Mechanotargeting: Mechanics‐Dependent Cellular Uptake of Nanoparticles

Targeted delivery of nanoparticle (NP)-based diagnostic and therapeutic agents to malignant cells and tissues has exclusively relied on chemotargeting, wherein NPs are surface-coated with ligands that specifically bind to overexpressed receptors on malignant cells. Here, it is demonstrated that cellular uptake of NPs can also be biased to malignant cells based on the differential mechanical states of cells, enabling mechanotargeting. Owing to mechanotransduction, cell lines (HeLa and HCT-8) cultured on hydrogels of various stiffness are directed into different stress states, measured by cellular force microscopies. In vitro NP delivery reveals that increases in cell stress suppress cellular uptake, counteracting the enhanced uptake that occurs with increases in exposed surface area of spread cells. Upon prolonged culture on stiff hydrogels, cohesive HCT-8 cell colonies undergo metastatic phenotypic change and disperse into individual malignant cells. The metastatic cells are of extremely low stress state and adopt an unspread, 3D morphology, resulting in several-fold higher uptake than the nonmetastatic counterparts. This study opens a new paradigm of harnessing mechanics for the design of future strategies in nanomedicine.

[1]  Changyou Gao,et al.  Influence of structure and properties of colloidal biomaterials on cellular uptake and cell functions. , 2013, Biomaterials science.

[2]  Jianping Fu,et al.  Cell shape and substrate rigidity both regulate cell stiffness. , 2011, Biophysical journal.

[3]  J. Rao,et al.  Nanomechanical analysis of cells from cancer patients. , 2007, Nature nanotechnology.

[4]  Silviya Zustiak,et al.  Multiwell stiffness assay for the study of cell responsiveness to cytotoxic drugs , 2014, Biotechnology and bioengineering.

[5]  P. Janmey,et al.  Tissue Cells Feel and Respond to the Stiffness of Their Substrate , 2005, Science.

[6]  J. Karp,et al.  Nanocarriers as an Emerging Platform for Cancer Therapy , 2022 .

[7]  J. Haga,et al.  Molecular basis of the effects of shear stress on vascular endothelial cells. , 2005, Journal of biomechanics.

[8]  Valerie M. Weaver,et al.  A tense situation: forcing tumour progression , 2009, Nature Reviews Cancer.

[9]  J. Fredberg,et al.  Collective cell guidance by cooperative intercellular forces , 2010, Nature materials.

[10]  Martin A. Schwartz,et al.  Cell adhesion: integrating cytoskeletal dynamics and cellular tension , 2010, Nature Reviews Molecular Cell Biology.

[11]  G. Salbreux,et al.  Stresses at the Cell Surface during Animal Cell Morphogenesis , 2014, Current Biology.

[12]  Huajian Gao,et al.  Physical Principles of Nanoparticle Cellular Endocytosis. , 2015, ACS nano.

[13]  T. Park,et al.  Diverse Applications of Nanomedicine , 2017, ACS nano.

[14]  Nastassja A. Lewinski,et al.  A new era for cancer treatment: gold-nanoparticle-mediated thermal therapies. , 2011, Small.

[15]  Huajian Gao,et al.  Mechanics of receptor-mediated endocytosis. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[16]  T. Saif,et al.  A mechanically-induced colon cancer cell population shows increased metastatic potential , 2014, Molecular Cancer.

[17]  A. Bretscher,et al.  ERM proteins and merlin: integrators at the cell cortex , 2002, Nature Reviews Molecular Cell Biology.

[18]  Subra Suresh,et al.  Size‐Dependent Endocytosis of Nanoparticles , 2009, Advanced materials.

[19]  T. Xia,et al.  Understanding biophysicochemical interactions at the nano-bio interface. , 2009, Nature materials.

[20]  Baoquan Ding,et al.  One-Step Microfluidic Synthesis of Nanocomplex with Tunable Rigidity and Acid-Switchable Surface Charge for Overcoming Drug Resistance. , 2017, Small.

[21]  Sean X. Sun,et al.  A mechanical model of actin stress fiber formation and substrate elasticity sensing in adherent cells , 2010, Proceedings of the National Academy of Sciences.

[22]  Jiaxi Zhou,et al.  Mechanical force affects expression of an in vitro metastasis-like phenotype in HCT-8 cells. , 2010, Biophysical journal.

[23]  A. J. Tavares,et al.  Analysis of nanoparticle delivery to tumours , 2016 .

[24]  H. Guillou,et al.  Spatial organization of the extracellular matrix regulates cell–cell junction positioning , 2012, Proceedings of the National Academy of Sciences.

[25]  P. Janmey,et al.  Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. , 2005, Cell motility and the cytoskeleton.

[26]  Leann Tilley,et al.  Multiple stiffening effects of nanoscale knobs on human red blood cells infected with Plasmodium falciparum malaria parasite , 2015, Proceedings of the National Academy of Sciences.

[27]  Arezou A Ghazani,et al.  Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. , 2006, Nano letters.

[28]  Sarah Seifert,et al.  Image-based analysis of lipid nanoparticle–mediated siRNA delivery, intracellular trafficking and endosomal escape , 2013, Nature Biotechnology.

[29]  K. Dawson,et al.  Effects of Transport Inhibitors on the Cellular Uptake of Carboxylated Polystyrene Nanoparticles in Different Cell Lines , 2011, PloS one.

[30]  André F. Vieira,et al.  Actin stress fiber organization promotes cell stiffening and proliferation of pre-invasive breast cancer cells , 2017, Nature Communications.

[31]  Y. Wang,et al.  Cell locomotion and focal adhesions are regulated by substrate flexibility. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[32]  Baoquan Ding,et al.  Tunable Rigidity of (Polymeric Core)–(Lipid Shell) Nanoparticles for Regulated Cellular Uptake , 2015, Advanced materials.

[33]  Justin L. Brown,et al.  The role of substrate topography on the cellular uptake of nanoparticles. , 2016, Journal of biomedical materials research. Part B, Applied biomaterials.

[34]  Sanjay Kumar,et al.  Mechanics, malignancy, and metastasis: The force journey of a tumor cell , 2009, Cancer and Metastasis Reviews.

[35]  M. Ferrari Cancer nanotechnology: opportunities and challenges , 2005, Nature Reviews Cancer.

[36]  Mark E. Davis,et al.  Nanoparticle therapeutics: an emerging treatment modality for cancer , 2008, Nature Reviews Drug Discovery.

[37]  P. Butler,et al.  Substrate stiffness regulates cellular uptake of nanoparticles. , 2013, Nano letters.

[38]  Joe Tien,et al.  Mechanotransduction at cell-matrix and cell-cell contacts. , 2004, Annual review of biomedical engineering.

[39]  Ulrich S Schwarz,et al.  United we stand – integrating the actin cytoskeleton and cell–matrix adhesions in cellular mechanotransduction , 2012, Journal of Cell Science.

[40]  G. Bao,et al.  Variable nanoparticle-cell adhesion strength regulates cellular uptake. , 2010, Physical review letters.

[41]  E. Isacoff,et al.  Evoked and Spontaneous Transmission Favored by Distinct Sets of Synapses , 2014, Current Biology.

[42]  Cheng Zhu,et al.  Mechanical regulation of a molecular clutch defines force transmission and transduction in response to matrix rigidity , 2016, Nature Cell Biology.

[43]  D. Ingber,et al.  Mechanotransduction across the cell surface and through the cytoskeleton , 1993 .

[44]  Manfred Radmacher,et al.  Softening of the actin cytoskeleton by inhibition of myosin II , 2008, Pflügers Archiv - European Journal of Physiology.

[45]  Warren C W Chan,et al.  Nanoparticle-mediated cellular response is size-dependent. , 2008, Nature nanotechnology.

[46]  Ben Fabry,et al.  Traction fields, moments, and strain energy that cells exert on their surroundings. , 2002, American journal of physiology. Cell physiology.

[47]  Huajian Gao,et al.  Cellular uptake of elastic nanoparticles. , 2011, Physical review letters.

[48]  P. Couvreur,et al.  Nanoparticles in cancer therapy and diagnosis. , 2002, Advanced drug delivery reviews.