G. M. Griffiths, A. Tsun, and J. C. Stinchcombe, The immunological synapse: a focal point for endocytosis and exocytosis, J. Cell Biol, vol.189, pp.399-406, 2010.

F. D. Batista and M. L. Dustin, Cell: cell interactions in the immune system, Immunol. Rev, vol.251, pp.7-12, 2013.

F. D. Batista, D. Iber, and M. S. Neuberger, B cells acquire antigen from target cells after synapse formation, Nature, vol.411, pp.489-494, 2001.

H. Qi, J. G. Egen, A. Y. Huang, and R. N. Germain, Extrafollicular activation of lymph node B cells by antigen-bearing dendritic cells, Science, vol.312, pp.1672-1676, 2006.

K. Suzuki, I. Grigorova, T. G. Phan, L. M. Kelly, and J. G. Cyster, Visualizing B cell capture of cognate antigen from follicular dendritic cells, J. Exp. Med, vol.206, pp.1485-1493, 2009.

Y. R. Carrasco, S. J. Fleire, T. Cameron, M. L. Dustin, and F. D. Batista, LFA-1/ ICAM-1 interaction lowers the threshold of B cell activation by facilitating B cell adhesion and synapse formation, Immunity, vol.20, pp.589-599, 2004.

F. D. Batista and N. E. Harwood, The who, how and where of antigen presentation to B cells, Nat. Rev. Immunol, vol.9, pp.15-27, 2009.

E. Natkanski, B cells use mechanical energy to discriminate antigen affinities. Materials and methods, Science, vol.340, pp.1587-1590, 2013.

M. Yuseff, Polarized secretion of lysosomes at the B cell synapse couples antigen extraction to processing and p, Immunity, vol.35, pp.361-374, 2011.

S. J. Fleire, B cell ligand discrimination through a spreading and contraction response, Science, vol.312, pp.738-741, 2006.

P. K. Mattila, The actin and tetraspanin networks organize receptor nanoclusters to regulate B cell receptor-mediated signaling, Immunity, vol.38, pp.461-474, 2013.

K. Kwak, Intrinsic properties of human germinal center B cells set antigen affinity thresholds, Sci. Immunol, vol.3, pp.1-14, 2018.

M. Dembo and Y. L. Wang, Stresses at the cell-to-substrate interface during locomotion of fibroblasts, Biophys. J, vol.76, pp.2307-2316, 1999.

B. Sabass, M. L. Gardel, C. M. Waterman, and U. S. Schwarz, High resolution traction force microscopy based on experimental and computational advances, Biophys. J, vol.94, pp.207-220, 2008.

K. Mandal, I. Wang, E. Vitiello, L. A. Orellana, and M. Balland, Cell dipole behaviour revealed by ECM sub-cellular geometry, Nat. Commun, vol.5, p.5749, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02063891

N. Bufi, Human primary immune cells exhibit distinct mechanical properties that are modified by inflammation, Biophys. J, vol.108, pp.2181-2190, 2015.

Z. Wan, B Cell Activation Is Regulated by the Stiffness Properties of the Substrate Presenting the Antigens, J. Immunol, vol.190, pp.4661-4675, 2013.

Y. Zeng, Substrate stiffness regulates B-cell activation, proliferation, class switch and T-cell-independent antibody responses in vivo, Eur. J. Immunol, vol.45, pp.1621-1634, 2015.

R. Hoogeboom, Myosin IIa promotes antibody responses by regulating B cell activation, acquisition of antigen, and proliferation, Cell Rep, vol.23, pp.2342-2353, 2018.

P. T. Sage, Antigen recognition is facilitated by invadosome-like protrusions formed by memory/effector T cells, J. Immunol, vol.188, pp.3686-3699, 2012.

D. Martino and J. , The microenvironment controls invadosome plasticity, J. Cell Sci, vol.129, pp.1759-1768, 2016.

M. Bretou, Lysosome signaling controls the migration of dendritic cells, Sci. Immunol, vol.2, p.9573, 2017.

J. Wang, Profiling the origin, dynamics, and function of traction force in B cell activation, Sci. Signal, vol.11, p.9192, 2018.

C. Liu, Analyzing actin dynamics during the activation of the B cell receptor in live B cells, Biochem. Biophys. Res. Commun, vol.427, pp.202-206, 2012.

S. Murugesan, Formin-generated actomyosin arcs propel T cell receptor microcluster movement at the immune synapse, J. Cell Biol, vol.215, pp.383-399, 2016.

A. Chaudhuri, B. Bhattacharya, K. Gowrishankar, S. Mayor, and M. Rao, Spatiotemporal regulation of chemical reactions by active cytoskeletal remodeling, Proc. Natl Acad. Sci. USA, vol.108, pp.14825-14830, 2011.

K. Gowrishankar, Active remodeling of cortical actin regulates spatiotemporal organization of cell surface molecules, Cell, vol.149, pp.1353-1367, 2012.

E. Cai, Visualizing dynamic microvillar search and stabilization during ligand detection by T cells, Science, vol.356, p.3118, 2017.

A. I. Shevchuk, An alternative mechanism of clathrin-coated pit closure revealed by ion conductance microscopy, J. Cell Biol, vol.197, pp.499-508, 2012.

C. Basquin, Membrane protrusion powers clathrin-independent endocytosis of interleukin-2 receptor, EMBO J, vol.34, pp.2147-2161, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01202886

W. S. Trimble and S. Grinstein, Barriers to the free diffusion of proteins and lipids in the plasma membrane, J. Cell Biol, vol.208, pp.259-271, 2015.

S. A. Freeman, Integrins form an expanding diffusional garrier that coordinates phagocytosis, Cell, vol.164, pp.128-140, 2016.

M. E. Maxson, Integrin-based diffusion barrier separates membrane domains enabling the formation of microbiostatic frustrated phagosomes, vol.7, pp.1-37, 2018.

A. Reversat, Polarity protein Par3 controls B-cell receptor dynamics and antigen extraction at the immune synapse, Mol. Biol. Cell, vol.26, pp.1273-1285, 2015.

M. Kaksonen, C. P. Toret, and D. G. Drubin, Harnessing actin dynamics for clathrin-mediated endocytosis, Nat. Rev. Mol. Cell Biol, vol.7, pp.404-414, 2006.

J. Cheng, A. Grassart, and D. G. Drubin, Myosin 1E coordinates actin assembly and cargo trafficking during clathrin-mediated endocytosis, Mol. Biol. Cell, vol.23, pp.2891-2904, 2012.

R. T. Pedersen and D. G. Drubin, Type I myosins anchor actin assembly to the plasma membrane during clathrin-mediated endocytosis, J. Cell Biol, vol.218, pp.1138-1147

Z. Wan, The activation of IgM-or isotype-switched IgG-and IgE-BCR exhibits distinct mechanical force sensitivity and threshold, Elife, vol.4, pp.1-24, 2015.

B. B. Mcintosh and E. M. Ostap, Myosin-I molecular motors at a glance, J. Cell Sci, vol.129, pp.2689-2695, 2016.

J. Lohner, Myosin filaments reversibly generate large forces in cells, Nat. Phys, 2019.

C. Watts, Antigen processing in the endocytic compartment, Curr. Opin. Immunol, vol.13, pp.26-31, 2001.

C. P. Heisenberg and Y. Bellaïche, Forces in tissue morphogenesis and patterning, Cell, vol.153, pp.948-962, 2013.

S. V. Plotnikov, A. M. Pasapera, B. Sabass, and C. M. Waterman, Force fluctuations within focal adhesions mediate ECM-rigidity sensing to guide directed cell migration, Cell, vol.151, pp.1513-1527, 2012.

B. Yang, Mechanosensing controlled directly by tyrosine kinases, Nano Lett, vol.16, pp.5951-5961, 2016.

J. Shiu, L. Aires, Z. Lin, and V. Vogel, Nanopillar force measurements reveal actin-cap-mediated YAP mechanotransduction, Nat. Cell Biol, vol.20, pp.262-271, 2018.

X. Trepat, Physical forces during collective cell migration, Nat. Phys, vol.5, pp.426-430, 2009.

J. Jacobelli, Confinement-optimized three-dimensional T cell amoeboid motility is modulated via myosin IIA-regulated adhesions, Nat. Immunol, vol.11, pp.953-961, 2010.

J. Riedl, Lifeact: a versatile marker to visualize F-actin, Nat. Methods, vol.5, pp.605-607, 2008.

C. C. Goodnow, Altered immunoglobulin expression and functional silencing of self-reactive B lymphocytes in transgenic mice, Nature, vol.334, pp.676-682, 1988.

R. Long, M. S. Hall, M. Wu, and C. Y. Hui, Effects of gel thickness on microscopic indentation measurements of gel modulus, Biophys. J, vol.101, pp.643-650, 2011.

K. Schauer, Probabilistic density maps to study global endomembrane organization, Nat. Methods, vol.7, pp.560-566, 2010.

J. P. Butler, I. V. Tolic, B. E. Fabry, and J. J. Fredberg, Traction fields, moments, and strain energy that cells exert on their surroundings, Am. J. Physiol. Cell Physiol, vol.282, pp.595-605, 2002.

N. Bertaux, D. Marguet, and A. Serge, Dynamic multiple-target tracing to probe spatiotemporal cartography of cell membranes, Nat. Methods, vol.5, pp.687-694, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00295014

D. Waithe, Super-resolved traction force microscopy (STFM), Nano Lett, vol.16, pp.2633-2638, 2016.

S. V. Plotnikov, B. Sabass, U. S. Schwarz, and C. M. Waterman, HighResolution Traction Force Microscopy, Methods in Cell Biology, vol.123, pp.367-394, 2014.

W. R. Legant, Multidimensional traction force microscopy reveals outof-plane rotational moments about focal adhesions, Proc. Natl Acad. Sci, vol.110, pp.881-886, 2013.