E. Application, F. Ferrage, S. Cousin, C. Charlier, T. Marquardsen et al., Method for two--eld nuclear magnetic resonance measurement

@. Cyril, C. , S. F. Cousin, and F. Ferrage, Protein Dynamics from Nuclear Magnetic Relaxation, Chem. Soc. Rev, pp.10-1039, 2016.

@. Samuel, F. Cousin, P. Kade?ávek, B. Haddou, C. Charlier et al., Recovering Invisible Signals by Two-Field NMR Spectroscopy, Angewandte Chemie International Edition

D. Kurzbach, . Weber, S. F. Jhajharia, A. Cousin, S. Sadet et al., Side-EEects of Cross-Polarization Enhanced Dissolution DNP of Deuterated Molecules

S. F. Cousin, C. Charlier, P. Kade?ávek, T. Marquardsen, J. Tyburn et al., Nuclear Magnetic Resonance Spectroscopy
URL : https://hal.archives-ouvertes.fr/cea-01393871

S. F. Cousin, P. Kade?ávek, N. Bolik-coulon, and F. Ferrage, Study of Protein Motions by High-Resolution Relaxometry Book chapter invited contribution, work in progress ? "Full broadband carbon-13 correlations with two--eld NMR spectroscopy

R. Jain, D. T. Nair, R. E. Johnson, L. Prakash, S. Prakash et al., Replication across Template T/U by Human DNA Polymerase-??, Structure, vol.17, issue.7, pp.974-980
DOI : 10.1016/j.str.2009.04.011

URL : http://doi.org/10.1016/j.str.2009.04.011

V. Motá?ková, H. ?anderová, L. ?ídek, J. Nová?ek, P. Padrta et al., Solution structure of the N-terminal domain of Bacillus subtilis ? subunit of RNA polymerase and its classiication based on structural homologs, Proteins: Structure, Function, and Bioinformatics, p.NA?NA, 2010.

E. Callaway, The revolution will not be crystallized: a new method sweeps through structural biology, Nature, vol.525, issue.7568, pp.172-174
DOI : 10.1038/525172a

K. K. Frederick, M. S. Marlow, K. G. Valentine, and A. Wand, Conformational entropy in molecular recognition by proteins, Nature, vol.10, issue.7151, pp.325-329
DOI : 10.1038/nature05959

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4156320

I. Gelis, A. M. Bonvin, D. Keramisanou, M. Koukaki, G. Gouridis et al., Structural Basis for Signal-Sequence Recognition by the Translocase Motor SecA as Determined by NMR, Cell, vol.131, issue.4, pp.756-769
DOI : 10.1016/j.cell.2007.09.039

R. Ernst, G. Bodenhausen, and A. Wokaun, Principles of Nuclear Magnetic Resonance in One and Two Dimensions. International Series of Monographs on Chemistry, 1990.

M. Liu, W. Leskes, A. J. Yu, L. Moore, P. M. Zhou et al., Cycling Li-O2 batteries via LiOH formation and decomposition, Science, vol.117, issue.32, pp.350530-533
DOI : 10.1149/1.3507922

URL : https://www.repository.cam.ac.uk/bitstream/1810/253469/1/Liu-et-al%202015%20Science.pdf

J. Nelson, D. Vigneron, J. Kurhanewicz, A. Chen, R. Bok et al., DNP-Hyperpolarized 13C Magnetic Resonance Metabolic Imaging for Cancer Applications, Applied Magnetic Resonance, vol.56, issue.3-4, pp.3-4533
DOI : 10.1007/s00723-008-0136-2

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2829774

J. Weekley, P. Bruins, M. Sisto, and M. P. Augustine, Using NMR to study full intact wine bottles, Journal of Magnetic Resonance, vol.161, issue.1, pp.91-98
DOI : 10.1016/S1090-7807(02)00177-5

URL : http://chemgroups.ucdavis.edu/~augustine/papers/2002_Using NMR to study full intact wine bottles.pdf

C. Lauterbur, Image Formation by Induced Local Interactions: Examples Employing Nuclear Magnetic Resonance, Nature, vol.178, issue.5394, pp.190-191
DOI : 10.1038/242190a0

K. Wüthrich, The way to NMR structures of proteins, Nature, vol.8, issue.11, pp.923-925, 2001.

C. Charlier, S. F. Cousin, and F. Ferrage, Protein dynamics from nuclear magnetic relaxation, Chem. Soc. Rev., vol.104, issue.9, 2016.
DOI : 10.1039/C5CS00832H

URL : https://hal.archives-ouvertes.fr/hal-01366187

A. G. Reddeld, High-resolution NMR field-cycling device for full-range relaxation and structural studies of biopolymers on a shared commercial instrument, Journal of Biomolecular NMR, vol.50, issue.2, pp.159-177
DOI : 10.1007/s10858-011-9594-1

C. Charlier, S. Nawaz-khan, T. Marquardsen, P. Pelupessy, V. Reiss et al., Nanosecond Time Scale Motions in Proteins Revealed by High-Resolution NMR Relaxometry, Journal of the American Chemical Society, vol.135, issue.49, pp.18665-18672
DOI : 10.1021/ja409820g

URL : http://doi.org/10.1021/ja409820g

N. Tjandra, S. E. Feller, R. W. Pastor, and A. Bax, Rotational diffusion anisotropy of human ubiquitin from 15N NMR relaxation, Journal of the American Chemical Society, vol.117, issue.50, pp.12562-12566
DOI : 10.1021/ja00155a020

F. Lienin, T. Bremi, B. Brutscher, R. Brüschweiler, and R. R. Ernst, Anisotropic Intramolecular Backbone Dynamics of Ubiquitin Characterized by NMR Relaxation and MD Computer Simulation, Journal of the American Chemical Society, vol.120, issue.38, pp.9870-9879, 1998.
DOI : 10.1021/ja9810179

A. Nils, T. Lakomek, S. Carlomagno, C. Becker, J. Griesinger et al., A Thorough Dynamic Interpretation of Residual Dipolar Couplings in Ubiquitin, Journal of Biomolecular NMR, vol.34, issue.2, pp.101-115

A. Abragam, The Principles of nuclear magnetism, 1961.

W. F. Mark, A. Fischer, E. R. Majumdar, and . Zuiderweg, Protein NMR relaxation: theory, applications and outlook, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.33, pp.3-4207

M. Korzhnev, M. Billeter, A. S. Arseniev, and V. Y. Orekhov, NMR studies of Brownian tumbling and internal motions in proteins, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.38, issue.3, pp.197-266
DOI : 10.1016/S0079-6565(00)00028-5

H. Malcolm and . Levitt, Spin Dynamica, 2015.

I. Kuprov, N. Wagner-rundell, and P. J. Hore, Bloch-Redfield-Wangsness theory engine implementation using symbolic processing software, Journal of Magnetic Resonance, vol.184, issue.2, pp.196-206
DOI : 10.1016/j.jmr.2006.09.023

URL : http://citeseerx.ist.psu.edu/viewdoc/summary?doi=10.1.1.573.5968

K. Berlin, A. Longhini, T. Kwaku-dayie, and D. Fushman, Deriving quantitative dynamics information for proteins and RNAs using ROTDIF with a graphical user interface, Journal of Biomolecular NMR, vol.128, issue.35, pp.333-352
DOI : 10.1007/s10858-013-9791-1

J. Frank, R. Millero, E. Dexter, and . Hoo, Density and viscosity of deuterium oxide solutions from 5-70. deg, Journal of Chemical & Engineering Data, vol.16, issue.1, pp.85-87, 1971.

X. Liao, D. Long, D. Li, R. Brüschweiler, and V. Tugarinov, Probing Side-Chain Dynamics in Proteins by the Measurement of Nine Deuterium Relaxation Rates Per Methyl Group, The Journal of Physical Chemistry B, vol.116, issue.1, pp.606-620
DOI : 10.1021/jp209304c

H. Haario, M. Laine, A. Mira, and E. Saksman, DRAM: Efficient adaptive MCMC, Statistics and Computing, vol.18, issue.2, pp.339-354
DOI : 10.1007/s11222-006-9438-0

URL : http://citeseerx.ist.psu.edu/viewdoc/summary?doi=10.1.1.467.9433

V. Tugarinov, R. Muhandiram, A. Ayed, and L. E. Kay, Four-Dimensional NMR Spectroscopy of a 723-Residue Protein:?? Chemical Shift Assignments and Secondary Structure of Malate Synthase G, Journal of the American Chemical Society, vol.124, issue.34, pp.10025-10035
DOI : 10.1021/ja0205636

L. Braunschweiler and R. R. Ernst, Coherence transfer by isotropic mixing: Application to proton correlation spectroscopy, Journal of Magnetic Resonance (1969), vol.53, issue.3, pp.521-528, 1969.
DOI : 10.1016/0022-2364(83)90226-3

K. Gheysen, C. Mihai, K. Conrath, and J. C. Martins, Rapid Identification of Common Hexapyranose Monosaccharide Units by a Simple TOCSY Matching Approach, Chemistry - A European Journal, vol.230, issue.29, pp.8869-8878
DOI : 10.1002/chem.200801081

C. John and R. Mark, Sensitivity Improvement in Isotropic Mixing (TOCSY) Experiments, Journal of Magnetic Resonance, vol.88, pp.72-85, 1990.

L. D. Hall and T. J. Norwood, Use of zero-quantum coherences to measure high resolution n.m.r. spectra in inhomogeneous magnetic elds, Journal of the Chemical Society, Chemical Communications, issue.1, p.44, 1986.

H. Akhtar, R. S. Mcdaniel, A. C. Feser, and . Oehlschlager, NMR study of hindered rotation in 1-aryl-3,3-dimethyltriazenes, Tetrahedron, vol.24, issue.10, pp.3899-3906
DOI : 10.1016/S0040-4020(01)92598-0

A. Arora, F. Abildgaard, J. H. Bushweller, and L. K. Tamm, Structure of outer membrane protein A transmembrane domain by NMR spectroscopy, Nature Structural Biology, vol.8, issue.4, pp.334-338, 2001.
DOI : 10.1038/86214

K. Wilkinson and A. Hershko, Irwin Allan Rose (1926???2015), Nature, vol.523, issue.7562, pp.532-532, 1926.
DOI : 10.1038/523532a

Z. Elan, O. Eisenmesser, W. Millet, D. M. Labeikovsky, M. Korzhnev et al., Intrinsic dynamics of an enzyme underlies catalysis, Nature, issue.7064, pp.438117-121

U. Lemieux and U. Spohr, How Emil Fischer was Led to the Lock and Key Concept for Enzyme Specificity11Presented at the symposium ???Emil Fischer: 100 Years of Carbohydrate Chemistry,??? 203rd National Meeting of the American Chemical Society, Division of Carbohydrate Chemistry, San Francisco, California, April 5???10, 1992., Adv Carbohydr Chem Biochem, vol.50, pp.1-20, 1994.
DOI : 10.1016/S0065-2318(08)60149-3

C. Kendrew, G. Bodo, H. M. Dintzis, R. G. Parrish, H. Wyckoo et al., A Three-Dimensional Model of the Myoglobin Molecule Obtained by X-Ray Analysis, Nature, vol.178, issue.4610, pp.181662-666
DOI : 10.1002/hlca.19490320118

C. Kendrew, R. E. Dickerson, B. E. Strandberg, R. G. Hart, D. R. Davies et al., Structure of Myoglobin: A Three-Dimensional Fourier Synthesis at 2 ???. Resolution, Nature, vol.178, issue.4711, pp.185422-427
DOI : 10.1107/S0365110X5900007X

T. D. Silverstein, R. E. Johnson, R. Jain, L. Prakash, S. Prakash et al., Structural basis for the suppression of skin cancers by DNA polymerase ??, Nature, vol.54, issue.7301, pp.1039-1043
DOI : 10.1038/nature09104

E. Peter, H. J. Wright, and . Dyson, Intrinsically unstructured proteins: re-assessing the protein structure-function paradigm, Journal of molecular biology, vol.293, issue.2, pp.321-331, 1999.

A. J. Dingley, I. Lorenzen, and J. Grötzinger, NMR Analysis of Viral Protein Structures, Plant Virology Protocols, pp.441-462, 2008.
DOI : 10.1007/978-1-59745-102-4_30

M. David, A. T. Ginter, C. J. Bell, and . Radke, Magic-angle-spinning NMR spectroscopy of gels, Journal of Magnetic Resonance, vol.81, issue.1, pp.217-219, 1969.

A. Loquet, N. G. Sgourakis, R. Gupta, K. Giller, D. Riedel et al., Atomic model of the type III secretion system needle, Nature, vol.309, pp.2012-2017
DOI : 10.1038/nature11079

V. Tugarinov, V. Kanelis, E. Lewis, and . Kay, Isotope labeling strategies for the study of high-molecular-weight proteins by solution NMR spectroscopy, Nature Protocols, vol.1, issue.2, pp.749-754
DOI : 10.1007/s10858-005-2614-2

K. Rime, Développement de stratégies de marquage isotopique des groupements méhyles pour l'éude d'assemblages protéques de grande taille par RMN, pp.2014-2023

G. Mas, E. Crublet, O. Hamelin, P. Gans, and J. Boisbouvier, Specific labeling and assignment strategies of valine methyl groups for NMR studies of high molecular weight proteins, Journal of Biomolecular NMR, vol.59, issue.3, pp.251-262
DOI : 10.1007/s10858-013-9785-z

URL : https://hal.archives-ouvertes.fr/hal-01101628

P. Schanda, H. Van-melckebeke, and B. Brutscher, Speeding Up Three-Dimensional Protein NMR Experiments to a Few Minutes, Journal of the American Chemical Society, vol.128, issue.28, pp.9042-9043
DOI : 10.1021/ja062025p

M. Nowakowski, S. Saxena-stanek, S. ?erko, and W. Ko?mi?ski, Applications of high dimensionality experiments to biomolecular NMR, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.90, issue.91, pp.90-9149
DOI : 10.1016/j.pnmrs.2015.07.001

A. Medek, J. S. Harwood, and L. Frydman, Multiple-Quantum Magic-Angle Spinning NMR: A New Method for the Study of Quadrupolar Nuclei in Solids, Journal of the American Chemical Society, vol.117, issue.51, pp.12779-12787, 1995.
DOI : 10.1021/ja00156a015

K. Wüthrich, NMR of proteins and nucleic acids. The George Fisher Baker non-resident lectureship in chemistry at Cornell University, 1986.

J. Fiaux, E. B. Bertelsen, A. L. Horwich, and K. Wuthrich, NMR analysis of a 900K GroEL???GroES complex, Nature, vol.24, issue.6894, pp.207-211
DOI : 10.1038/7573

S. Gil-caballero, A. Favier, and B. Brutscher, HNCA+, HNCO+, and HNCACB+ experiments: improved performance by simultaneous detection of orthogonal coherence transfer pathways, Journal of Biomolecular NMR, vol.239, issue.1, pp.1-9
DOI : 10.1007/s10858-014-9847-x

URL : https://hal.archives-ouvertes.fr/hal-01321280

X. Cavalli, C. M. Salvatella, M. Dobson, and . Vendruscolo, Protein structure determination from NMR chemical shifts, Proceedings of the National Academy of Sciences, pp.9615-9620
DOI : 10.1016/S0022-2836(03)00021-4

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1887584

J. Korukottu, R. Schneider, V. Vijayan, A. Lange, O. Pongs et al., High-Resolution 3D Structure Determination of Kaliotoxin by Solid-State NMR Spectroscopy, PLoS ONE, vol.440, issue.6, pp.2359-2008
DOI : 10.1371/journal.pone.0002359.s003

K. Pervushin, R. Riek, and K. Wüthrich, Attenuated T2 relaxation by mutual cancellation of dipole-dipole coupling and chemical shift anisotropy indicates an avenue to NMR structures of very large biological macromolecules in solution, Proc. Natl. Acad. Sci. USA, pp.12366-12371, 1997.
DOI : 10.1007/BF02192855

V. Tugarinov, R. Sprangers, and L. E. Kay, C NMR Spectroscopy, Journal of the American Chemical Society, vol.126, issue.15, pp.4921-4925
DOI : 10.1021/ja039732s

M. Dmitry, K. Korzhnev, V. Kloiber, V. Kanelis, L. E. Tugarinov et al., Probing Slow Dynamics in High Molecular Weight Proteins by Methyl-TROSY NMR Spectroscopy: Application to a 723-Residue Enzyme, Journal of the American Chemical Society, vol.126, issue.12, pp.3964-3973

G. Roth, Insights into Superconducting Ultra High Field Magnet Technology, pp.2015-2019

G. Lipari and A. Szabo, Model-free approach to the interpretation of nuclear magnetic resonance relaxation in macromolecules. 1. Theory and range of validity, Journal of the American Chemical Society, vol.104, issue.17, pp.4546-4559, 1982.
DOI : 10.1021/ja00381a009

M. Clore, A. Szabo, A. Bax, L. E. Kay, P. C. Driscoll et al., Deviations from the simple two-parameter model-free approach to the interpretation of nitrogen-15 nuclear magnetic relaxation of proteins, Journal of the American Chemical Society, vol.112, issue.12, pp.1124989-4991, 1990.
DOI : 10.1021/ja00168a070

A. M. Mandel, M. Akke, A. G. Palmer, and I. , Backbone Dynamics ofEscherichia coliRibonuclease HI: Correlations with Structure and Function in an Active Enzyme, Journal of Molecular Biology, vol.246, issue.1, pp.144-163
DOI : 10.1006/jmbi.1994.0073

W. Jeerey, G. Peng, and . Wagner, Mapping of Spectral Density Functions Using Heteronuclear NMR Relaxation Measurements, Journal of Magnetic Resonance, vol.98, pp.308-332, 1992.

R. Kimmich and E. Anoardo, Field-cycling NMR relaxometry, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.44, issue.3-4, pp.257-320
DOI : 10.1016/j.pnmrs.2004.03.002

C. Chou, M. Chu, C. Chang, and T. Huang, A compact high-speed mechanical sample shuttle for field-dependent high-resolution solution NMR, Journal of Magnetic Resonance, vol.214, pp.302-308
DOI : 10.1016/j.jmr.2011.12.001

M. W. Clarkson, M. Lei, E. Z. Eisenmesser, W. Labeikovsky, A. Reddeld et al., Mesodynamics in the SARS nucleocapsid measured by NMR field cycling, Journal of Biomolecular NMR, vol.357, issue.1-2, pp.217-225
DOI : 10.1007/s10858-009-9347-6

V. Chevelkov, Y. Xue, R. Linser, N. R. Skrynnikov, and B. Reif, Comparison of Solid-State Dipolar Couplings and Solution Relaxation Data Provides Insight into Protein Backbone Dynamics, Journal of the American Chemical Society, vol.132, issue.14, pp.5015-5017
DOI : 10.1021/ja100645k

F. Ferrage, P. Pelupessy, D. Cowburn, and G. Bodenhausen, Cross-Relaxation in NMR Spectroscopy, Journal of the American Chemical Society, vol.128, issue.34, pp.11072-11078
DOI : 10.1021/ja0600577

URL : https://hal.archives-ouvertes.fr/hal-00103657

R. A. Venters, C. Huang, B. T. Farmer, I. , R. Trolard et al., High-level 2H/13C/15N labeling of proteins for NMR studies, Journal of Biomolecular NMR, vol.5, issue.4, pp.339-344, 1995.
DOI : 10.1007/BF00182275

W. Tugarinov, V. Choy, . Yu, L. E. Orekhov, and . Kay, Solution NMR-derived global fold of a monomeric 82-kDa enzyme, Proceedings of the National Academy of Sciences, vol.8, issue.1, pp.622-627
DOI : 10.1016/0263-7855(96)00009-4

T. Yamazaki, H. Tochio, J. Furui, S. Aimoto, and Y. Kyogoku, N-Labeled Sample, Journal of the American Chemical Society, vol.119, issue.5, pp.872-880
DOI : 10.1021/ja962945f

M. Kainosho, T. Torizawa, Y. Iwashita, T. Terauchi, A. M. Ono et al., Optimal isotope labelling for NMR protein structure determinations, Nature, vol.30, issue.7080, pp.52-57
DOI : 10.1038/nature04525

Y. Miyanoiri, M. Takeda, K. Okuma, A. M. Ono, T. Terauchi et al., Differential isotope-labeling for Leu and Val residues in a protein by E. coli cellular expression using stereo-specifically methyl labeled amino acids, Journal of Biomolecular NMR, vol.335, issue.3, pp.237-249
DOI : 10.1007/s10858-013-9784-0

K. G. Vignesh-kasinath, A. Valentine, and . Wand, C Labeling Strategy Reveals a Range of Aromatic Side Chain Motion in Calmodulin, Journal of the American Chemical Society, vol.135, issue.26, pp.9560-9563
DOI : 10.1021/ja4001129

R. J. Lichtenecker, K. Weinhäupl, W. Schmid, and R. Konrat, ??-Ketoacids as precursors for phenylalanine and tyrosine labelling in cell-based protein overexpression, Journal of Biomolecular NMR, vol.121, issue.4, pp.327-331
DOI : 10.1007/s10858-013-9796-9

P. J. Hajduk, D. J. Augeri, J. Mack, R. Mendoza, J. Yang et al., C-Labeled Methyl Groups, Journal of the American Chemical Society, vol.122, issue.33, pp.7898-7904
DOI : 10.1021/ja000350l

L. Religa, R. Sprangers, and L. E. Kay, Dynamic Regulation of Archaeal Proteasome Gate Opening As Studied by TROSY NMR, Science, vol.27, issue.5, pp.98-102
DOI : 10.1016/j.molcel.2007.06.033

K. Wangness and F. Bloch, The Dynamical Theory of Nuclear Induction, Physical Review, vol.89, issue.4, pp.728-739, 1953.
DOI : 10.1103/PhysRev.89.728

A. G. Reddeld, On the Theory of Relaxation Processes, IBM Journal of Research and Development, vol.1, issue.1, pp.19-31, 1957.
DOI : 10.1147/rd.11.0019

D. Frueh, Internal motions in proteins and interference effects in nuclear magnetic resonance, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.41, issue.3-4, pp.305-324, 2002.
DOI : 10.1016/S0079-6565(02)00051-1

M. Goldman, Formal Theory of Spin???Lattice Relaxation, Journal of Magnetic Resonance, vol.149, issue.2, pp.160-187
DOI : 10.1006/jmre.2000.2239

J. Kowalewski and L. Mäler, Nuclear spin relaxation in liquids: theory, experiments, and applications . Number 2 in Series in chemical physics, 2006.
DOI : 10.1201/9781420012194

URL : https://repozitorij.uni-lj.si/Dokument.php?id=39829

M. P. Nicholas, E. Eryilmaz, F. Ferrage, D. Cowburn, and R. Ghose, Nuclear spin relaxation in isotropic and anisotropic media, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.57, issue.2, pp.111-158
DOI : 10.1016/j.pnmrs.2010.04.003

URL : https://hal.archives-ouvertes.fr/hal-00605407

. Van-de-wiele, Rotations and angular moments in quantum mechanics, Annales de Physique, vol.26, issue.6, pp.1-167, 2001.
DOI : 10.1051/anphys:200106001

URL : https://hal.archives-ouvertes.fr/in2p3-00019832

A. Kumar, R. C. , R. Grace, and P. K. Madhu, Cross-correlations in NMR, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.37, issue.3, p.191, 2000.
DOI : 10.1016/S0079-6565(00)00023-6

M. Goldman, Interference EEects in the Relaxaation of a Pair of Unlike Spin 1/2 Nuclei, Journal of Magnetic Resonance, vol.60, pp.437-452, 1984.

D. 100-fabien-ferrage, R. Cowburn, and . Ghose, Accurate Sampling of High-Frequency Motions in Proteins by Steady-State 15 N-{ 1 H} Nuclear Overhauser EEect Measurements in the Presence of Cross-Correlated Relaxation, Journal of the American Chemical Society, issue.17, pp.1316048-6049

F. Ferrage, A. Reichel, S. Battacharya, D. Cowburn, and R. Ghose, On the measurement of 15N???{1H} nuclear Overhauser effects. 2. Effects of the saturation scheme and water signal suppression, Journal of Magnetic Resonance, vol.207, issue.2, pp.294-303
DOI : 10.1016/j.jmr.2010.09.014

URL : https://hal.archives-ouvertes.fr/hal-00422173

C. Charlier, Protein Dynamics from Nuclear Spin Relaxation, pp.2015-2022
URL : https://hal.archives-ouvertes.fr/tel-01333232

R. James, A. Brainard, and . Szabo, Theory for nuclear magnetic relaxation of probes in anisotropic systems: application to cholesterol in phospholipid vesicles, Biochemistry, vol.20, issue.16, pp.4618-4628

V. Calandrini, D. Abergel, and G. R. Kneller, Fractional protein dynamics seen by nuclear magnetic resonance spectroscopy: Relating molecular dynamics simulation and experiment, The Journal of Chemical Physics, vol.93, issue.14, p.145101, 2010.
DOI : 10.1021/ja00168a070

URL : https://hal.archives-ouvertes.fr/hal-00593171

P. Calligari and D. Abergel, Toward the Characterization of Fractional Stochastic Processes Underlying Methyl Dynamics in Proteins, The Journal of Physical Chemistry B, vol.116, issue.43, pp.12955-12965
DOI : 10.1021/jp307050v

B. Halle, The physical basis of model-free analysis of NMR relaxation data from proteins and complex uids, The Journal of Chemical Physics, issue.22, p.131224507, 2009.

R. Brüschweiler and P. E. Wright, NMR Order Parameters of Biomolecules: A New Analytical Representation and Application to the Gaussian Axial Fluctuation Model, Journal of the American Chemical Society, vol.116, issue.18, pp.8426-8427, 1994.
DOI : 10.1021/ja00097a084

A. Polimeno and J. H. Freed, Slow Motional ESR in Complex Fluids: The Slowly Relaxing Local Structure Model of Solvent Cage Effects, The Journal of Physical Chemistry, vol.99, issue.27, pp.10995-11006, 1995.
DOI : 10.1021/j100027a047

A. V. Buevich and J. Baum, Dynamics of Unfolded Proteins:?? Incorporation of Distributions of Correlation Times in the Model Free Analysis of NMR Relaxation Data, Journal of the American Chemical Society, vol.121, issue.37, pp.8671-8672
DOI : 10.1021/ja9910412

K. Modig and F. M. Poulsen, Model-independent interpretation of NMR relaxation data for unfolded proteins: the acid-denatured state of ACBP, Journal of Biomolecular NMR, vol.44, issue.3, pp.163-177
DOI : 10.1007/s10858-008-9280-0

N. Shahid, C. Khan, R. Charlier, N. Augustyniak, V. Salvi et al., Philippe Pelupessy, and Fabien Ferrage. Distribution of Pico-and Nanosecond Motions in Disordered Proteins from Nuclear Spin Relaxation, Biophysical Journal, vol.109, issue.5, pp.988-999

S. A. Buchli, R. Waldauer, M. L. Walser, R. Donten, N. Ppster et al., Kinetic response of a photoperturbed allosteric protein, Proceedings of the National Academy of Sciences, pp.11725-11730
DOI : 10.1002/jcc.20065

I. Paul, N. De-bakker, . Furnham, L. Tom, . Blundell et al., Conformer generation under restraints, Current Opinion in Structural Biology, vol.16, issue.2, pp.160-165

J. S. Fraser, M. W. Clarkson, S. C. Degnan, R. Erion, D. Kern et al., Hidden alternative structures of proline isomerase essential for catalysis, Nature, vol.71, issue.7273, pp.669-673
DOI : 10.1038/nature08615

T. Lang, J. M. Holton, J. S. Fraser, and T. Alber, Protein structural ensembles are revealed by redefining X-ray electron density noise, Proceedings of the National Academy of Sciences, vol.28, issue.1, pp.237-242
DOI : 10.1093/nar/28.1.235

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3890839

P. Schanda, H. Beat, M. Meier, and . Ernst, Quantitative Analysis of Protein Backbone Dynamics in Microcrystalline Ubiquitin by Solid-State NMR Spectroscopy, Journal of the American Chemical Society, vol.132, issue.45, pp.15957-15967
DOI : 10.1021/ja100726a

P. Schanda, M. Huber, J. Boisbouvier, H. Beat, M. Meier et al., Solid-State NMR Measurements of Asymmetric Dipolar Couplings Provide Insight into Protein Side-Chain Motion, Angewandte Chemie International Edition, vol.120, issue.46, pp.11005-11009
DOI : 10.1002/anie.201103944

J. L. Lorieau and A. E. Mcdermott, Conformational Flexibility of a Microcrystalline Globular Protein: Order Parameters by Solid-State NMR Spectroscopy, Journal of the American Chemical Society, vol.128, issue.35, pp.11505-11512
DOI : 10.1021/ja062443u

K. K. Frederick, M. S. Marlow, K. G. Valentine, and A. Wand, Conformational entropy in molecular recognition by proteins, Nature, vol.10, issue.7151, pp.325-329
DOI : 10.1038/nature05959

R. Joel, K. Tolman, and . Ruan, NMR Residual Dipolar Couplings as Probes of Biomolecular Dynamics, Chemical Reviews, vol.106, issue.5, pp.1720-1736

G. 123-loïc-salmon, P. Bouvignies, N. Markwick, S. Lakomek, D. Showalter et al., Protein Conformational Flexibility from Structure-Free Analysis of NMR Dipolar Couplings: Quantitative and Absolute Determination of Backbone Motion in Ubiquitin, Angewandte Chemie, vol.317, issue.23, pp.4218-4221
DOI : 10.1002/ange.200900476

N. Lakomek, F. A. Korvin, C. Walter, O. F. Farès, B. L. Lange et al., Self-consistent residual dipolar coupling based model-free analysis for the robust determination of nanosecond to microsecond protein dynamics, Journal of Biomolecular NMR, vol.23, issue.3, pp.41139-155
DOI : 10.1007/s10858-008-9244-4

C. Farès, N. Lakomek, F. A. Korvin, . Walter, T. C. Benedikt et al., Accessing ns?????s side chain dynamics in ubiquitin with methyl RDCs, Journal of Biomolecular NMR, vol.122, issue.51???55, pp.23-44
DOI : 10.1007/s10858-009-9354-7

K. B. Briggman and J. R. Tolman, De Novo Determination of Bond Orientations and Order Parameters from Residual Dipolar Couplings with High Accuracy, Journal of the American Chemical Society, vol.125, issue.34, pp.10164-10165, 2003.
DOI : 10.1021/ja035904+

W. Peti, J. Meiler, R. Brüschweiler, and C. Griesinger, Model-Free Analysis of Protein Backbone Motion from Residual Dipolar Couplings, Journal of the American Chemical Society, vol.124, issue.20, pp.5822-5833
DOI : 10.1021/ja011883c

L. E. Kay, D. A. Torchia, and A. Bax, Backbone dynamics of proteins as studied by nitrogen-15 inverse detected heteronuclear NMR spectroscopy: application to staphylococcal nuclease, Biochemistry, vol.28, issue.23, pp.288972-8979, 1989.
DOI : 10.1021/bi00449a003

N. A. Farrow, R. Muhandiram, A. U. Singer, S. M. Pascal, C. M. Kay et al., Backbone Dynamics of a Free and a Phosphopeptide-Complexed Src Homology 2 Domain Studied by 15N NMR Relaxation, Biochemistry, vol.33, issue.19, pp.335984-6003
DOI : 10.1021/bi00185a040

S. Ramboarina and C. Reddeld, Probing the Effect of Temperature on the Backbone Dynamics of the Human ??-Lactalbumin Molten Globule, Journal of the American Chemical Society, vol.130, issue.46, pp.15318-15326
DOI : 10.1021/ja802967k

P. Lundström, K. Teilum, T. Carstensen, I. Bezsonova, S. Wiesner et al., Fractional 13C enrichment of isolated carbons using [1-13C]- or [2-13C]-glucose facilitates the accurate measurement of dynamics at backbone C?? and side-chain methyl positions in proteins, Journal of Biomolecular NMR, vol.7, issue.3, pp.199-212
DOI : 10.1007/s10858-007-9158-6

J. Michael, J. Plevin, and . Boisbouvier, Chapter 1. Isotope-Labelling of Methyl Groups for NMR Studies of Large Proteins, RSC Biomolecular Sciences, pp.1-24, 2012.

Y. Zhang, L. Zhou, L. Rouge, H. Aaron, C. Phillips et al., Conformational stabilization of ubiquitin yields potent and selective inhibitors of USP7, Nature Chemical Biology, vol.20, issue.1, pp.51-58
DOI : 10.1038/nchembio.1134

F. Lange, N. Lakomek, C. Fares, G. F. Schroder, K. F. Walter et al., Recognition Dynamics Up to Microseconds Revealed from an RDC-Derived Ubiquitin Ensemble in Solution, Science, vol.117, issue.7147, pp.3201471-1475
DOI : 10.1038/nature05858

. Ochsenbein, Dynamical characterization of residual and non-native structures in a partially folded protein by 15N NMR relaxation using a model based on a distribution of correlation times, Protein Science, vol.11, issue.4, pp.957-964
DOI : 10.1110/ps.4000102

V. Tugarinov, Z. Liang, Y. E. Shapiro, J. H. Freed, and E. Meirovitch, N NMR Relaxation in Proteins, Journal of the American Chemical Society, vol.123, issue.13, pp.3055-3063
DOI : 10.1021/ja003803v

E. Persson and B. Halle, Nanosecond to Microsecond Protein Dynamics Probed by Magnetic Relaxation Dispersion of Buried Water Molecules, Journal of the American Chemical Society, vol.130, issue.5, pp.1774-1787
DOI : 10.1021/ja0775873

O. Millet, D. R. Muhandiram, N. R. Skrynnikov, and L. E. Kay, Deuterium Spin Probes of Side-Chain Dynamics in Proteins. 1. Measurement of Five Relaxation Rates per Deuteron in 13C- Labeled and Fractionally 2H-Enriched Proteins in Solution, Journal of the American Chemical Society, issue.22, pp.1246439-6448

N. R. Skrynnikov, O. Millet, and L. E. Kay, Deuterium Spin Probes of Side-Chain Dynamics in Proteins. 2. Spectral Density Mapping and Identification of Nanosecond Time-Scale Side-Chain Motions, Journal of the American Chemical Society, vol.124, issue.22, pp.6449-6460
DOI : 10.1021/ja012498q

A. Scott, E. Showalter, M. Johnson, R. Rance, and . Brüschweiler, Toward Quantitative Interpretation of Methyl Side-Chain Dynamics from NMR by Molecular Dynamics Simulations, Journal of the American Chemical Society, vol.129, issue.46, pp.14146-14147

W. Ole, J. C. Sørensen, . Madsen, H. Nielsen-chr-nielsen, . Bildsøe et al., An improved refocused INEPT experiment. Application for sensitivity enhancement and spectral editing in 13C NMR, Journal of Magnetic Resonance, vol.77, issue.1, pp.170-174, 1969.

R. E. Deverell, J. H. Morgan, and . Strange, Studies of chemical exchange by nuclear magnetic relaxation in the rotating frame, Molecular Physics, vol.249, issue.4, pp.553-559
DOI : 10.1021/ja00862a015

F. Massi, E. Johnson, C. Wang, M. Rance, and A. G. Palmer, Rotating-Frame Relaxation with Weak Radio Frequency Fields, Journal of the American Chemical Society, vol.126, issue.7, pp.2247-2256
DOI : 10.1021/ja038721w

D. Ban, A. D. Gossert, K. Giller, S. Becker, C. Griesinger et al., Exceeding the limit of dynamics studies on biomolecules using high spin-lock field strengths with a cryogenically cooled probehead, Journal of Magnetic Resonance, vol.221, pp.1-4
DOI : 10.1016/j.jmr.2012.05.005

A. 145-fabien-ferrage, D. Piserchio, R. Cowburn, and . Ghose, On the measurement of 15N???{1H} nuclear Overhauser effects, Journal of Magnetic Resonance, vol.192, issue.2, pp.302-313
DOI : 10.1016/j.jmr.2008.03.011

L. Kay, P. Keifer, and T. Saarinen, Pure absorption gradient enhanced heteronuclear single quantum correlation spectroscopy with improved sensitivity, Journal of the American Chemical Society, vol.114, issue.26, pp.10663-10665
DOI : 10.1021/ja00052a088

H. Akaike, A new look at the statistical model identiication. Automatic Control, IEEE Transactions on, vol.19, issue.6, pp.716-723, 1974.

J. Edward and P. R. Gooley, Optimisation of NMR dynamic models II. A new methodology for the dual optimisation of the model-free parameters and the Brownian rotational diiusion tensor, Journal of Biomolecular NMR, vol.40, issue.2, pp.121-133

J. Edward and P. R. Gooley, Optimisation of NMR dynamic models I. Minimisation algorithms and their performance within the model-free and Brownian rotational diiusion spaces, Journal of Biomolecular NMR, vol.40, issue.2, pp.107-119

A. L. Lee, P. F. Flynn, and A. Wand, C NMR Relaxation Techniques for the Study of Protein Methyl Group Dynamics in Solution, Journal of the American Chemical Society, vol.121, issue.12, pp.2891-2902
DOI : 10.1021/ja983758f

M. Roos, M. Hofmann, S. Link, M. Ott, J. Balbach et al., The ???long tail??? of the protein tumbling correlation function: observation by 1H NMR relaxometry in a wide frequency and concentration range, Journal of Biomolecular NMR, vol.75, issue.4, pp.403-415
DOI : 10.1007/s10858-015-0001-1

R. Kerfah and J. Boisbouvier, AILM 2015 Patractical school, pp.2015-2016

P. Weitekamp, A. Bielecki, D. Zax, K. Zilm, and A. Pines, Zero--eld nuclear magnetic resonance. Physical review letters, p.501807, 1983.

B. Zax, A. Bielecki, K. W. Zilm, A. Pines, and D. P. Weitekamp, Zero field NMR and NQR, The Journal of Chemical Physics, vol.5, issue.10, p.4877, 1985.
DOI : 10.1063/1.435038

D. Reese, T. Lennartz, P. Marquardsen, A. Höfer, P. Tavernier et al., Construction of a Liquid-State NMR DNP Shuttle Spectrometer: First Experimental Results and Evaluation of Optimal Performance Characteristics, Applied Magnetic Resonance, vol.106, issue.3-4, pp.3-4301
DOI : 10.1007/s00723-008-0131-7

H. Jan, B. Ardenkj\a-er-larsen, A. Fridlund, G. Gram, L. Hansson et al., Increase in signal-to-noise ratio of> 10,000 times in liquid-state NMR, Proceedings of the National Academy of Sciences, vol.100, issue.18, pp.10158-10163, 2003.

A. Bornet, S. Jannin, and G. Bodenhausen, Three--eld NMR to preserve hyperpolarized proton magnetization as long-lived states in moderate magnetic elds, Chemical Physics Letters, vol.512, pp.4-6151

F. Grosse, H. Gubaydullin, H. M. Scheelken, A. V. Vieth, and . Yurkovskaya, Field cycling by fast NMR probe transfer: Design and application in field-dependent CIDNP experiments, Applied Magnetic Resonance, vol.46, issue.2-3, pp.211-225
DOI : 10.1007/BF03162162

O. Lips, A. F. Privalov, S. V. Dvinskikh, and F. Fujara, Magnet Design with High B0 Homogeneity for Fast-Field-Cycling NMR Applications, Journal of Magnetic Resonance, vol.149, issue.1, pp.22-28
DOI : 10.1006/jmre.2000.2279

. Noack, NMR field-cycling spectroscopy: principles and a]lications, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.18, issue.3, pp.171-276
DOI : 10.1016/0079-6565(86)80004-8

K. Modig and B. Halle, Proton Magnetic Shielding Tensor in Liquid Water, Journal of the American Chemical Society, vol.124, issue.40, pp.12031-12041
DOI : 10.1021/ja026981s

G. Allen, P. C. Stephenson, and J. H. Strange, Morphology of porous media studied by nuclear magnetic resonance, The Journal of Chemical Physics, vol.106, issue.18, p.7802, 1997.
DOI : 10.1016/0730-725X(91)90365-S

P. Korb, Nuclear magnetic relaxation of liquids in porous media, New Journal of Physics, vol.13, issue.3, pp.35016-2011
DOI : 10.1088/1367-2630/13/3/035016

W. P. Slichter, NMR studies of multiple relaxations in polymers, Journal of Polymer Science Part C: Polymer Symposia, vol.70, issue.1, pp.33-48
DOI : 10.1002/polc.5070140107

K. Hatada and T. Kitayama, NMR Relaxation, NMR Spectroscopy of Polymers, pp.169-197, 2004.
DOI : 10.1007/978-3-662-08982-8_7

A. Krahn, P. Lottmann, T. Marquardsen, A. Tavernier, M. Türke et al., Shuttle DNP spectrometer with a two-center magnet, Physical Chemistry Chemical Physics, vol.15, issue.3, p.125830, 2010.
DOI : 10.1039/c002814m

URL : http://hdl.handle.net/11858/00-001M-0000-0012-D69D-7

A. G. Reddeld, Shuttling device for high-resolution measurements of relaxation and related phenomena in solution at low field, using a shared commercial 500 MHz NMR instrument, Magnetic Resonance in Chemistry, vol.84, issue.10, pp.753-768
DOI : 10.1002/mrc.1264

. Vacuumschmelze, Soft Magnetic Materials and semi--nished Products. 2002-01. US Patent App, p.466932

J. Nová?ek, L. Janda, R. Dopitová, L. ?ídek, and V. Sklená?, Efficient protocol for backbone and side-chain assignments of large, intrinsically disordered proteins: transient secondary structure analysis of 49.2??kDa microtubule associated protein 2c, Journal of Biomolecular NMR, vol.52, issue.4, pp.291-301
DOI : 10.1007/s10858-013-9761-7

M. Sattler, J. Schleucher, and C. Griesinger, Heteronuclear multidimensional NMR experiments for the structure determination of proteins in solution, Progress in nuclear magnetic resonance spectroscopy, pp.93-158, 1999.

T. Gaetano, B. A. Montelione, S. D. Lyons, M. Emerson, and . Tashiro, An eecient triple resonance experiment using carbon-13 isotropic mixing for determining sequence-speciic resonance assignments of isotopically-enriched proteins, Journal of the American Chemical Society, vol.114, issue.27, pp.10974-10975

T. Bennett, R. A. Farmer, and . Venters, Assignment of side-chain 13C resonances in perdeuterated proteins, Journal of the American Chemical Society, vol.117, issue.14, pp.4187-4188, 1995.

J. Shaka, C. J. Lee, and A. Pines, Iterative schemes for bilinear operators; application to spin decoupling, Journal of Magnetic Resonance (1969), vol.77, issue.2, pp.274-293, 1969.
DOI : 10.1016/0022-2364(88)90178-3

O. Kadkhodaie, M. Rivas, A. Tan, A. J. Mohebbi, and . Shaka, Broadband homonuclear cross polarization using ip--op spectroscopy, Journal of Magnetic Resonance, issue.2, pp.91437-443, 1969.
DOI : 10.1016/0022-2364(91)90210-k

K. L. Ivanov, A. N. Pravdivtsev, A. V. Yurkovskaya, H. Vieth, and R. Kaptein, The role of level anti-crossings in nuclear spin hyperpolarization, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.81, pp.1-36
DOI : 10.1016/j.pnmrs.2014.06.001

L. Suzanne, . Duce, D. Laurance, T. J. Hall, and . Norwood, High-resolution heteronuclear NMR spectroscopy in an inhomogeneous magnetic eld, Journal of Magnetic Resonance, vol.89, issue.2, pp.273-286, 1969.

A. Bax, R. Freeman, T. Frenkiel, and M. H. Levitt, Assignment of carbon-13 NMR spectra via double-quantum coherence, Journal of Magnetic Resonance (1969), vol.43, issue.3, pp.478-483, 1969.
DOI : 10.1016/0022-2364(81)90060-3

A. Bax and R. Freeman, Investigation of complex networks of spin-spin coupling by twodimensional NMR, Journal of Magnetic Resonance, vol.44, issue.3, pp.542-561, 1969.

A. W. Overhauser, Polarization of Nuclei in Metals, Physical Review, vol.92, issue.2, pp.411-415
DOI : 10.1103/PhysRev.92.411

R. Carver and C. P. Slichter, Polarization of Nuclear Spins in Metals, Physical Review, vol.92, issue.1, pp.212-213
DOI : 10.1103/PhysRev.92.212.2

J. Shaka, J. Keeler, T. Frenkiel, and R. A. Freeman, An improved sequence for broadband decoupling: WALTZ-16, Journal of Magnetic Resonance (1969), vol.52, issue.2, pp.335-338, 1969.
DOI : 10.1016/0022-2364(83)90207-X

K. Marion and . Wüthrich, Application of phase sensitive two-dimensional correlated spectroscopy (COSY) for measurements of 1H-1H spin-spin coupling constants in proteins, Biochemical and Biophysical Research Communications, vol.113, issue.3, pp.967-974
DOI : 10.1016/0006-291X(83)91093-8

L. Mueller, Sensitivity enhanced detection of weak nuclei using heteronuclear multiple quantum coherence, Journal of the American Chemical Society, vol.101, issue.16, pp.4481-4484, 1979.
DOI : 10.1021/ja00510a007

G. Bodenhausen, H. Kogler, and R. R. Ernst, Selection of coherence-transfer pathways in NMR pulse experiments, Journal of Magnetic Resonance, vol.58, issue.3, pp.370-388, 1969.
URL : https://hal.archives-ouvertes.fr/hal-00730604

K. Marion and . Wuthrich, Application of Phase Sensitive Two-Dimensional Correlated Spectroscopy (COSY) for measurements of 1H-1H Spin-Spin Coupling Constants in Proteins, NMR in Structural Biology: A Collection of Papers by Kurt Wüthrich, p.114, 1995.

J. Shaka, P. B. Barker, and R. Freeman, Computer-optimized decoupling scheme for wideband applications and low-level operation, Journal of Magnetic Resonance (1969), vol.64, issue.3, pp.547-552, 1969.
DOI : 10.1016/0022-2364(85)90122-2

V. Tugarinov, P. M. Hwang, J. E. Ollerenshaw, and L. E. Kay, C NMR Spectroscopy of Methyl Groups in Very High Molecular Weight Proteins and Protein Complexes, Journal of the American Chemical Society, vol.125, issue.34, pp.10420-10428
DOI : 10.1021/ja030153x

C. Reddeld, R. A. Smith, and C. M. Dobson, Structural characterization of a highly???ordered ???molten globule??? at low pH, Nature Structural Biology, vol.31, issue.1, pp.23-29
DOI : 10.1107/S0021889891004399

J. and P. Wright, Formation of a molten globule intermediate early in the kinetic folding pathway of apomyoglobin, Science, vol.262, issue.5135, pp.892-896

B. A. Schulman, P. S. Kim, C. M. Dobson, and C. Reddeld, A residue-specific NMR view of the non-cooperative unfolding of a molten globule, Natural Structural Biology, vol.29, issue.8, pp.630-634
DOI : 10.1016/0022-2836(91)80073-4

. Reddeld, Using nuclear magnetic resonance spectroscopy to study molten globule states of proteins, Methods, vol.34, issue.1, pp.121-132
DOI : 10.1016/j.ymeth.2004.03.009

A. D. Bain, Chemical exchange in NMR, Progress in Nuclear Magnetic Resonance Spectroscopy, vol.43, issue.3-4, pp.63-103
DOI : 10.1016/j.pnmrs.2003.08.001

S. Christian, S. J. Rondestvedt, and . Davis, 1-Aryl-3,3-dialkyltriazenes as Tumor Inhibitors, The Journal of Organic Chemistry, vol.22, issue.2, pp.200-203

H. Haario, E. Saksman, and J. Tamminen, An Adaptive Metropolis Algorithm, Bernoulli, vol.7, issue.2, pp.2001-2005
DOI : 10.2307/3318737

URL : http://citeseerx.ist.psu.edu/viewdoc/summary?doi=10.1.1.48.8948

C. Chou, F. Ferrage, G. Aubert, and D. Sakellariou, Simple method for the generation of multiple homogeneous field volumes inside the bore of superconducting magnets, Scientific Reports, vol.100, issue.1, pp.12200-2015
DOI : 10.1073/pnas.1133497100

URL : https://hal.archives-ouvertes.fr/hal-01223310

S. Laurent, L. Vander-elst, and R. N. Muller, Comparative study of the physicochemical properties of six clinical low molecular weight gadolinium contrast agents, Contrast Media & Molecular Imaging, vol.7, issue.3, pp.128-137
DOI : 10.1002/cmmi.100

S. Aime, M. Botta, S. Geninatti-crich, G. B. Giovenzana, R. Pagliarin et al., Towards MRI contrast agents of improved efficacy. NMR relaxometric investigations of the binding interaction to HSA of a novel heptadentate macrocyclic triphosphonate Gd(III)-complex, Journal of Biological Inorganic Chemistry, vol.2, issue.4, pp.470-479, 1997.
DOI : 10.1007/s007750050158

Y. Gossuin, Z. Serhan, L. Sandiford, D. Henrard, T. Marquardsen et al., Sample Shuttling Relaxometry of Contrast Agents: NMRD Profiles above 1 T with a Single Device, Applied Magnetic Resonance, vol.293, issue.3, pp.237-246
DOI : 10.1007/s00723-015-0751-7

URL : https://hal.archives-ouvertes.fr/hal-01266671

C. Dalvit, P. Pevarello, M. Tatò, M. Veronesi, A. Vulpetti et al., Identiication of compounds with binding aanity to proteins via magnetization transfer from bulk water, Journal of Biomolecular NMR, vol.18, issue.1, pp.65-68, 2000.
DOI : 10.1023/A:1008354229396

A. G. Reddeld, Field-Cycling NMR Applied to Macromolecular Structure and Dynamics, NMR as a Structural Tool for Macromolecules: Current Status and Future Directions, pp.123-132, 1996.

V. Borsi, C. Luchinat, and G. Parigi, Global and Local Mobility of Apocalmodulin Monitored through Fast-Field Cycling Relaxometry, Biophysical Journal, vol.97, issue.6, pp.1765-1771
DOI : 10.1016/j.bpj.2009.07.005

Z. Gan, P. Gor-'kov, T. A. Cross, A. Samoson, and D. Massiot, Seeking Higher Resolution and Sensitivity for NMR of Quadrupolar Nuclei at Ultrahigh Magnetic Fields, Journal of the American Chemical Society, vol.124, issue.20, pp.5634-5635
DOI : 10.1021/ja025849p