Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Virol ; 90(8): 4078-4092, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26865716

ABSTRACT

UNLABELLED: Mengovirus, a member of thePicornaviridaefamily, has a broad cell tropism and can cause encephalitis and myocarditis in multiple mammalian species. Attenuation has been achieved by shortening the polycytidine tract in the 5' noncoding region (NCR). A poly(C)-truncated strain of mengovirus, vMC24, resulted in significant tumor regression in immunocompetent BALB/c mice bearing syngeneic MPC-11 plasmacytomas, but the associated toxicities were unacceptable. To enhance its safety profile, microRNA target sequences complementary to miR-124 or miR-125 (enriched in nervous tissue), miR-133 and miR-208 (enriched in cardiac tissue), or miR-142 (control; enriched in hematopoietic tissues) were inserted into the vMC24NCRs. The microRNA-detargeted viruses showed reduced replication and cell killing specifically in cells expressing the cognate microRNAs, but certain insertions additionally were associated with nonspecific suppression of viral fitnessin vivo. In vivotoxicity testing confirmed that miR-124 targets within the 5' NCR suppressed virus replication in the central nervous system while miR-133 and miR-208 targets in the 3' NCR suppressed viral replication in cardiac tissue. A dual-detargeted virus named vMC24-NC, with miR-124 targets in the 5' NCR and miR-133 plus miR-208 targets in the 3' NCR, showed the suppression of replication in both nervous and cardiac tissues but retained full oncolytic potency when administered by intratumoral (10(6)50% tissue culture infectious doses [TCID50]) or intravenous (10(7)to 10(8)TCID50) injection into BALB/c mice bearing MPC-11 plasmacytomas. Overall survival of vMC24-NC-treated tumor-bearing mice was significantly improved compared to that of nontreated mice. MicroRNA-detargeted mengoviruses offer a promising oncolytic virotherapy platform that merits further development for clinical translation. IMPORTANCE: The clinical potential of oncolytic virotherapy for cancer treatment has been well demonstrated, justifying the continued development of novel oncolytic viruses with enhanced potency. Here, we introduce mengovirus as a novel oncolytic agent. Mengovirus is appealing as an oncolytic virotherapy platform because of its small size, simple genome structure, rapid replication cycle, and broad cell/species tropism. However, mengovirus can cause encephalomyelitis and myocarditis. It can be partially attenuated by shortening the poly(C) tract in the 5' NCR but remains capable of damaging cardiac and nervous tissue. Here, we further enhanced the safety profile of a poly(C)-truncated mengovirus by incorporating muscle- and neuron-specific microRNA target sequences into the viral genome. This dual-detargeted virus has reduced pathogenesis but retained potent oncolytic activity. Our data show that microRNA targeting can be used to further increase the safety of an attenuated mengovirus, providing a basis for its development as an oncolytic platform.


Subject(s)
Mengovirus , MicroRNAs/genetics , Multiple Myeloma/therapy , Oncolytic Virotherapy , Animals , Cardiovirus Infections/etiology , Cardiovirus Infections/prevention & control , Cell Line , Cytopathogenic Effect, Viral , Female , Gene Targeting , Genomic Instability , Humans , Immunocompromised Host , Mengovirus/genetics , Mice , Mice, Inbred BALB C , Multiple Myeloma/immunology , Neurotoxicity Syndromes/prevention & control , Neurotoxicity Syndromes/virology , Oncolytic Virotherapy/adverse effects , RNA, Untranslated/genetics , Virus Replication
2.
J Virol ; 87(7): 3752-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23325695

ABSTRACT

We are developing oncolytic vesicular stomatitis viruses (VSVs) for systemic treatment of multiple myeloma, an incurable malignancy of antibody-secreting plasma cells that are specifically localized in the bone marrow. One of the presumed advantages for using VSV as an oncolytic virus is that human infections are rare and preexisting anti-VSV immunity is typically lacking in cancer patients, which is very important for clinical success. However, our studies show that nonimmune human and mouse serum can neutralize clinical-grade VSV, reducing the titer by up to 4 log units in 60 min. In addition, we show that neutralizing anti-VSV antibodies negate the antitumor efficacy of VSV, a concern for repeat VSV administration. We have investigated the potential use of covalent modification of VSV with polyethylene glycol (PEG) or a function-spacer-lipid (FSL)-PEG construct to inhibit serum neutralization and to limit hepatosplenic sequestration of systemically delivered VSV. We report that in mice passively immunized with neutralizing anti-VSV antibodies, PEGylation of VSV improved the persistence of VSV in the blood circulation, maintaining a more than 1-log-unit increase in VSV genome copies for up to 1 h compared to the genome copy numbers for the non-PEGylated virus, which was mostly cleared within 10 min after intravenous injection. We are currently investigating if this increase in PEGylated VSV circulating half-life can translate to increased virus delivery and better efficacy in mouse models of multiple myeloma.


Subject(s)
Multiple Myeloma/therapy , Oncolytic Virotherapy/methods , Polyethylene Glycols/pharmacology , Vesicular stomatitis Indiana virus/metabolism , Viremia/blood , Animals , Chlorocebus aethiops , Chromatography , DNA Primers/genetics , Drug Discovery/methods , Green Fluorescent Proteins , Humans , Interferon-beta , Kaplan-Meier Estimate , Mice , Multiple Myeloma/immunology , Neutralization Tests , Real-Time Polymerase Chain Reaction , Vero Cells , Vesicular stomatitis Indiana virus/immunology
3.
J Virol Methods ; 176(1-2): 78-84, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21703308

ABSTRACT

Tools that can aid in vitro and in vivo imaging and also noninvasively determine half-life and biodistribution are required to advance clinical developments. A Function-Spacer-Lipid construct (FSL) incorporating fluorescein (FSL-FLRO4) was used to label vesicular stomatitis virus (VSV), measles virus MV-NIS (MV) and influenza virus (H1N1). The ability of FSL constructs to label these virions was established directly by FACScan of FSL-FLRO4 labeled VSV and MV, and indirectly following labeled H1N1 and MV binding to a cells. FSL-FLRO4 labeling of H1N1 was shown to maintain higher infectivity of the virus when compared with direct fluorescein virus labeling. A novel tyrosine (125)I radioiodinated FSL construct was synthesized (FSL-(125)I) from FSL-tyrosine. This was used to label VSV (VSV-FSL-(125)I), which was infused into the peritoneal cavity of laboratory mice. Bioscanning showed VSV-FSL-(125)I to localize in the liver, spleen and bloodstream in contrast to the free labels FSL-(125)I or (125)I, which localized predominantly in the liver and thyroid respectively. This is a proof-of-principle novel and rapid method for modifying virions and demonstrates the potential of FSL constructs to improve in vivo imaging of virions and noninvasively observe in vivo biodistribution.


Subject(s)
Flow Cytometry/methods , Fluorescein/chemistry , Iodine Radioisotopes/chemistry , Lipids/chemistry , Staining and Labeling/methods , Virion/chemistry , Animals , Biomarkers/chemistry , Cell Line , Chlorocebus aethiops , Female , Humans , Image Processing, Computer-Assisted , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/pathogenicity , Measles virus/chemistry , Mice , Mice, Inbred C57BL , Tissue Distribution , Vero Cells , Vesicular stomatitis Indiana virus/chemistry , Vesicular stomatitis Indiana virus/pathogenicity
4.
Mol Ther ; 19(6): 1041-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21505425

ABSTRACT

The feasibility of using a nonviral vector formulation to initiate an oncolytic viral infection has not been previously demonstrated. We therefore sought to determine whether infectious nucleic acid (INA) could be used in place of virus particles to initiate an oncolytic picornavirus infection in vivo. Infectious RNA encoding coxsackievirus A21 (CVA21) was transcribed from plasmid DNA using T7 polymerase. Within 48 hours of injecting this RNA into KAS6/1 myeloma xenografts, high titers of infectious CVA21 virions were detected in the bloodstream. Tumors regressed rapidly thereafter and mice developed signs of myositis. At euthanasia, CVA21 was recovered from regressing tumors and from skeletal muscles. Treatment outcomes were comparable following intratumoral injection of naked RNA or fully infectious CVA21 virus. Dose-response studies showed that an effective oncolytic infection could be established by intratumoral injection of 1 µg of infectious RNA. The oncolytic infection could also be initiated by intravenous injection of infectious RNA. Our study demonstrates that INA is a highly promising alternative drug formulation for oncolytic virotherapy.


Subject(s)
Enterovirus/genetics , Genetic Vectors/genetics , Multiple Myeloma/therapy , RNA, Viral/physiology , Animals , Cell Line, Tumor , HeLa Cells , Humans , Mice , Mice, SCID , Oncolytic Virotherapy , RNA, Viral/genetics
5.
PLoS Pathog ; 6(3): e1000820, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20333250

ABSTRACT

In addition to modulating the function and stability of cellular mRNAs, microRNAs can profoundly affect the life cycles of viruses bearing sequence complementary targets, a finding recently exploited to ameliorate toxicities of vaccines and oncolytic viruses. To elucidate the mechanisms underlying microRNA-mediated antiviral activity, we modified the 3' untranslated region (3'UTR) of Coxsackievirus A21 to incorporate targets with varying degrees of homology to endogenous microRNAs. We show that microRNAs can interrupt the picornavirus life-cycle at multiple levels, including catalytic degradation of the viral RNA genome, suppression of cap-independent mRNA translation, and interference with genome encapsidation. In addition, we have examined the extent to which endogenous microRNAs can suppress viral replication in vivo and how viruses can overcome this inhibition by microRNA saturation in mouse cancer models.


Subject(s)
Cancer Vaccines/genetics , Coxsackievirus Infections/virology , Enterovirus/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Melanoma/therapy , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Disease Models, Animal , Enterovirus/growth & development , HeLa Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/virology , Melanoma/virology , Mice , Mice, SCID , Molecular Sequence Data , Mutagenesis , Neoplasm Transplantation , Protein Biosynthesis/genetics , RNA Interference , RNA, Viral/genetics , Transplantation, Heterologous
6.
AJR Am J Roentgenol ; 192(1): 279-87, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19098211

ABSTRACT

OBJECTIVE: Our objectives were to, first, determine the oncolytic potential of an engineered measles virus expressing the sodium-iodide symporter gene (MV-NIS) for intratumoral (i.t.) therapy of pancreatic cancer and, second, evaluate NIS as a reporter gene for in vivo monitoring and quantitation of MV-NIS delivery, viral spread, and gene expression in this tumor model. MATERIALS AND METHODS: Cultured human pancreatic cancer cells were infected with MV-NIS. Light microscopy, cell viability, and iodide uptake assays were used to confirm viral infection and NIS gene expression and function in vitro. Human pancreatic tumor xenografts were established in mice and infected via i.t. MV-NIS injections. NIS-mediated i.t. iodide uptake was quantitated by (123)I micro-SPECT/CT. i.t. MV-NIS infection was confirmed by immunohistochemistry of excised pancreatic xenografts. The oncolytic efficacy of MV-NIS was determined by measurement of tumor growth and mouse survival. RESULTS: Infection of human pancreatic cancer cell lines with MV-NIS in vitro resulted in syncytia formation, marked iodide uptake, and ultimately cell death. Tumor xenografts infected with MV-NIS concentrated radioiodine, allowing serial quantitative imaging with (123)I micro-SPECT/CT. i.t. MV-NIS therapy of human pancreatic cancer xenografts resulted in a significant reduction in tumor volume and increased survival time of the treated mice compared with the control mice. CONCLUSION: MV-NIS efficiently infects human pancreatic tumor cells and results in sufficient radioiodine uptake to enable noninvasive serial imaging and quantitation of the intensity, distribution, and time course of NIS gene expression. MV-NIS also shows oncolytic activity in human pancreatic cancer xenografts: Tumor growth is reduced and survival is increased in mice treated with the virus.


Subject(s)
Genetic Therapy/methods , Measles virus/metabolism , Molecular Probe Techniques , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/therapy , Symporters/metabolism , Symporters/therapeutic use , Animals , Cell Line, Tumor , Cell Survival , Female , Humans , Measles virus/genetics , Mice , Mice, Nude , Pancreatic Neoplasms/virology , Protein Engineering/methods , Radionuclide Imaging , Transfection/methods
7.
Nat Med ; 14(11): 1278-83, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18953352

ABSTRACT

The cellular tropisms of eukaryotic viruses are shaped by their need for entry receptors and intracellular transcription factors. Here we show that viral tropisms can also be regulated by tissue-specific microRNAs (miRNAs). Target sequences complementary to muscle-specific miRNAs were inserted into the 3' untranslated region (UTR) of an oncolytic picornavirus that causes lethal myositis in tumor-bearing mice. The recombinant virus still propagated in subcutaneous tumors, causing total regression and sustained viremia, but could not replicate in cells expressing complementary miRNAs and therefore did not cause myositis. This altered tropism was not due to insertional attenuation, as a control virus containing a 3' UTR insert with a disrupted miRNA target sequence fully retained its lethal myotropism. Tissue-specific destabilization of viral genomes by miRNA target insertion provides a potentially versatile new mechanism for controlling the tropism of replicating viruses for therapy and may serve as a new modality for attenuating viruses for vaccine purposes.


Subject(s)
Enterovirus/genetics , Enterovirus/pathogenicity , Genetic Engineering/methods , Genetic Therapy/methods , MicroRNAs/genetics , Animals , Base Sequence , Cell Line , Down-Regulation , Humans , Mice , Mice, SCID , Molecular Sequence Data , Myositis/genetics , Myositis/metabolism , Myositis/pathology
8.
Mol Imaging Biol ; 8(6): 324-32, 2006.
Article in English | MEDLINE | ID: mdl-17053863

ABSTRACT

PURPOSE: This study was undertaken to determine the ability of micro-single photon emission computed tomography (micro-SPECT)/computed tomography (CT) to accurately quantitate intratumoral radioisotope uptake in vivo and to compare these measurements with planar imaging and micro-SPECT imaging alone. PROCEDURES: Human pancreatic cancer xenografts were established in 10 mice. Intratumoral radioisotope uptake was achieved via intratumoral injection of an attenuated measles virus vector expressing the NIS gene (MV-NIS). On various days after MV-NIS injection, (123)I planar and micro-SPECT/CT imaging was performed. Tumor activity was determined by dose calibrator measurements and region-of-interest (ROI) image analysis. Agreement and reproducibility of tumor activity measurements were assessed by Bland-Altman plots and Lin's concordance correlation coefficient (CCC). RESULTS: Intratumoral radioisotope uptake was detected in all mice. Scatterplots demonstrate strong agreement (CCC = 0.93) between micro-SPECT/CT ROI image analysis and dose calibrator tumor activity measurements. The differences between dose calibrator activity measurements and those obtained with ROI image analysis of micro-SPECT alone and planar imaging are less accurate and more variable (CCC = 0.84 and 0.78, respectively). CONCLUSIONS: Micro-SPECT/CT can be used to accurately quantify intratumoral radioisotope uptake in vivo and is more reliable than planar or micro-SPECT imaging alone.


Subject(s)
Iodine Radioisotopes/pharmacokinetics , Tomography, Emission-Computed, Single-Photon/methods , Animals , Dose-Response Relationship, Drug , Female , Mice , Mice, Nude , Subtraction Technique , Tomography, Emission-Computed, Single-Photon/standards
9.
J Med Chem ; 49(3): 850-63, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16451051

ABSTRACT

An understanding of the molecular basis of drug action provides opportunities for refinement of drug properties and for development of more potent and selective molecules that act at the same biological target. In this work, we have identified the active enantiomers in racemic mixtures of structurally related benzophenone derivatives of 1,5-benzodiazepines, representing both antagonist and agonist ligands of the type A cholecystokinin receptor. The parent compounds of the 1,5-benzodiazepine CCK receptor photoaffinity ligands were originally prepared in an effort to develop orally active drugs. The enantiomeric compounds reported in this study selectively photoaffinity-labeled the CCK receptor, resulting in the identification of a site of attachment for the photolabile moiety of the antagonist probe deep within the receptor's membrane-spanning region at Leu(88), a residue within transmembrane segment two. In contrast, the agonist probe labeled a region including extracellular loop one and a portion of transmembrane segment three. The antagonist covalent attachment site to the receptor served as a guide in the construction of theoretical three-dimensional molecular models for the antagonist-receptor complex. These models provided a means for visualization of physically plausible ligand-receptor interactions in the context of all currently available biological data that address small molecule interactions with the CCK receptor. Our approach, featuring the use of novel photolabile compounds targeting the membrane-spanning receptor domain to probe the binding site region, introduces powerful tools and a strategy for direct and selective investigation of nonpeptidyl ligand binding to peptide receptors.


Subject(s)
Benzodiazepines/chemical synthesis , Benzophenones/chemical synthesis , Photoaffinity Labels/chemical synthesis , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/antagonists & inhibitors , Animals , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Benzophenones/chemistry , Benzophenones/pharmacology , Binding Sites , CHO Cells , Cell Membrane/metabolism , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Devazepide/chemistry , Devazepide/pharmacology , In Vitro Techniques , Ligands , Models, Molecular , Pancreas/cytology , Pancreas/drug effects , Pancreas/metabolism , Photoaffinity Labels/chemistry , Photoaffinity Labels/pharmacology , Protein Structure, Tertiary , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/metabolism , Stereoisomerism , Structure-Activity Relationship
10.
Mol Pharmacol ; 67(6): 1892-900, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15774770

ABSTRACT

Understanding the structures of active and inactive agonist- and antagonist-bound receptor complexes is of great interest. In this work, we focus on position 30 of cholecystokinin (CCK) and its spatial approximation with the type A CCK receptor. For this, we developed two photoaffinity labeling probes, replacing the naturally occurring tryptophan with p-benzoyl-l-phenylalanine (Bpa) or p-nitro-phenylalanine (NO(2)-Phe). The Bpa probe was shown to represent an antagonist, whereas the NO(2)-Phe probe stimulated intracellular calcium as a fully efficacious agonist (EC(50) = 81 +/- 15 nM). Both ligands bound to the receptor specifically, although with lower affinity than CCK (K(i) values: Bpa probe, 270 +/- 72 nM; NO(2)-Phe probe, 180 +/- 40 nM). Both probes covalently labeled the receptor in an efficient manner. The Bpa antagonist labeled the receptor in two distinct regions as identified by cyanogen bromide cleavage, with labeled bands migrating at M(r) = 25,000 and 4500. The former represented the glycosylated amino-terminal fragment, with the site of attachment further localized by endoproteinase Lys-C cleavage to the region between Asn(10) and Lys(37). The latter was shown to represent the first extracellular loop using further cleavage and sequencing of the wild-type and a mutant receptor. Following the same approach, the NO(2)-Phe agonist probe was shown to also label the first extracellular loop region. Radiochemical sequencing identified that the Bpa antagonist probe labeled receptor residue Lys(105), whereas the NO(2)-Phe agonist probe labeled residue Leu(99). These data extend our understanding of the molecular basis of binding and the conformational states of this important receptor.


Subject(s)
Receptor, Cholecystokinin A/chemistry , Receptor, Cholecystokinin A/genetics , Tetragastrin/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Receptor, Cholecystokinin A/metabolism , Tetragastrin/metabolism
11.
Virology ; 329(2): 217-25, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15518802

ABSTRACT

Receptor specificity is a critical determinant of viral tropism, but the capacity of viruses to switch to alternative receptors has not been extensively studied. Here, we engineered the attachment protein of an attenuated measles virus and generated truly retargeted viruses that are blind to the native receptors CD46 and SLAM, but which propagate efficiently and exclusively via alternative cellular receptors, epidermal growth factor receptor, or CD38. The engineered receptor tropisms were stably maintained during multiple serial virus passage without reversion to native receptor usage, even on cells offering the choice of both native and alternative receptors. We conclude that paramyxoviruses have a remarkably flexible and adaptable entry mechanism.


Subject(s)
Measles virus/physiology , Receptors, Virus/metabolism , Recombination, Genetic , Viral Proteins/physiology , ADP-ribosyl Cyclase/metabolism , ADP-ribosyl Cyclase 1 , Animals , Antigens, CD/metabolism , CHO Cells , Chlorocebus aethiops , Cricetinae , ErbB Receptors/metabolism , Glycoproteins/metabolism , Immunoglobulins/metabolism , Measles virus/genetics , Membrane Cofactor Protein , Membrane Glycoproteins/metabolism , Receptors, Cell Surface , Signaling Lymphocytic Activation Molecule Family Member 1 , Tropism , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Recept Channels ; 9(2): 83-91, 2003.
Article in English | MEDLINE | ID: mdl-12916469

ABSTRACT

Cysteine residues play a unique role in structural analysis. We examined endogenous cysteine residues in the cholecystokinin receptor to determine participation in disulfide bonds and accessibility to methanethiosulfonate (MTS) reagents. Bonds linking Cys114 to Cys196 and Cys18 to Cys29 were demonstrated, with the first functionally important and the amino-terminal bond having no apparent function. Cys94, in the second transmembrane segment, was also accessible. Mutation of this residue to serine (C94S) was key for establishing a null cysteine-reactive pseudo-wild type receptor that could act as a template for insertion of a reactive cysteine (N102C, A204C, and T341C). Modification of T341C with a negatively charged MTS reagent reduced CCK agonist binding, while this binding was enhanced by a positively charged MTS reagent. This pattern was repeated in mutants having the same residue directly replaced with a charged residue.


Subject(s)
Cysteine/chemistry , Disulfides/chemistry , Receptors, Cholecystokinin/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Cysteine/metabolism , Disulfides/metabolism , Mesylates/pharmacology , Mutation , Receptors, Cholecystokinin/metabolism , Serine/chemistry , Serine/metabolism , Sulfhydryl Reagents/pharmacology
13.
Mol Pharmacol ; 63(5): 993-1001, 2003 May.
Article in English | MEDLINE | ID: mdl-12695527

ABSTRACT

The amino-terminal domain of class B G protein-coupled receptors is critically important for natural peptide agonist binding and action. The precise role it plays and the molecular basis of the interaction between ligand and this domain are not well understood. In the current work, we have developed a new probe for affinity labeling the secretin receptor through a photolabile benzoyl-phenylalanine residue in position 13. This represented a high affinity ligand (K(i) = 56 +/- 8 nM) that was a potent full agonist to stimulate cellular cAMP (EC(50) = 236 +/- 22 pM). It covalently labeled the secretin receptor saturably in a single site. This was localized to the amino-terminal domain near the first transmembrane segment using a series of chemical and enzymatic digestions. Edman degradation sequencing of radiolabeled cyanogen bromide and skatole digestion products that were attached to glass beads and further cleaved with endoproteinase Asp-N demonstrated that the labeled residue represented Val(103). This is in contrast with previous photoaffinity labeling through positions 6, 18, 22, and 26 of secretin that all labeled the distal end of the amino terminus of this receptor. Together, these five pairs of residue-residue approximations provide important constraints to better understand the molecular conformation of the agonist-bound receptor.


Subject(s)
Receptors, Gastrointestinal Hormone/metabolism , Secretin/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Models, Molecular , Molecular Sequence Data , Photoaffinity Labels , Precipitin Tests , Protein Structure, Tertiary , Rats , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...