with ligand and very different from that seen in the unactivated forms of ErbB3 or EGFR. The structure explains the inability of ErbB2 to bind known ligands and suggests why ErbB2 fails to form homodimers. Together, the data suggest a model in which ErbB2 is already in the activated conformation and ready to interact with other ligand-activated ErbB receptors.


Subject(s)
Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , CHO Cells , Cricetinae , Crystallography, X-Ray , DNA, Complementary/genetics , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , In Vitro Techniques , Ligands , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Structure, Tertiary , Receptor, ErbB-2/genetics , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Static Electricity
18.
J Mol Biol ; 326(2): 341-51, 2003 Feb 14.
Article in English | MEDLINE | ID: mdl-12559905

ABSTRACT

Diabodies (scFv dimers) are small, bivalent antibody mimetics of approximately 55kDa in size that possess rapid in vivo targeting pharmacokinetics compared to the intact parent antibody, and may prove highly suitable for imaging and therapeutic applications. Here, we describe T84.66Di, the first diabody crystal structure in which the scFvs comprise V domains linked in the V(L)-to-V(H) orientation. The structure was determined by X-ray diffraction analysis to 2.6 A resolution. The T84.66Di scFv was constructed from the anti-carcinoembryonic antigen (anti-CEA) antibody T84.66 variable domains connected by an eight residue peptide linker to provide flexibility between Fv modules and promote dimer formation with bivalent affinity to the cell-surface target, CEA. Therefore, it was surprising to observe a close association of some Fv module complementarity-determining regions in the T84.66 diabody crystal, especially compared to other diabody structures all of which are linked in the opposite V(H)-to-V(L) orientation. The differences between the arrangement of Fv modules in the T84.66Di V(L)-to-V(H) linked diabody structure compared to the crystal structure of L5MK16 and other proposed V(H)-to-V(L) linked diabodies has been investigated and their potential for flexibility discussed. The comparison between V(H)-to-V(L) and V(L)-to-V(H) linked diabodies revealed in this study represents a limited repertoire of possible diabody Fv orientations, but one that reveals the potential flexibility of these molecules. This analysis therefore provides some signposts that may impact on future molecular designs for these therapeutic molecules with respect to diabody flexibility and avidity.


Subject(s)
Antigens, Neoplasm/immunology , Carcinoembryonic Antigen/immunology , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/immunology , Antibodies, Bispecific , Antigen-Antibody Reactions , Antigens, Neoplasm/metabolism , Crystallization , Crystallography, X-Ray , Humans , Immunoglobulin Fc Fragments , Immunoglobulin Fragments/immunology , Immunoglobulin Heavy Chains/immunology , Isoenzymes/immunology , Models, Molecular , Neuraminidase/immunology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Phospholipase C delta , Protein Conformation , Recombinant Fusion Proteins , Type C Phospholipases/immunology
19.
Cell ; 110(6): 763-73, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12297049

ABSTRACT

We report the crystal structure, at 2.5 A resolution, of a truncated human EGFR ectodomain bound to TGFalpha. TGFalpha interacts with both L1 and L2 domains of EGFR, making many main chain contacts with L1 and interacting with L2 via key conserved residues. The results indicate how EGFR family members can bind a family of highly variable ligands. In the 2:2 TGFalpha:sEGFR501 complex, each ligand interacts with only one receptor molecule. There are two types of dimers in the asymmetric unit: a head-to-head dimer involving contacts between the L1 and L2 domains and a back-to-back dimer dominated by interactions between the CR1 domains of each receptor. Based on sequence conservation, buried surface area, and mutagenesis experiments, the back-to-back dimer is favored to be biologically relevant.


Subject(s)
ErbB Receptors/chemistry , ErbB Receptors/metabolism , Models, Molecular , Transforming Growth Factor alpha/chemistry , Transforming Growth Factor alpha/metabolism , 3T3 Cells , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Conserved Sequence , Crystallization , Crystallography, X-Ray , Dimerization , Disulfides/chemistry , Humans , Ligands , Mice , Molecular Sequence Data , Molecular Structure , Mutation , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Transforming Growth Factor alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...