Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
bioRxiv ; 2024 Feb 29.
Article En | MEDLINE | ID: mdl-38464100

Doublecortin (DCX) is a microtubule-associated protein critical for brain development. Although most highly expressed in the developing central nervous system, the molecular function of DCX in neuron morphogenesis remains unknown and controversial. We demonstrate that DCX function is intimately linked to its microtubule-binding activity. By using human induced pluripotent stem cell (hiPSC)- derived cortical i 3 Neurons genome engineered to express mEmerald-tagged DCX from the endogenous locus, we find that DCX-MT interactions become highly polarized very early during neuron morphogenesis. DCX becomes enriched only on straight microtubules in advancing growth cones with approximately 120 DCX molecules bound per micrometer of growth cone microtubule. At a similar saturation, microtubule-bound DCX molecules begin to impede lysosome transport, and thus can potentially control growth cone organelle entry. In addition, by comparing control, DCX-mEmerald and knockout DCX -/Y i 3 Neurons, we find that DCX stabilizes microtubules in the growth cone peripheral domain by reducing the microtubule catastrophe frequency and the depolymerization rate. DCX -/Y i 3 Neuron morphogenesis was inhibited in soft microenvironments that mimic the viscoelasticity of brain tissue and DCX -/Y neurites failed to grow toward brain-derived neurotrophic factor (BDNF) gradients. Together with high resolution traction force microscopy data, we propose a model in which DCX-decorated, rigid growth cone microtubules provide intracellular mechanical resistance to actomyosin generated contractile forces in soft physiological environments in which weak and transient adhesion-mediated forces in the growth cone periphery may be insufficient for productive growth cone advance. These data provide a new mechanistic understanding of how DCX mutations cause lissencephaly-spectrum brain malformations by impacting growth cone dynamics during neuron morphogenesis in physiological environments.

2.
Proc Natl Acad Sci U S A ; 120(41): e2308941120, 2023 10 10.
Article En | MEDLINE | ID: mdl-37782785

Impaired lymphatic drainage and lymphedema are major morbidities whose mechanisms have remained obscure. To study lymphatic drainage and its impairment, we engineered a microfluidic culture model of lymphatic vessels draining interstitial fluid. This lymphatic drainage-on-chip revealed that inflammatory cytokines that are known to disrupt blood vessel junctions instead tightened lymphatic cell-cell junctions and impeded lymphatic drainage. This opposing response was further demonstrated when inhibition of rho-associated protein kinase (ROCK) was found to normalize fluid drainage under cytokine challenge by simultaneously loosening lymphatic junctions and tightening blood vessel junctions. Studies also revealed a previously undescribed shift in ROCK isoforms in lymphatic endothelial cells, wherein a ROCK2/junctional adhesion molecule-A (JAM-A) complex emerges that is responsible for the cytokine-induced lymphatic junction zippering. To validate these in vitro findings, we further demonstrated in a genetic mouse model that lymphatic-specific knockout of ROCK2 reversed lymphedema in vivo. These studies provide a unique platform to generate interstitial fluid pressure and measure the drainage of interstitial fluid into lymphatics and reveal a previously unappreciated ROCK2-mediated mechanism in regulating lymphatic drainage.


Junctional Adhesion Molecule A , Lymphatic Vessels , Lymphedema , rho-Associated Kinases , Animals , Mice , Biomimetics , Cytokines/metabolism , Endothelial Cells/metabolism , Intercellular Junctions , Junctional Adhesion Molecule A/metabolism , Lymphatic Vessels/metabolism , Lymphedema/genetics , Lymphedema/metabolism , rho-Associated Kinases/metabolism
3.
J Cell Sci ; 136(19)2023 10 01.
Article En | MEDLINE | ID: mdl-37795818

Emergent cell behaviors that drive tissue morphogenesis are the integrated product of instructions from gene regulatory networks, mechanics and signals from the local tissue microenvironment. How these discrete inputs intersect to coordinate diverse morphogenic events is a critical area of interest. Organ-on-chip technology has revolutionized the ability to construct and manipulate miniaturized human tissues with organotypic three-dimensional architectures in vitro. Applications of organ-on-chip platforms have increasingly transitioned from proof-of-concept tissue engineering to discovery biology, furthering our understanding of molecular and mechanical mechanisms that operate across biological scales to orchestrate tissue morphogenesis. Here, we provide the biological framework to harness organ-on-chip systems to study tissue morphogenesis, and we highlight recent examples where organ-on-chips and associated microphysiological systems have enabled new mechanistic insight in diverse morphogenic settings. We further highlight the use of organ-on-chip platforms as emerging test beds for cell and developmental biology.


Microphysiological Systems , Tissue Engineering , Humans , Tissue Engineering/methods , Morphogenesis
4.
J Cell Biol ; 222(12)2023 12 04.
Article En | MEDLINE | ID: mdl-37796194

Notch receptors control tissue morphogenic processes that involve coordinated changes in cell architecture and gene expression, but how a single receptor can produce these diverse biological outputs is unclear. Here, we employ a 3D model of a human ductal epithelium to reveal tissue morphogenic defects result from loss of Notch1, but not Notch1 transcriptional signaling. Instead, defects in duct morphogenesis are driven by dysregulated epithelial cell architecture and mitogenic signaling which result from the loss of a transcription-independent, Notch1 cortical signaling mechanism that ultimately functions to stabilize adherens junctions and cortical actin. We identify that Notch1 localization and cortical signaling are tied to apical-basal cell restructuring and discover that a Notch1-FAM83H interaction underlies control of epithelial adherens junctions and cortical actin. Together, these results offer new insights into Notch1 signaling and regulation and advance a paradigm in which transcriptional and cell adhesive programs might be coordinated by a single receptor.


Actins , Adherens Junctions , Cell Adhesion , Receptor, Notch1 , Humans , Adherens Junctions/genetics , Cell Proliferation , Epithelial Cells , Proteins , Receptor, Notch1/genetics , Signal Transduction
5.
Nat Commun ; 14(1): 3561, 2023 06 15.
Article En | MEDLINE | ID: mdl-37322009

Intratumor heterogeneity associates with poor patient outcome. Stromal stiffening also accompanies cancer. Whether cancers demonstrate stiffness heterogeneity, and if this is linked to tumor cell heterogeneity remains unclear. We developed a method to measure the stiffness heterogeneity in human breast tumors that quantifies the stromal stiffness each cell experiences and permits visual registration with biomarkers of tumor progression. We present Spatially Transformed Inferential Force Map (STIFMap) which exploits computer vision to precisely automate atomic force microscopy (AFM) indentation combined with a trained convolutional neural network to predict stromal elasticity with micron-resolution using collagen morphological features and ground truth AFM data. We registered high-elasticity regions within human breast tumors colocalizing with markers of mechanical activation and an epithelial-to-mesenchymal transition (EMT). The findings highlight the utility of STIFMap to assess mechanical heterogeneity of human tumors across length scales from single cells to whole tissues and implicates stromal stiffness in tumor cell heterogeneity.


Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Mechanical Phenomena , Elasticity , Collagen , Neural Networks, Computer , Microscopy, Atomic Force/methods
6.
Cell Syst ; 14(4): 252-257, 2023 04 19.
Article En | MEDLINE | ID: mdl-37080161

Collective cell behavior contributes to all stages of cancer progression. Understanding how collective behavior emerges through cell-cell interactions and decision-making will advance our understanding of cancer biology and provide new therapeutic approaches. Here, we summarize an interdisciplinary discussion on multicellular behavior in cancer, draw lessons from other scientific disciplines, and identify future directions.


Mass Behavior , Neoplasms , Humans , Communication
7.
Sci Adv ; 9(7): eade8939, 2023 02 15.
Article En | MEDLINE | ID: mdl-36791204

Somatic activating mutations of PIK3CA are associated with development of vascular malformations (VMs). Here, we describe a microfluidic model of PIK3CA-driven VMs consisting of human umbilical vein endothelial cells expressing PIK3CA activating mutations embedded in three-dimensional hydrogels. We observed enlarged, irregular vessel phenotypes and the formation of cyst-like structures consistent with clinical signatures and not previously observed in cell culture models. Pathologic morphologies occurred concomitant with up-regulation of Rac1/p21-activated kinase (PAK), mitogen-activated protein kinase cascades (MEK/ERK), and mammalian target of rapamycin (mTORC1/2) signaling networks. We observed differential effects between alpelisib, a PIK3CA inhibitor, and rapamycin, an mTORC1 inhibitor, in mitigating matrix degradation and network topology. While both were effective in preventing vessel enlargement, rapamycin failed to reduce MEK/ERK and mTORC2 activity and resulted in hyperbranching, while inhibiting PAK, MEK1/2, and mTORC1/2 mitigates abnormal growth and vascular dilation. Collectively, these findings demonstrate an in vitro platform for VMs and establish a role of dysregulated Rac1/PAK and mTORC1/2 signaling in PIK3CA-driven VMs.


TOR Serine-Threonine Kinases , Vascular Malformations , Humans , TOR Serine-Threonine Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Sirolimus/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Vascular Malformations/metabolism , rac1 GTP-Binding Protein/metabolism
8.
bioRxiv ; 2023 Jan 23.
Article En | MEDLINE | ID: mdl-36747830

Notch receptors control tissue morphogenic processes that involve coordinated changes in cell architecture and gene expression, but how a single receptor can produce these diverse biological outputs is unclear. Here we employ a 3D organotypic model of a ductal epithelium to reveal tissue morphogenic defects result from loss of Notch1, but not Notch1 transcriptional signaling. Instead, defects in duct morphogenesis are driven by dysregulated epithelial cell architecture and mitogenic signaling which result from loss of a transcription-independent Notch1 cortical signaling mechanism that ultimately functions to stabilize adherens junctions and cortical actin. We identify that Notch1 localization and cortical signaling are tied to apical-basal cell restructuring and discover a Notch1-FAM83H interaction underlies stabilization of adherens junctions and cortical actin. Together, these results offer new insights into Notch1 signaling and regulation, and advance a paradigm in which transcriptional and cell adhesive programs might be coordinated by a single receptor.

9.
Nat Cell Biol ; 24(12): 1739-1753, 2022 12.
Article En | MEDLINE | ID: mdl-36456828

Adherens junctions (AJs) create spatially, chemically and mechanically discrete microdomains at cellular interfaces. Here, using a mechanogenetic platform that generates artificial AJs with controlled protein localization, clustering and mechanical loading, we find that AJs also organize proteolytic hotspots for γ-secretase with a spatially regulated substrate selectivity that is critical in the processing of Notch and other transmembrane proteins. Membrane microdomains outside of AJs exclusively organize Notch ligand-receptor engagement (LRE microdomains) to initiate receptor activation. Conversely, membrane microdomains within AJs exclusively serve to coordinate regulated intramembrane proteolysis (RIP microdomains). They do so by concentrating γ-secretase and primed receptors while excluding full-length Notch. AJs induce these functionally distinct microdomains by means of lipid-dependent γ-secretase recruitment and size-dependent protein segregation. By excluding full-length Notch from RIP microdomains, AJs prevent inappropriate enzyme-substrate interactions and suppress spurious Notch activation. Ligand-induced ectodomain shedding eliminates size-dependent segregation, releasing Notch to translocate into AJs for processing by γ-secretase. This mechanism directs radial differentiation of ventricular zone-neural progenitor cells in vivo and more broadly regulates the proteolysis of other large cell-surface receptors such as amyloid precursor protein. These findings suggest an unprecedented role of AJs in creating size-selective spatial switches that choreograph γ-secretase processing of multiple transmembrane proteins regulating development, homeostasis and disease.


Amyloid Precursor Protein Secretases , Amyloid Precursor Protein Secretases/genetics , Ligands
10.
Am J Physiol Cell Physiol ; 323(5): C1333-C1344, 2022 11 01.
Article En | MEDLINE | ID: mdl-36121131

Tumor metastasis via the circulation requires crossing the vascular barrier twice: first, during intravasation when tumor cells disseminate from the primary site through proximal vasculature, and second, during extravasation, when tumor cells exit the circulation to form distant metastatic seeds. During these key metastatic events, chemomechanical signaling between tumor cells and endothelial cells elicits reciprocal changes in cell morphology and behavior that are necessary to breach the vessel wall. Existing experimental systems have provided a limited understanding of the diverse mechanisms underlying tumor-endothelial interactions during intravasation and extravasation. Recent advances in microphysiological systems have revolutionized the ability to generate miniaturized human tissues with tailored three-dimensional architectures, physiological cell interfaces, and precise chemical and physical microenvironments. By doing so, microphysiological systems enable experimental access to complex morphogenic processes associated with human tumor progression with unprecedented resolution and biological control. Here, we discuss recent examples in which microphysiological systems have been leveraged to reveal new mechanistic insight into cellular and molecular control systems operating at the tumor-endothelial interface during intravasation and extravasation.


Endothelial Cells , Neoplasms , Humans , Endothelial Cells/pathology , Neoplasms/pathology , Endothelium , Signal Transduction , Neoplasm Metastasis , Tumor Microenvironment
11.
Dev Cell ; 56(6): 826-841.e4, 2021 03 22.
Article En | MEDLINE | ID: mdl-33705692

We describe a cellular contractile mechanism employed by fibroblasts and mesenchymal cancer cells to migrate in 3D collagen gels. During 3D spreading, fibroblasts strongly deform the matrix. They protrude, polarize, and initiate migration in the direction of highest extracellular matrix (ECM) deformation (prestrain). This prestrain is maintained through anterior cellular contractions behind the leading edge prior to protrusion, coordinating a distinct 3D migration cycle that varies between cell types. Myosin IIA is required for strain polarization, generating anterior contractions, and maintaining prestrain for efficient directional cell migration. Local matrix severing disrupts the matrix prestrain, suppressing directional protrusion. We show that epithelial cancer and endothelial cells rarely demonstrate the sustained prestrain or anterior contractions. We propose that mesenchymal cells sense ECM stiffness in 3D and generate their own matrix prestrain. This requires myosin IIA to generate polarized periodic anterior contractions for maintaining a 3D migration cycle.


Breast Neoplasms/pathology , Cell Movement , Extracellular Matrix/physiology , Fibroblasts/physiology , Mesoderm/physiology , Nonmuscle Myosin Type IIA/metabolism , Stress, Mechanical , Breast Neoplasms/metabolism , Cell Adhesion , Cells, Cultured , Female , Fibroblasts/cytology , Humans , Mesoderm/cytology
12.
Nat Commun ; 11(1): 3377, 2020 07 06.
Article En | MEDLINE | ID: mdl-32632100

The mammary gland is a highly vascularized tissue capable of expansion and regression during development and disease. To enable mechanistic insight into the coordinated morphogenic crosstalk between the epithelium and vasculature, we introduce a 3D microfluidic platform that juxtaposes a human mammary duct in proximity to a perfused endothelial vessel. Both compartments recapitulate stable architectural features of native tissue and the ability to undergo distinct forms of branching morphogenesis. Modeling HER2/ERBB2 amplification or activating PIK3CA(H1047R) mutation each produces ductal changes observed in invasive progression, yet with striking morphogenic and behavioral differences. Interestingly, PI3KαH1047R ducts also elicit increased permeability and structural disorganization of the endothelium, and we identify the distinct secretion of IL-6 as the paracrine cause of PI3KαH1047R-associated vascular dysfunction. These results demonstrate the functionality of a model system that facilitates the dissection of 3D morphogenic behaviors and bidirectional signaling between mammary epithelium and endothelium during homeostasis and pathogenesis.


Mammary Glands, Human/metabolism , Morphogenesis/genetics , Mutation , Paracrine Communication/genetics , Biomimetics/methods , Cell Line , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Endothelium, Vascular/growth & development , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mammary Glands, Human/blood supply , Mammary Glands, Human/growth & development , Phenotype , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
13.
Nat Protoc ; 14(5): 1425-1454, 2019 05.
Article En | MEDLINE | ID: mdl-30953042

The vascular endothelium forms the inner lining of blood vessels and actively regulates vascular permeability in response to chemical and physical stimuli. Understanding the molecular pathways and mechanisms that regulate the permeability of blood vessels is of critical importance for developing therapies for cardiovascular dysfunction and disease. Recently, we developed a novel microfluidic human engineered microvessel (hEMV) platform to enable controlled blood flow through a human endothelial lumen within a physiologic 3D extracellular matrix (ECM) into which pericytes and other stromal cells can be introduced to recapitulate tissue-specific microvascular physiology. This protocol describes how to design and fabricate the silicon hEMV device master molds (takes ~1 week) and elastomeric substrates (takes 3 d); how to seed, culture, and apply calibrated fluid shear stress to hEMVs (takes 1-7 d); and how to assess vascular barrier function (takes 1 d) and perform immunofluorescence imaging (takes 3 d).


Capillary Permeability/physiology , Cell Culture Techniques/instrumentation , Microfluidic Analytical Techniques/instrumentation , Microvessels , Models, Cardiovascular , Tissue Engineering/instrumentation , Cells, Cultured , Endothelial Cells/cytology , Equipment Design , Humans , Microvessels/cytology , Microvessels/physiology , Stromal Cells/cytology
14.
Dev Cell ; 44(1): 87-96.e5, 2018 01 08.
Article En | MEDLINE | ID: mdl-29316444

Truncating mutations in the sarcomere protein titin cause dilated cardiomyopathy due to sarcomere insufficiency. However, it remains mechanistically unclear how these mutations decrease sarcomere content in cardiomyocytes. Utilizing human induced pluripotent stem cell-derived cardiomyocytes, CRISPR/Cas9, and live microscopy, we characterize the fundamental mechanisms of human cardiac sarcomere formation. We observe that sarcomerogenesis initiates at protocostameres, sites of cell-extracellular matrix adhesion, where nucleation and centripetal assembly of α-actinin-2-containing fibers provide a template for the fusion of Z-disk precursors, Z bodies, and subsequent striation. We identify that ß-cardiac myosin-titin-protocostamere form an essential mechanical connection that transmits forces required to direct α-actinin-2 centripetal fiber assembly and sarcomere formation. Titin propagates diastolic traction stresses from ß-cardiac myosin, but not α-cardiac myosin or non-muscle myosin II, to protocostameres during sarcomerogenesis. Ablating protocostameres or decoupling titin from protocostameres abolishes sarcomere assembly. Together these results identify the mechanical and molecular components critical for human cardiac sarcomerogenesis.


Actinin/metabolism , Cell-Matrix Junctions/physiology , Connectin/metabolism , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Sarcomeres/physiology , Ventricular Myosins/metabolism , Actinin/genetics , Adolescent , Adult , Cells, Cultured , Connectin/genetics , Humans , Induced Pluripotent Stem Cells/physiology , Male , Middle Aged , Myocytes, Cardiac/physiology , Ventricular Myosins/genetics
15.
APL Bioeng ; 2(4): 046107, 2018 Dec.
Article En | MEDLINE | ID: mdl-31069329

Physical features of the extracellular matrix (ECM) heavily influence cell migration strategies and efficiency. Migration in and on fibrous ECMs is of significant physiologic importance, but limitations in the ability to experimentally define the diameter, density, and alignment of native ECMs in vitro have hampered our understanding of how these properties affect this basic cell function. Here, we designed a high-throughput in vitro platform that models fibrous ECM as collections of lines of cell-adhesive fibronectin on a flat surface to eliminate effects of dimensionality and topography. Using a microcontact printing approach to orthogonally vary line alignment, density, and size, we determined each factor's individual influence on NIH3T3 fibroblast migration. High content imaging and statistical analyses revealed that ECM alignment is the most critical parameter in influencing cell morphology, polarization, and migratory behavior. Specifically, increasing ECM alignment led cells to adopt an elongated uniaxial morphology and migrate with enhanced speed and persistence. Intriguingly, migration speeds were tightly correlated with the organization of focal adhesions, where cells with the most aligned adhesions migrated fastest. Highly organized focal adhesions and associated actin stress fibers appeared to define the number and location of protrusive fronts, suggesting that ECM alignment influences active Rac1 localization. Utilizing a novel microcontact-printing approach that lacks confounding influences of substrate dimensionality, mechanics, or differences in the adhesive area, this work highlights the effect of ECM alignment on orchestrating the cytoskeletal machinery that governs directed uniaxial cell migration.

16.
Nature ; 552(7684): 258-262, 2017 12 14.
Article En | MEDLINE | ID: mdl-29160307

The vascular barrier that separates blood from tissues is actively regulated by the endothelium and is essential for transport, inflammation, and haemostasis. Haemodynamic shear stress plays a critical role in maintaining endothelial barrier function, but how this occurs remains unknown. Here we use an engineered organotypic model of perfused microvessels to show that activation of the transmembrane receptor NOTCH1 directly regulates vascular barrier function through a non-canonical, transcription-independent signalling mechanism that drives assembly of adherens junctions, and confirm these findings in mouse models. Shear stress triggers DLL4-dependent proteolytic activation of NOTCH1 to expose the transmembrane domain of NOTCH1. This domain mediates establishment of the endothelial barrier; expression of the transmembrane domain of NOTCH1 is sufficient to rescue defects in barrier function induced by knockout of NOTCH1. The transmembrane domain restores barrier function by catalysing the formation of a receptor complex in the plasma membrane consisting of vascular endothelial cadherin, the transmembrane protein tyrosine phosphatase LAR, and the RAC1 guanidine-exchange factor TRIO. This complex activates RAC1 to drive assembly of adherens junctions and establish barrier function. Canonical transcriptional signalling via Notch is highly conserved in metazoans and is required for many processes in vascular development, including arterial-venous differentiation, angiogenesis and remodelling. We establish the existence of a non-canonical cortical NOTCH1 signalling pathway that regulates vascular barrier function, and thus provide a mechanism by which a single receptor might link transcriptional programs with adhesive and cytoskeletal remodelling.


Adherens Junctions/metabolism , Endothelium, Vascular/metabolism , Multiprotein Complexes/metabolism , Receptor, Notch1/metabolism , Adherens Junctions/enzymology , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line , Endothelium, Vascular/enzymology , Female , Guanine Nucleotide Exchange Factors/metabolism , Humans , Mice , Multiprotein Complexes/chemistry , Phosphoproteins/metabolism , Protein Domains , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptor, Notch1/chemistry , Signal Transduction , rac GTP-Binding Proteins/metabolism
17.
Curr Opin Cell Biol ; 42: 73-79, 2016 10.
Article En | MEDLINE | ID: mdl-27209346

The effects of hemodynamic and interstitial mechanical forces on endothelial biology in vivo have been appreciated for over half a century, regulating vessel network development, homeostatic function, and progression of vascular disease. Investigations using cultures of endothelial cells on two-dimensional (2D) substrates have elucidated important mechanisms by which microenvironmental stresses are sensed and transduced into chemical signaling responses. However recent studies in vivo and in three-dimensional (3D) in vitro models of vascular beds have enabled the investigation of forces and cellular behaviors previously not possible in traditional 2D culture systems. These studies support a developing paradigm that the 3D chemo-mechanical architecture of the vascular niche impacts how endothelial cells both sense and respond to microenvironmental forces. We present evolving concepts in endothelial force sensing and mechanical signaling and highlight recent insights gained from in vivo and 3D in vitro vascular models.


Blood Vessels/cytology , Blood Vessels/physiology , Imaging, Three-Dimensional , Mechanotransduction, Cellular , Animals , Biomechanical Phenomena , Endothelial Cells/physiology , Humans
18.
Small GTPases ; 6(1): 16-9, 2015.
Article En | MEDLINE | ID: mdl-25862162

Investigating cell migration in 3D settings has revealed that specific extracellular matrix environments require differential activities of the Rho GTPases for efficient migration. However, it is largely unknown how the activities of specific Rho GTPases are modulated to direct cell migration in response to different extracellular matrix cues. We have recently reported that extracellular matrix-dependent regulation of a specific Rho GEF is a fundamental mechanism governing cell migration in different microenvironments, providing a direct mechanism for extracellular matrix-specific regulation of Rho GTPase activity directing cell motility. We discovered that the Rho GEF ßPix has a unique function during cell migration in fibrillar collagen environments by restraining RhoA signaling through a conserved signaling axis involving Cdc42 and the Rho GAP srGAP1. In this Commentary, we expand upon this new pathway and discuss potential mechanotransductive and therapeutic applications. Additionally, we speculate on a generalized role for Rho GEFs and GAPs in providing localized, context-dependent responses to the cellular microenvironment during cell migration and other cellular processes.


Extracellular Matrix/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Cell Movement , Collagen/metabolism , Extracellular Matrix/genetics , Fibronectins/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Rho Guanine Nucleotide Exchange Factors/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
19.
Nat Cell Biol ; 16(9): 909-17, 2014 Sep.
Article En | MEDLINE | ID: mdl-25150978

Rho-family GTPases govern distinct types of cell migration on different extracellular matrix proteins in tissue culture or three-dimensional (3D) matrices. We searched for mechanisms selectively regulating 3D cell migration in different matrix environments and discovered a form of Cdc42-RhoA crosstalk governing cell migration through a specific pair of GTPase activator and inhibitor molecules. We first identified ßPix, a guanine nucleotide exchange factor (GEF), as a specific regulator of migration in 3D collagen using an affinity-precipitation-based GEF screen. Knockdown of ßPix specifically blocks cell migration in fibrillar collagen microenvironments, leading to hyperactive cellular protrusion accompanied by increased collagen matrix contraction. Live FRET imaging and RNAi knockdown linked this ßPix knockdown phenotype to loss of polarized Cdc42 but not Rac1 activity, accompanied by enhanced, de-localized RhoA activity. Mechanistically, collagen phospho-regulates ßPix, leading to its association with srGAP1, a GTPase-activating protein (GAP), needed to suppress RhoA activity. Our results reveal a matrix-specific pathway controlling migration involving a GEF-GAP interaction of ßPix with srGAP1 that is critical for maintaining suppressive crosstalk between Cdc42 and RhoA during 3D collagen migration.


Cell Movement , Collagen/metabolism , GTPase-Activating Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors/physiology , rhoA GTP-Binding Protein/metabolism , Cell Line, Tumor , Cell Shape , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Integrin alpha2beta1/metabolism , Protein Phosphatase 2/metabolism , Protein Processing, Post-Translational , Protein Transport , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
20.
Curr Opin Cell Biol ; 25(5): 642-9, 2013 Oct.
Article En | MEDLINE | ID: mdl-23850350

The importance of cell migration for both normal physiological functions and disease processes has been clear for the past 50 years. Although investigations of two-dimensional (2D) migration in regular tissue culture have elucidated many important molecular mechanisms, recent evidence suggests that cell migration depends profoundly on the dimensionality of the extracellular matrix (ECM). Here we review a number of evolving concepts revealed when cell migration is examined in different dimensions.


Cell Movement , Animals , Cell Physiological Phenomena , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Humans , Signal Transduction
...