Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Med Chem ; 62(4): 1989-1998, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30707835

ABSTRACT

Circadian rhythms are controlled by transcriptional feedback loops of clock genes and proteins. The stability of clock proteins is regulated by post-translational modification, such as phosphorylation by kinases. In particular, casein kinase I (CKI) phosphorylates the PER protein to regulate proteasomal degradation and nuclear localization. Therefore, CKI inhibition can modulate mammalian circadian rhythms. In the present study, we have developed novel CKIα and CKIδ dual inhibitors by extensive structural modification of N9 and C2 position of longdaysin. We identified NCC007 that showed stronger period effects (0.32 µM for 5 h period lengthening) in a cell-based circadian assay. The following in vitro kinase assay showed that NCC007 inhibited CKIα and CKIδ with an IC50 of 1.8 and 3.6 µM. We further demonstrated that NCC007 lengthened the period of mouse behavioral rhythms in vivo. Thus, NCC007 is a valuable tool compound to control circadian rhythms through CKI inhibition.


Subject(s)
Adenine/analogs & derivatives , Casein Kinase Ialpha/antagonists & inhibitors , Circadian Rhythm/drug effects , Enzyme Inhibitors/pharmacology , Adenine/chemical synthesis , Adenine/metabolism , Adenine/pharmacology , Animals , Binding Sites , Casein Kinase Ialpha/chemistry , Casein Kinase Ialpha/metabolism , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Humans , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Protein Binding
2.
PLoS One ; 11(3): e0152934, 2016.
Article in English | MEDLINE | ID: mdl-27031502

ABSTRACT

We describe the identification of a novel, tumor-specific missense mutation in the active site of casein kinase 1α (CSNK1A1) using activity-based proteomics. Matched normal and tumor colon samples were analyzed using an ATP acyl phosphate probe in a kinase-targeted LC-MS2 platform. An anomaly in the active-site peptide from CSNK1A1 was observed in a tumor sample that was consistent with an altered catalytic aspartic acid. Expression and analysis of the suspected mutant verified the presence of asparagine in the probe-labeled, active-site peptide for CSNK1A1. Genomic sequencing of the colon tumor samples confirmed the presence of a missense mutation in the catalytic aspartic acid of CSNK1A1 (GAC→AAC). To our knowledge, the D163N mutation in CSNK1A1 is a newly defined mutation to the conserved, catalytic aspartic acid of a protein kinase and the first missense mutation identified using activity-based proteomics. The tumorigenic potential of this mutation remains to be determined.


Subject(s)
Adenocarcinoma/genetics , Casein Kinase Ialpha/genetics , Colonic Neoplasms/genetics , Mutation, Missense , Adenocarcinoma/pathology , Base Sequence , Casein Kinase Ialpha/chemistry , Catalytic Domain , Colon/pathology , Colonic Neoplasms/pathology , HEK293 Cells , Humans , Molecular Sequence Data , Proteomics/methods
3.
Nature ; 532(7597): 127-30, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26909574

ABSTRACT

Thalidomide and its derivatives, lenalidomide and pomalidomide, are immune modulatory drugs (IMiDs) used in the treatment of haematologic malignancies. IMiDs bind CRBN, the substrate receptor of the CUL4-RBX1-DDB1-CRBN (also known as CRL4(CRBN)) E3 ubiquitin ligase, and inhibit ubiquitination of endogenous CRL4(CRBN) substrates. Unexpectedly, IMiDs also repurpose the ligase to target new proteins for degradation. Lenalidomide induces degradation of the lymphoid transcription factors Ikaros and Aiolos (also known as IKZF1 and IKZF3), and casein kinase 1α (CK1α), which contributes to its clinical efficacy in the treatment of multiple myeloma and 5q-deletion associated myelodysplastic syndrome (del(5q) MDS), respectively. How lenalidomide alters the specificity of the ligase to degrade these proteins remains elusive. Here we present the 2.45 Å crystal structure of DDB1-CRBN bound to lenalidomide and CK1α. CRBN and lenalidomide jointly provide the binding interface for a CK1α ß-hairpin-loop located in the kinase N-lobe. We show that CK1α binding to CRL4(CRBN) is strictly dependent on the presence of an IMiD. Binding of IKZF1 to CRBN similarly requires the compound and both, IKZF1 and CK1α, use a related binding mode. Our study provides a mechanistic explanation for the selective efficacy of lenalidomide in del(5q) MDS therapy. We anticipate that high-affinity protein-protein interactions induced by small molecules will provide opportunities for drug development, particularly for targeted protein degradation.


Subject(s)
Casein Kinase Ialpha/metabolism , Thalidomide/analogs & derivatives , Ubiquitin-Protein Ligases/metabolism , Binding Sites/drug effects , Casein Kinase Ialpha/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Ikaros Transcription Factor/chemistry , Ikaros Transcription Factor/metabolism , Lenalidomide , Models, Molecular , Protein Binding/drug effects , Proteolysis/drug effects , Structure-Activity Relationship , Substrate Specificity/drug effects , Thalidomide/chemistry , Thalidomide/metabolism , Thalidomide/pharmacology , Ubiquitin-Protein Ligases/chemistry , Ubiquitination/drug effects
4.
BMC Struct Biol ; 12: 28, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23148689

ABSTRACT

BACKGROUND: Interaction of non-structural protein 5A (NS5A) of Hepatitis C virus (HCV) with human kinases namely, casein kinase 1α (ck1α) and protein kinase R (PKR) have different functional implications such as regulation of viral replication and evasion of interferon induced immune response respectively. Understanding the structural and molecular basis of interactions of the viral protein with two different human kinases can be useful in developing strategies for treatment against HCV. RESULTS: Serine 232 of NS5A is known to be phosphorylated by human ck1α. A structural model of NS5A peptide containing phosphoacceptor residue Serine 232 bound to ck1α has been generated using the known 3-D structures of kinase-peptide complexes. The substrate interacting residues in ck1α has been identified from the model and these are found to be conserved well in the ck1 family. ck1α - substrate peptide complex has also been used to understand the structural basis of association between ck1α and its other viral stress induced substrate, tumour suppressor p53 transactivation domain which has a crystal structure available.Interaction of NS5A with another human kinase PKR is primarily genotype specific. NS5A from genotype 1b has been shown to interact and inhibit PKR whereas NS5A from genotype 2a/3a are unable to bind and inhibit PKR efficiently. This is one of the main reasons for the varied response to interferon therapy in HCV patients across different genotypes. Using PKR crystal structure, sequence alignment and evolutionary trace analysis some of the critical residues responsible for the interaction of NS5A 1b with PKR have been identified. CONCLUSIONS: The substrate interacting residues in ck1α have been identified using the structural model of kinase - substrate peptide. The PKR interacting NS5A 1b residues have also been predicted using PKR crystal structure, NS5A sequence analysis along with known experimental results. Functional significance and nature of interaction of interferon sensitivity determining region and variable region 3 of NS5A in different genotypes with PKR which was experimentally shown are also supported by the findings of evolutionary trace analysis. Designing inhibitors to prevent this interaction could enable the HCV genotype 1 infected patients respond well to interferon therapy.


Subject(s)
Casein Kinase Ialpha/chemistry , Casein Kinase Ialpha/metabolism , Hepacivirus/chemistry , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , eIF-2 Kinase/chemistry , eIF-2 Kinase/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Evolution, Molecular , Genotype , Hepacivirus/genetics , Humans , Models, Molecular , Molecular Sequence Data , Peptides/metabolism , Phosphoserine/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Sequence Alignment , Structural Homology, Protein , Substrate Specificity , Transcriptional Activation , Tumor Suppressor Protein p53/chemistry
5.
Cell Physiol Biochem ; 30(2): 407-17, 2012.
Article in English | MEDLINE | ID: mdl-22814238

ABSTRACT

Pharmacological modification of protein kinase CK1 (casein kinase 1) has previously been shown to influence suicidal erythrocyte death or eryptosis, which is triggered by activation of Cl(-)-sensitive Ca(2+)-permeable cation channels. Ca(2+) entering through those channels stimulates cell membrane scrambling and opens Ca(2+)-activated K(+)-channels resulting in KCl exit and thus cell shrinkage. The specific CK1-inhibitor D4476 (1 µM) blunted, whereas the specific CK1 αactivator pyrvinium pamoate (10 µM) enhanced cell membrane scrambling. The substances were at least partially effective through modification of cytosolic Ca(2+)-activity. The present study explored, whether pyrvinium pamoate indeed influences Cl(-)-sensitive cation-channels in erythrocytes. As a result, removal of Cl(-)increased Fluo3-fluorescence (reflecting cytosolic Ca(2+)-activity), triggered cell membrane scrambling (apparent from annexin-V-binding), and decreased forward scatter (pointing to cell shrinkage). Pyrvinium pamoate significantly augmented the effect of Cl(-)-removal on Fluo3 fluorescence and annexin-V-binding, but blunted the effect on forward scatter. According to whole cell patch clamp recording, Cl(-)removal activated a cation current, which was significantly enhanced by pyrvinium pamoate. Pyrvinium pamoate inhibited Ca(2+)-activated K(+)-channels. Ca(2+)-ionophore ionomycin (1 µM) decreased forward scatter, an effect significantly blunted by pyrvinium pamoate. In conclusion, pyrvinium pamoate activates Cl(-)-sensitive Ca(2+)-permeable cation channels with subsequent Ca(2+)-entry and inhibits Ca(2+)-activated K(+)-channels thus blunting the stimulating effect of Ca(2+) on those channels, K(+)-exit and thus cell shrinkage.


Subject(s)
Casein Kinase Ialpha/metabolism , Erythrocytes/drug effects , Ion Channels/metabolism , Pyrvinium Compounds/pharmacology , Aniline Compounds/chemistry , Annexin A5/metabolism , Calcium/metabolism , Casein Kinase Ialpha/chemistry , Cations/metabolism , Cell Membrane/drug effects , Cell Size/drug effects , Electrophysiological Phenomena/drug effects , Erythrocytes/metabolism , Humans , Ionomycin/pharmacology , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels, Calcium-Activated/metabolism , Protein Binding , Xanthenes/chemistry
6.
J Biomol Struct Dyn ; 29(5): 843-61, 2012.
Article in English | MEDLINE | ID: mdl-22292947

ABSTRACT

The canonical Wnt-pathway plays a number of crucial roles in the development of organism. Malfunctions of this pathway lead to various diseases including cancer. In the inactivated state, this pathway involves five proteins, Axin, CKI-α, GSK-3ß, APC, and ß-catenin. We analyzed these proteins by a number of computational tools, such as PONDR(r)VLXT, PONDR(r)VSL2, MoRF-II predictor and Hydrophobic Cluster Analysis (HCA) to show that each of the Wnt-pathway proteins contains several intrinsically disordered regions. Based on a comprehensive analysis of published data we conclude that these disordered regions facilitate protein-protein interactions, post-translational modifications, and signaling. The scaffold protein Axin and another large protein, APC, act as flexible concentrators in gathering together all other proteins involved in the Wnt-pathway, emphasizing the role of intrinsically disordered regions in orchestrating the complex protein-protein interactions. We further explore the intricate roles of highly disordered APC in regulation of ß-catenin function. Intrinsically disordered APC helps the collection of ß-catenin from cytoplasm, facilitates the b-catenin delivery to the binding sites on Axin, and controls the final detachment of ß-catenin from Axin.


Subject(s)
Adenomatous Polyposis Coli Protein/chemistry , Axin Protein/chemistry , Casein Kinase Ialpha/chemistry , Glycogen Synthase Kinase 3/chemistry , Wnt Signaling Pathway , beta Catenin/chemistry , Adenomatous Polyposis Coli Protein/metabolism , Amino Acid Sequence , Axin Protein/metabolism , Binding Sites , Casein Kinase Ialpha/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Proline , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , beta Catenin/metabolism
7.
Anal Biochem ; 405(2): 246-54, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20561505

ABSTRACT

Glycogen synthase kinase-3beta (GSK3beta) and casein kinase-1alpha (CK1alpha) are multifunctional kinases that play critical roles in the regulation of a number of cellular processes. In spite of their importance, molecular imaging tools for noninvasive and real-time monitoring of their kinase activities have not been devised. Here we report development of the bioluminescent GSK3beta and CK1alpha reporter (BGCR) based on firefly luciferase complementation. Treatment of SW620 cells stably expressing the reporter with inhibitors of GSK3beta (SB415286 and LiCl) or CK1alpha (CKI-7) resulted in dose- and time-dependent increases in BGCR activity that were validated using Western blotting. No increase in bioluminescence was observed in the case of S37A mutant (GSK3beta inhibitors) or S45A mutant (CKI-7), demonstrating the specificity of the reporter. Imaging of mice tumor xenograft generated with BGCR-expressing SW620 cells following treatment with LiCl showed unique oscillations in GSK3beta activity that were corroborated by phosphorylated GSK3beta immunoblotting. Taken together, the BGCR is a novel molecular imaging tool that reveals unique insight into GSK3beta and CK1alpha kinase activities and may provide a powerful tool in experimental therapeutics for rapid optimization of dose and schedule of targeted therapies and for monitoring therapeutic response.


Subject(s)
Casein Kinase Ialpha/chemistry , Glycogen Synthase Kinase 3/chemistry , Molecular Imaging/methods , Animals , Casein Kinase Ialpha/metabolism , Cells, Cultured , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Male , Mice , Transplantation, Heterologous , Wnt Proteins/chemistry , Wnt Proteins/metabolism , Wnt3 Protein
8.
Cell Mol Life Sci ; 67(7): 1105-18, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20041275

ABSTRACT

The ability of three isoforms of protein kinase CK1 (alpha, gamma(1), and delta) to phosphorylate the N-terminal region of p53 has been assessed using either recombinant p53 or a synthetic peptide reproducing its 1-28 sequence. Both substrates are readily phosphoylated by CK1delta and CK1alpha, but not by the gamma isoform. Affinity of full size p53 for CK1 is 3 orders of magnitude higher than that of its N-terminal peptide (K (m) 0.82 muM vs 1.51 mM). The preferred target is S20, whose phosphorylation critically relies on E17, while S6 is unaffected despite displaying the same consensus (E-x-x-S). Our data support the concept that non-primed phosphorylation of p53 by CK1 is an isoform-specific reaction preferentially affecting S20 by a mechanism which is grounded both on a local consensus and on a remote docking site mapped to the K(221)RQK(224) loop according to modeling and mutational analysis.


Subject(s)
Casein Kinase I/metabolism , Casein Kinase Ialpha/metabolism , Casein Kinase Idelta/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Binding Sites , Casein Kinase I/chemistry , Casein Kinase Ialpha/chemistry , Casein Kinase Idelta/chemistry , Computer Simulation , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Molecular Sequence Data , Mutation , Phosphorylation , Tumor Suppressor Protein p53/chemistry , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
9.
J Cell Biochem ; 106(3): 399-408, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19115251

ABSTRACT

CK1 constitutes a protein kinase subfamily that is involved in many important physiological processes. However, there is limited knowledge about mechanisms that regulate their activity. Isoforms CK1delta and CK1epsilon were previously shown to autophosphorylate carboxy-terminal sites, a process which effectively inhibits their catalytic activity. Mass spectrometry of CK1alpha and splice variant CK1alphaL has identified the autophosphorylation of the last four carboxyl-end serines and threonines and also for CK1alphaS, the same four residues plus threonine-327 and serine-332 of the S insert. Autophosphorylation occurs while the recombinant proteins are expressed in Escherichia coli. Mutation of four carboxy-terminal phosphorylation sites of CK1alpha to alanine demonstrates that these residues are the principal but not unique sites of autophosphorylation. Treatment of autophosphorylated CK1alpha and CK1alphaS with lambda phosphatase causes an activation of 80-100% and 300%, respectively. Similar treatment fails to stimulate the CK1alpha mutants lacking autophosphorylation sites. Incubation of dephosphorylated enzymes with ATP to allow renewed autophosphorylation causes significant inhibition of CK1alpha and CK1alphaS. The substrate for these studies was a synthetic canonical peptide for CK1 (RRKDLHDDEEDEAMS*ITA). The stimulation of activity seen upon dephosphorylation of CK1alpha and CK1alphaS was also observed using the known CK1 protein substrates DARPP-32, beta-catenin, and CK2beta, which have different CK1 recognition sequences. Autophosphorylation effects on CK1alpha activity are not due to changes in Km(app) for ATP or for peptide substrate but rather to the catalytic efficiency per pmol of enzyme. This work demonstrates that CK1alpha and its splice variants can be regulated by their autophosphorylation status.


Subject(s)
Casein Kinase Ialpha/metabolism , Amino Acid Sequence , Animals , Biocatalysis , Casein Kinase Ialpha/chemistry , Casein Kinase Ialpha/genetics , Enzyme Activation , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Molecular Sequence Data , Mutation/genetics , Phosphorylation , Protein Subunits/genetics , Protein Subunits/metabolism , Substrate Specificity , Time Factors , Zebrafish/genetics , Zebrafish/metabolism
10.
J Biol Chem ; 282(8): 5536-44, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17166835

ABSTRACT

The hepatitis C virus encodes a single polyprotein that is processed by host and viral proteases to yield at least 10 mature viral proteins. The nonstructural (NS) protein 5A is a phosphoprotein, and experimental data indicate that the phosphorylation state of NS5A is important for the outcome of viral RNA replication. We were able to identify kinase inhibitors that specifically inhibit the formation of the hyperphosphorylated form of NS5A (p58) in cells. These kinase inhibitors were used for inhibitor affinity chromatography in order to identify the cellular targets of these compounds. The kinases casein kinase I (CKI), p38 MAPK, CIT (Citron Rho-interacting kinase), GAK, JNK2, PKA, RSK1/2, and RIPK2 were identified in the high affinity binding fractions of two NS5A hyperphosphorylation inhibitors (NS5A-p58-i). Even though these kinases are targets of the NS5A-p58-i, the only kinase showing an effect on NS5A hyperphosphorylation was confirmed to be CKI-alpha. Although this finding does not exclude the possibility that other kinase(s) might be involved in basal or regulatory phosphorylation of NS5A, we show here that NS5A is a direct substrate of CKI-alpha. Moreover, in vitro phosphorylation of NS5A by CKI-alpha resulted for the first time in the production of basal and hyperphosphorylated forms resembling those produced in cells. In vitro kinase reactions performed with NS5A peptides show that Ser-2204 is a preferred substrate residue for CKI-alpha after pre-phosphorylation of Ser-2201.


Subject(s)
Casein Kinase Ialpha/metabolism , Hepacivirus/metabolism , Protein Processing, Post-Translational/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , Animals , Casein Kinase Ialpha/chemistry , Casein Kinase Ialpha/isolation & purification , Cell Line , Chromatography, Affinity , Hepacivirus/chemistry , Humans , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/isolation & purification , Virus Replication/drug effects
11.
Dev Biol ; 299(1): 221-37, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16987508

ABSTRACT

The Wingless (Wg)/Wnt signaling pathway regulates a myriad of developmental processes and its malfunction leads to human disorders including cancer. Recent studies suggest that casein kinase I (CKI) family members play pivotal roles in the Wg/Wnt pathway. However, genetic evidence for the involvement of CKI family members in physiological Wg/Wnt signaling events is lacking. In addition, there are conflicting reports regarding whether a given CKI family member functions as a positive or negative regulator of the pathway. Here we examine the roles of seven CKI family members in Wg signaling during Drosophila limb development. We find that increased CKIepsilon stimulates whereas dominant-negative or a null CKIepsilon mutation inhibits Wg signaling. In contrast, inactivation of CKIalpha by RNA interference (RNAi) leads to ectopic Wg signaling. Interestingly, hypomorphic CKIepsilon mutations synergize with CKIalpha RNAi to induce ectopic Wg signaling, revealing a negative role for CKIepsilon. Conversely, CKIalpha RNAi enhances the loss-of-Wg phenotypes caused by CKIepsilon null mutation, suggesting a positive role for CKIalpha. While none of the other five CKI isoforms can substitute for CKIalpha in its inhibitory role in the Wg pathway, several CKI isoforms including CG12147 exhibit a positive role based on overexpression. Moreover, loss of Gilgamesh (Gish)/CKIgamma attenuates Wg signaling activity. Finally, we provide evidence that several CKI isoforms including CKIalpha and Gish/CKIgamma can phosphorylate the Wg coreceptor Arrow (Arr), which may account, at least in part, for their positive roles in the Wg pathway.


Subject(s)
Casein Kinase I/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/metabolism , Extremities/embryology , Proto-Oncogene Proteins/metabolism , Signal Transduction , Animals , Casein Kinase 1 epsilon/chemistry , Casein Kinase 1 epsilon/deficiency , Casein Kinase 1 epsilon/metabolism , Casein Kinase I/chemistry , Casein Kinase Ialpha/chemistry , Casein Kinase Ialpha/metabolism , Genes, Dominant/genetics , Isoenzymes/chemistry , Isoenzymes/metabolism , Mutation/genetics , Phosphorylation , RNA Interference , Receptors, Cell Surface/metabolism , Wings, Animal/cytology , Wnt1 Protein , Xenopus
12.
Biomacromolecules ; 6(6): 3018-29, 2005.
Article in English | MEDLINE | ID: mdl-16283722

ABSTRACT

We present a statistical mechanical approach to predicting the properties of mixed copolymer layers using the Scheutjens-Fleer self-consistent-field theory. Our model copolymers are based on the primary structures of the major bovine casein monomers, alpha(s1)-casein and beta-casein. Numerical calculations have been carried out to determine the polymer segment density profiles at an isolated hydrophobic surface and the interaction forces as a pair of polymer-coated surfaces is brought to close interlayer separation. For a copolymer model containing hydrophilic and hydrophobic segments, we show how the steric stabilizing capacity of a casein-like macromolecule at very low surface coverage is enhanced in the presence of a thin dense layer of shorter tethered amphiphilic chains. Using a more refined protein model, which also distinguishes between the charged and uncharged hydrophilic segments along the chain, we clearly demonstrate that the enhanced steric repulsion from beta-casein exceeds that from alpha(s1)-casein. These calculations explain how the replacement of just a few percent of beta-lactoglobulin by casein can inhibit the heat-induced thickening and flocculation behavior observed experimentally with some whey protein-stabilized oil-in-water emulsions.


Subject(s)
Biocompatible Materials/chemistry , Caseins/chemistry , Macromolecular Substances/chemistry , Polymers/chemistry , Adsorption , Animals , Casein Kinase Ialpha/chemistry , Lactoglobulins/chemistry , Milk Proteins/chemistry , Models, Statistical , Models, Theoretical , Oils/chemistry , Static Electricity , Stereoisomerism , Thermodynamics , Time Factors , Water/chemistry , Whey Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...