Network


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

Hotspot


Dive into the research topics where David E. Reichert is active.

Publication


Featured researches published by David E. Reichert.


Coordination Chemistry Reviews | 1999

Metal complexes as diagnostic tools

David E. Reichert; Jason S. Lewis; Carolyn J. Anderson

Abstract This review summarizes some of the developments of metal complexes and metal-complex-bioconjugates for the diagnosis of disease states that have occurred over the past 10 years. The diagnostic imaging modalities discussed are gamma scintigraphy, positron emission tomography (PET) and magnetic resonance imaging (MRI). Metal complexes are utilized in all three imaging modalities to image a broad array of diseases, including heart disease, brain disorders and cancer. There are a wide variety of different radiometals that have been utilized in the synthesis of coordination compounds for gamma scintigraphy and PET, and these will be discussed individually. The field of metal-complex-bioconjugate chemistry is covered extensively, describing radiometals labeled to biomolecules such as receptor ligands for diseases such as neurological disorders and cancer. A section is devoted to the development of coordination compounds for MRI enhancement agents, and specifically details the agents that have been evaluated in vivo, both in animal models and in humans. Overall, the goal of this review is to demonstrate the significant progress made in the field of coordination chemistry as it applies to the development of diagnostic imaging agents.


Journal of Immunology | 2004

Staging the Initiation of Autoantibody-Induced Arthritis: A Critical Role for Immune Complexes

Brian T. Wipke; Zheng Wang; Wouter Nagengast; David E. Reichert; Paul M. Allen

In the K/B×N mouse model of arthritis, autoantibodies against glucose-6-phosphate isomerase cause joint-specific inflammation and destruction. We have shown using micro-positron emission tomography that these glucose-6-phosphate isomerase-specific autoantibodies rapidly localize to distal joints of mice. In this study we used micro-positron emission tomography to delineate the stages involved in the development of arthritis. Localization of Abs to the joints depended upon mast cells, neutrophils, and FcRs, but not on C5. Surprisingly, anti-type II collagen Abs alone did not accumulate in the distal joints, but could be induced to do so by coinjection of irrelevant preformed immune complexes. Control Abs localized to the joint in a similar manner. Thus, immune complexes are essential initiators of arthritis by sequential activation of neutrophils and mast cells to allow Abs access to the joints, where they must bind a target Ag to initiate inflammation. Our findings support a four-stage model for the development of arthritis and identify checkpoints where the disease is reversible.


Nuclear Medicine and Biology | 2003

Preparation of 66Ga- and 68Ga-labeled Ga(III)-deferoxamine-folate as potential folate-receptor-targeted PET radiopharmaceuticals

Carla J. Mathias; Michael R. Lewis; David E. Reichert; Richard Laforest; Terry L. Sharp; Jason S. Lewis; Zhen Fan Yang; David J. Waters; Paul W. Snyder; Philip S. Low; Michael J. Welch; Mark A. Green

A folate-receptor-targeting radiopharmaceutical, Ga(III)-deferoxamine-folate (Ga-DF-Folate), was radiolabeled with two positron-emitting isotopes of gallium, cyclotron-produced (66)Ga (9.5 hour half-life) and generator-produced (68)Ga (68 minute half-life). The [(66)Ga]Ga-DF-Folate was administered to athymic mice with folate-receptor-positive human KB cell tumor xenografts to demonstrate that microPET mouse tumor imaging is feasible with (66)Ga, despite the relatively high positron energy of this radionuclide. Using the athymic mouse KB tumor xenograft model, dual-isotope autoradiography was also performed following i.v. co-administration of [(18)F]-FDG, a marker of regional metabolic activity, and folate-receptor-targeted [(111)In]In-DTPA-Folate. The autoradiographic images of 1 mm tumor sections demonstrate the gross heterogeneity of the KB cell tumor xenograft, as well as subtle disparity in the regional accumulation of the two radiotracers.


Molecular Pharmacology | 2008

Mutations of the GABA-A receptor α1 subunit M1 domain reveal unexpected complexity for modulation by neuroactive steroids

Gustav Akk; Ping Li; John Bracamontes; David E. Reichert; Douglas F. Covey; Joe Henry Steinbach

Neuroactive steroids are among the most efficacious modulators of the mammalian GABA-A receptor. Previous work has proposed that receptor potentiation is mediated by steroid interactions with a site defined by the residues α1Asn407/Tyr410 in the M4 transmembrane domain and residue α1Gln241 in the M1 domain. We examined the role of residues in the α1 subunit M1 domain in the modulation of the rat α1β2γ2L GABA-A receptor by neuroactive steroids. The data demonstrate that the region is critical to the actions of potentiating neuroactive steroids. Receptors containing the α1Q241W or α1Q241L mutations were insensitive to (3α,5α)-3-hydroxypregnan-20-one (3α5αP), albeit with different underlying mechanisms. The α1Q241S mutant was potentiated by 3α5αP, but the kinetic mode of potentiation was altered by the mutation. It is noteworthy that the α1Q241L mutation had no effect on channel potentiation by (3α,5α)-3-hydroxymethyl-pregnan-20-one, but mutation of the neighboring residue, α1Ser240, prevented channel modulation. A steroid lacking an H-bonding group on C3 (5α-pregnan-20-one) potentiated the wild-type receptor but not the α1Q241L mutant. The findings are consistent with a model in which the α1Ser240 and α1Gln241 residues shape the surface to which steroid molecules bind.


Organic and Biomolecular Chemistry | 2009

[18F]- and [11C]-Labeled N-benzyl-isatin sulfonamide analogues as PET tracers for Apoptosis: synthesis, radiolabeling mechanism, and in vivo imaging study of apoptosis in Fas-treated mice using [11C]WC-98

Dong Zhou; Wenhua Chu; Delphine L. Chen; Qi Wang; David E. Reichert; Justin Rothfuss; André D'avignon; Michael J. Welch; Robert H. Mach

The radiolabeled isatin sulfonamide caspase-3 inhibitor, [18F] 2 (WC-II-89), is a potential PET radiotracer for noninvasive imaging of apoptosis. The radiolabeling mechanism was studied by 13C NMR, ESI/MS, and computational calculations. It was found that the high electrophilicity of the C3 carbonyl group in the isatin ring, which served as a trap for [18F]fluoride, was responsible for the failure of the radiolabeling via nucleophilic substitution of the mesylate group in 7a by [18F]fluoride. Once treated with a strong base, 7a opened the isatin ring completely to form an isatinate intermediate 16, which lost the ability to trap [18F]fluoride, thereby allowing the displacement of the mesylate group to afford the 18F-labeled isatinate 17. [18F] 17 can be converted to isatin [18F] 2 efficiently under acidic conditions. The ring-opening and re-closure of the isatin ring under basic and acidic conditions were confirmed by reversed phase HPLC analysis, ESI/MS and 13C NMR studies. Computational studies of model compounds also support the above proposed mechanism. Similarly, the ring-opening and re-closure method was used successfully in the synthesis of the 11C labeled isatin sulfonamide analogue [11C] 4 (WC-98). A microPET imaging study using [11C] 4 in the Fas liver apoptosis model demonstrated retained activity in the target organ (liver) of the treated mice. Increased caspase-3 activation in the liver was verified by the fluorometric caspase-3 enzyme assay. Therefore, this study provides a useful method for radio-synthesis of isatin derivative radiotracers for PET and SPECT studies, and [11C] 4 is a potential PET radiotracer for noninvasive imaging of apoptosis.


Molecular Pharmacology | 2012

Neurosteroid Analog Photolabeling of a Site in the Third Transmembrane Domain of the β3 Subunit of the GABAA Receptor

Zi-Wei Chen; Brad D. Manion; R. Reid Townsend; David E. Reichert; Douglas F. Covey; Joe Henry Steinbach; Werner Sieghart; Karoline Fuchs; Alex S. Evers

Accumulated evidence suggests that neurosteroids modulate GABAA receptors through binding interactions with transmembrane domains. To identify these neurosteroid binding sites directly, a neurosteroid-analog photolabeling reagent, (3α,5β)-6-azi-pregnanolone (6-AziP), was used to photolabel membranes from Sf9 cells expressing high-density, recombinant, His8-β3 homomeric GABAA receptors. 6-AziP inhibited 35S-labeled t-butylbicyclophosphorothionate binding to the His8-β3 homomeric GABAA receptors in a concentration-dependent manner (IC50 = 9 ± 1 μM), with a pattern consistent with a single class of neurosteroid binding sites. [3H]6-AziP photolabeled proteins of 30, 55, 110, and 150 kDa, in a concentration-dependent manner. The 55-, 110-, and 150-kDa proteins were identified as His8-β3 subunits through immunoblotting and through enrichment on a nickel affinity column. Photolabeling of the β3 subunits was stereoselective, with [3H]6-AziP producing substantially greater labeling than an equal concentration of its diastereomer [3H](3β,5β)-6-AziP. High-resolution mass spectrometric analysis of affinity-purified, 6-AziP-labeled His8-β3 subunits identified a single photolabeled peptide, ALLEYAF-6-AziP, in the third transmembrane domain. The identity of this peptide and the site of incorporation on Phe301 were confirmed through high-resolution tandem mass spectrometry. No other sites of photoincorporation were observed despite 90% sequence coverage of the whole β3 subunit protein, including 84% of the transmembrane domains. This study identifies a novel neurosteroid binding site and demonstrates the feasibility of identifying neurosteroid photolabeling sites by using mass spectrometry.


Nuclear Medicine and Biology | 2002

Production and purification of gallium-66 for preparation of tumor-targeting radiopharmaceuticals

Michael R. Lewis; David E. Reichert; Richard Laforest; William Margenau; Ruth E. Shefer; Robert E. Klinkowstein; Barbara J. Hughey; Michael J. Welch

Gallium-66 (T(1/2) = 9.49 h) is an intermediate-lived radionuclide that has potential for positron emission tomography (PET) imaging of biological processes with intermediate to slow target tissue uptake. We have produced (66)Ga by the (66)Zn(p,n) (66)Ga nuclear reaction using a small biomedical cyclotron and have investigated methods for purifying (66)Ga that could be applied to the development of an automated processing system. Measured yields of (66)Ga were very high with a production yield of nearly 14 mCi/microA-h at 14.5 MeV bombardment energy, a value in excellent agreement with theoretical predictions based on literature cross sections for the (66)Zn(p,n) (66)Ga reaction. Gallium-66 has been purified from irradiated zinc targets two ways, by cation-exchange chromatography and diisopropyl ether extraction. The concentrations of stable contaminants in (66)Ga following the two processing methods were determined, and it was found that iron and zinc were present at levels up to an order of magnitude higher after cation-exchange chromatography. The bioconjugates DOTA-Tyr(3)-octreotide and DOTA-biotin were labeled with (66)Ga purified by both methods. Following purification of (66)Ga by solvent extraction, radiochemical yields in excess of 85% were obtained for both compounds, in contrast to much lower labeling yields (less than 20%) obtained after the cation-exchange separation. Higher concentrations of stable contaminants likely contributed to the poor radiochemical yields for labeling DOTA-Tyr(3)-octreotide and DOTA-biotin with cation-exchanged (66)Ga. The lower purity and radiolabeling yields obtained using cation-exchange do not warrant the development of an automated processing system based on this method. Therefore, work is in progress to automate the diisopropyl ether extraction method for routine processing of (66)Ga.


Journal of Computer-aided Molecular Design | 2003

CoMFA and docking study of novel estrogen receptor subtype selective ligands.

Peter Wolohan; David E. Reichert

We present the results from a Comparative Molecular Field Analysis (CoMFA) and docking study of a diverse set of 36 estrogen receptor ligands whose relative binding affinities (RBA) with respect to 17β-Estradiol were available in both isoforms of the nuclear estrogen receptors (ERα, ERβ). Initial CoMFA models exhibited a correlation between the experimental relative binding affinities and the molecular steric and electrostatic fields; ERα: r2=0.79, q2=0.44 ERβ: r2=0.93, q2=0.63. Addition of the solvation energy of the isolated ligand improved the predictive nature of the ERβ model initially; r2=0.96, q2=0.70 but upon rescrambling of the data-set and reselecting the training set at random, inclusion of the ligand solvation energy was found to have little effect on the predictive nature of the CoMFA models. The ligands were then docked inside the ligand binding domain (LBD) of both ERα and ERβ utilizing the docking program Gold, after-which the program CScore was used to rank the resulting poses. Inclusion of both the Gold and CScore scoring parameters failed to improve the predictive ability of the original CoMFA models. The subtype selectivity expressed as RBA(ERα/ERβ) of the test sets was predicted using the most predictive CoMFA models, as illustrated by the cross-validated r2. In each case the most selective ligands were ranked correctly illustrating the utility of this method as a prescreening tool in the development of novel estrogen receptor subtype selective ligands.


Nuclear Medicine and Biology | 1999

Evaluation of Gallium-68 Tris(2-Mercaptobenzyl)Amine: A Complex with Brain and Myocardial Uptake

Cathy S. Cutler; M.Cecilia Giron; David E. Reichert; Abraham Z. Snyder; Pilar Herrero; Carolyn J. Anderson; Duncan A. Quarless; Stephen A. Koch; Michael J. Welch

Previous research into development of a gallium-radiolabeled agent that crosses the blood-brain barrier has met with limited success. In this study, we focused our attention on a Ga(III) complex of a 4-coordinate amine trithiolate tripod ligand, tris(2-mercaptobenzyl) amine (S3N). The Ga(III) S3N complex is small, neutral, and lipophilic, meeting the requirements for a potential brain imaging agent. The Ga-68 complex was easily formed with a radiochemical purity of >95%. In vitro stability of the Ga-S3N complex, determined in rat serum incubated at 37 degrees C, was greater than 95% intact at 2 h by silica gel and reversed-phase radio-thin layer chromatography. Biodistribution studies conducted in female Sprague-Dawley rats showed the complex cleared rapidly from the blood with initial high liver uptake followed by rapid washout. Significant uptake was observed in the brain, with brain:blood ratios increasing from 0.11 at 2 min postinjection to 3.8 at 60 min postinjection. Uptake was also observed in the heart going from a heart:blood ratio of 2.3 at 2 min postinjection to 11 at 60 min postinjection. Molecular mechanics were used to determine the coordination number, and demonstrated that the Ga(III) complex prefers to be 4-coordinate. Imaging studies with 68Ga-S3N in a Nemestrina macaque showed significant brain uptake, similar to other lipophilic agents. The extraction of 68Ga-S3N into the brains of both rodents and primates, higher than any 68Ga agent reported in the literature, suggests that this compound may have potential as a brain imaging agent for positron emission tomography.


Nuclear Medicine and Biology | 2013

Microfluidic radiolabeling of biomolecules with PET radiometals

Dexing Zeng; Amit V. Desai; David Ranganathan; Tobias D. Wheeler; Paul J. A. Kenis; David E. Reichert

INTRODUCTION A robust, versatile and compact microreactor has been designed, fabricated and tested for the labeling of bifunctional chelate conjugated biomolecules (BFC-BM) with PET radiometals. METHODS The developed microreactor was used to radiolabel a chelate, either 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) or 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) that had been conjugated to cyclo(Arg-Gly-Asp-DPhe-Lys) peptide, with both ⁶⁴Cu and ⁶⁸Ga respectively. The microreactor radiolabeling conditions were optimized by varying temperature, concentration and residence time. RESULTS Direct comparisons between the microreactor approach and conventional methods showed improved labeling yields and increased reproducibility with the microreactor under identical labeling conditions, due to enhanced mass and heat transfer at the microscale. More importantly, over 90% radiolabeling yields (incorporation of radiometal) were achieved with a 1:1 stoichiometry of bifunctional chelate biomolecule conjugate (BFC-BM) to radiometal in the microreactor, which potentially obviates extensive chromatographic purification that is typically required to remove the large excess of unlabeled biomolecule in radioligands prepared using conventional methods. Moreover, higher yields for radiolabeling of DOTA-functionalized BSA protein (Bovine Serum Albumin) were observed with ⁶⁴Cu/⁶⁸Ga using the microreactor, which demonstrates the ability to label both small and large molecules. CONCLUSIONS A robust, reliable, compact microreactor capable of chelating radiometals with common chelates has been developed and validated. Based on our radiolabeling results, the reported microfluidic approach overall outperforms conventional radiosynthetic methods, and is a promising technology for the radiometal labeling of commonly utilized BFC-BM in aqueous solutions.

Collaboration


Dive into the David E. Reichert's collaboration.

Top Co-Authors

Avatar

Michael J. Welch

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Hairong Li

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Dexing Zeng

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Robert H. Mach

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Alex S. Evers

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

David Ranganathan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Douglas F. Covey

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Haiying Zhou

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Paul Kenis

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Qi Wang

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge