Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell ; 168(6): 1135-1148.e12, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28262351

ABSTRACT

Investigation of host-environment interactions in the gut would benefit from a culture system that maintained tissue architecture yet allowed tight experimental control. We devised a microfabricated organ culture system that viably preserves the normal multicellular composition of the mouse intestine, with luminal flow to control perturbations (e.g., microbes, drugs). It enables studying short-term responses of diverse gut components (immune, neuronal, etc.). We focused on the early response to bacteria that induce either Th17 or RORg+ T-regulatory (Treg) cells in vivo. Transcriptional responses partially reproduced in vivo signatures, but these microbes elicited diametrically opposite changes in expression of a neuronal-specific gene set, notably nociceptive neuropeptides. We demonstrated activation of sensory neurons by microbes, correlating with RORg+ Treg induction. Colonic RORg+ Treg frequencies increased in mice lacking TAC1 neuropeptide precursor and decreased in capsaicin-diet fed mice. Thus, differential engagement of the enteric nervous system may partake in bifurcating pro- or anti-inflammatory responses to microbes.


Subject(s)
Clostridium/growth & development , Intestines/growth & development , Intestines/microbiology , Organ Culture Techniques , Animals , Clostridium/classification , Clostridium/physiology , Intestines/cytology , Mice , Symbiosis
2.
Cell ; 158(4): 822-832, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25126787

ABSTRACT

Molecular motors in cells typically produce highly directed motion; however, the aggregate, incoherent effect of all active processes also creates randomly fluctuating forces, which drive diffusive-like, nonthermal motion. Here, we introduce force-spectrum-microscopy (FSM) to directly quantify random forces within the cytoplasm of cells and thereby probe stochastic motor activity. This technique combines measurements of the random motion of probe particles with independent micromechanical measurements of the cytoplasm to quantify the spectrum of force fluctuations. Using FSM, we show that force fluctuations substantially enhance intracellular movement of small and large components. The fluctuations are three times larger in malignant cells than in their benign counterparts. We further demonstrate that vimentin acts globally to anchor organelles against randomly fluctuating forces in the cytoplasm, with no effect on their magnitude. Thus, FSM has broad applications for understanding the cytoplasm and its intracellular processes in relation to cell physiology in healthy and diseased states.


Subject(s)
Cytoplasm/chemistry , Microscopy, Atomic Force/methods , Animals , Biomechanical Phenomena , Embryo, Mammalian/cytology , Fibroblasts/chemistry , Mice , Proteins/chemistry , Vimentin/chemistry
3.
Proc Natl Acad Sci U S A ; 120(28): e2301285120, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37399392

ABSTRACT

Yes-associated protein (YAP) is a key mechanotransduction protein in diverse physiological and pathological processes; however, a ubiquitous YAP activity regulatory mechanism in living cells has remained elusive. Here, we show that YAP nuclear translocation is highly dynamic during cell movement and is driven by nuclear compression arising from cell contractile work. We resolve the mechanistic role of cytoskeletal contractility in nuclear compression by manipulation of nuclear mechanics. Disrupting the linker of nucleoskeleton and cytoskeleton complex reduces nuclear compression for a given contractility and correspondingly decreases YAP localization. Conversely, decreasing nuclear stiffness via silencing of lamin A/C increases nuclear compression and YAP nuclear localization. Finally, using osmotic pressure, we demonstrated that nuclear compression even without active myosin or filamentous actin regulates YAP localization. The relationship between nuclear compression and YAP localization captures a universal mechanism for YAP regulation with broad implications in health and biology.


Subject(s)
Adaptor Proteins, Signal Transducing , YAP-Signaling Proteins , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Mechanotransduction, Cellular , Transcription Factors/genetics , Transcription Factors/metabolism , Cytoskeleton/metabolism
4.
Biophys J ; 123(10): 1222-1239, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38605521

ABSTRACT

Controlling mesenchymal stem cell (MSC) differentiation remains a critical challenge in MSCs' therapeutic application. Numerous biophysical and mechanical stimuli influence stem cell fate; however, their relative efficacy and specificity in mechanically directed differentiation remain unclear. Yes-associated protein (YAP) is one key mechanosensitive protein that controls MSC differentiation. Previous studies have related nuclear mechanics with YAP activity, but we still lack an understanding of what nuclear deformation specifically regulates YAP and its relationship with mechanical stimuli. Here, we report that maximum nuclear curvature is the most precise biophysical determinant for YAP mechanotransduction-mediated MSC differentiation and is a relevant parameter for stem cell-based therapies. We employed traction force microscopy and confocal microscopy to characterize the causal relationships between contractility and nuclear deformation in regulating YAP activity in MSCs. We observed that an increase in contractility compresses nuclei anisotropically, whereby the degree of asymmetric compression increased the bending curvature of the nuclear membrane. We then examined membrane curvature and tension using thin micropatterned adhesive substrate lines and an FRET-based tension sensor, revealing the direct role of curvature in YAP activity driven by both active and passive nuclear import. Finally, we employed micropatterned lines to control nuclear curvature and precisely direct MSC differentiation. This work illustrates that nuclear curvature subsumes other biophysical aspects to control YAP-mediated differentiation in MSCs and may provide a deterministic solution to some of the challenges in mesenchymal stem cell therapies.


Subject(s)
Cell Differentiation , Cell Nucleus , Mesenchymal Stem Cells , YAP-Signaling Proteins , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Cell Nucleus/metabolism , YAP-Signaling Proteins/metabolism , Humans , Adaptor Proteins, Signal Transducing/metabolism , Transcription Factors/metabolism , Mechanotransduction, Cellular , Protein Transport
5.
Biophys J ; 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39033326

ABSTRACT

Traction-force microscopy (TFM) has emerged as a widely used standard methodology to measure cell-generated traction forces and determine their role in regulating cell behavior. While TFM platforms have enabled many discoveries, their implementation remains limited due to complex experimental procedures, specialized substrates, and the ill-posed inverse problem whereby low-magnitude high-frequency noise in the displacement field severely contaminates the resulting traction measurements. Here, we introduce deep morphology traction microscopy (DeepMorphoTM), a deep-learning alternative to conventional TFM approaches. DeepMorphoTM first infers cell-induced substrate displacement solely from a sequence of cell shapes and subsequently computes cellular traction forces, thus avoiding the requirement of a specialized fiduciarily marked deformable substrate or force-free reference image. Rather, this technique drastically simplifies the overall experimental methodology, imaging, and analysis needed to conduct cell-contractility measurements. We demonstrate that DeepMorphoTM quantitatively matches conventional TFM results while offering stability against the biological variability in cell contractility for a given cell shape. Without high-frequency noise in the inferred displacement, DeepMorphoTM also resolves the ill-posedness of traction computation, increasing the consistency and accuracy of traction analysis. We demonstrate the accurate extrapolation across several cell types and substrate materials, suggesting robustness of the methodology. Accordingly, we present DeepMorphoTM as a capable yet simpler alternative to conventional TFM for characterizing cellular contractility in two dimensions.

6.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Article in English | MEDLINE | ID: mdl-34887356

ABSTRACT

Membrane invagination and vesicle formation are key steps in endocytosis and cellular trafficking. Here, we show that endocytic coat proteins with prion-like domains (PLDs) form hemispherical puncta in the budding yeast, Saccharomyces cerevisiae These puncta have the hallmarks of biomolecular condensates and organize proteins at the membrane for actin-dependent endocytosis. They also enable membrane remodeling to drive actin-independent endocytosis. The puncta, which we refer to as endocytic condensates, form and dissolve reversibly in response to changes in temperature and solution conditions. We find that endocytic condensates are organized around dynamic protein-protein interaction networks, which involve interactions among PLDs with high glutamine contents. The endocytic coat protein Sla1 is at the hub of the protein-protein interaction network. Using active rheology, we inferred the material properties of endocytic condensates. These experiments show that endocytic condensates are akin to viscoelastic materials. We use these characterizations to estimate the interfacial tension between endocytic condensates and their surroundings. We then adapt the physics of contact mechanics, specifically modifications of Hertz theory, to develop a quantitative framework for describing how interfacial tensions among condensates, the membrane, and the cytosol can deform the plasma membrane to enable actin-independent endocytosis.


Subject(s)
Cytoskeletal Proteins/metabolism , Endocytosis/physiology , Prions/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Cell Membrane , Cytoskeletal Proteins/genetics , Cytosol/physiology , Gene Expression Regulation, Fungal , Glutamine/chemistry , Mechanotransduction, Cellular , Protein Conformation , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Viscoelastic Substances
7.
J Cell Sci ; 134(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34028539

ABSTRACT

While diverse cellular components have been identified as mechanotransduction elements, the deformation of the nucleus itself is a critical mechanosensory mechanism, implying that nuclear stiffness is essential in determining responses to intracellular and extracellular stresses. Although the nuclear membrane protein lamin A/C is known to contribute to nuclear stiffness, bulk moduli of nuclei have not been reported for various levels of lamin A/C. Here, we measure the nuclear bulk moduli as a function of lamin A/C expression and applied osmotic stress, revealing a linear dependence within the range of 2-4 MPa. We also find that the nuclear compression is anisotropic, with the vertical axis of the nucleus being more compliant than the minor and major axes in the substrate plane. We then related the spatial distribution of lamin A/C with submicron 3D nuclear envelope deformation, revealing that local areas of the nuclear envelope with higher density of lamin A/C have correspondingly lower local deformations. These findings describe the complex dispersion of nuclear deformations as a function of lamin A/C expression and distribution, implicating a lamin A/C role in mechanotransduction. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Lamin Type A , Mechanotransduction, Cellular , Cell Nucleus/metabolism , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Nuclear Envelope/metabolism
8.
Biophys J ; 121(4): 629-643, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34999131

ABSTRACT

Tissue and cell mechanics are crucial factors in maintaining homeostasis and in development, with aberrant mechanics contributing to many diseases. During the epithelial-to-mesenchymal transition (EMT), a highly conserved cellular program in organismal development and cancer metastasis, cells gain the ability to detach from their original location and autonomously migrate. While a great deal of biochemical and biophysical changes at the single-cell level have been revealed, how the physical properties of multicellular assemblies change during EMT, and how this may affect disease progression, is unknown. Here we introduce cell monolayer deformation microscopy (CMDM), a new methodology to measure the planar mechanical properties of cell monolayers by locally applying strain and measuring their resistance to deformation. We employ this new method to characterize epithelial multicellular mechanics at early and late stages of EMT, finding the epithelial monolayers to be relatively compliant, ductile, and mechanically homogeneous. By comparison, the transformed mesenchymal monolayers, while much stiffer, were also more brittle, mechanically heterogeneous, displayed more viscoelastic creep, and showed sharp yield points at significantly lower strains. Here, CMDM measurements identify specific biophysical functional states of EMT and offer insight into how cell aggregates fragment under mechanical stress. This mechanical fingerprinting of multicellular assemblies using new quantitative metrics may also offer new diagnostic applications in healthcare to characterize multicellular mechanical changes in disease.


Subject(s)
Epithelial-Mesenchymal Transition , Microscopy , Stress, Mechanical
9.
Biochem Biophys Res Commun ; 586: 27-33, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34823219

ABSTRACT

While it is now well-established that substrate stiffness regulates vascular endothelial growth factor-A (VEGF-A) mediated signaling and functions, causal mechanisms remain poorly understood. Here, we report an underlying role for the PI3K/Akt/mTOR signaling pathway. This pathway is activated on stiffer substrates, is amplified by VEGF-A stimulation, and correlates with enhanced endothelial cell (EC) proliferation, contraction, pro-angiogenic secretion, and capillary-like tube formation. In the settings of advanced age-related macular degeneration, characterized by EC and retinal pigment epithelial (RPE)-mediated angiogenesis, these data implicate substrate stiffness as a novel causative mechanism and Akt/mTOR inhibition as a novel therapeutic pathway.


Subject(s)
Endothelial Cells/metabolism , Mechanotransduction, Cellular/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Retinal Pigment Epithelium/metabolism , TOR Serine-Threonine Kinases/genetics , Vascular Endothelial Growth Factor A/genetics , Biomechanical Phenomena , Cell Line , Cell Movement , Cell Proliferation , Elasticity , Endothelial Cells/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Models, Biological , Neovascularization, Pathologic/genetics , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , Retinal Pigment Epithelium/cytology , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L323-L330, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31774304

ABSTRACT

In asthma, acute bronchospasm is driven by contractile forces of airway smooth muscle (ASM). These forces can be imaged in the cultured ASM cell or assessed in the muscle strip and the tracheal/bronchial ring, but in each case, the ASM is studied in isolation from the native airway milieu. Here, we introduce a novel platform called tissue traction microscopy (TTM) to measure ASM contractile force within porcine and human precision-cut lung slices (PCLS). Compared with the conventional measurements of lumen area changes in PCLS, TTM measurements of ASM force changes are 1) more sensitive to bronchoconstrictor stimuli, 2) less variable across airways, and 3) provide spatial information. Notably, within every human airway, TTM measurements revealed local regions of high ASM contraction that we call "stress hotspots". As an acute response to cyclic stretch, these hotspots promptly decreased but eventually recovered in magnitude, spatial location, and orientation, consistent with local ASM fluidization and resolidification. By enabling direct and precise measurements of ASM force, TTM should accelerate preclinical studies of airway reactivity.


Subject(s)
Lung/physiology , Microscopy , Muscle Contraction/physiology , Traction , Animals , Animals, Newborn , Biomechanical Phenomena , Bronchoconstriction/physiology , Humans , Muscle, Smooth/physiology , Stress, Mechanical , Swine
11.
Proc Natl Acad Sci U S A ; 114(39): E8147-E8154, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28900011

ABSTRACT

Biological complexity presents challenges for understanding natural phenomenon and engineering new technologies, particularly in systems with molecular heterogeneity. Such complexity is present in myosin motor protein systems, and computational modeling is essential for determining how collective myosin interactions produce emergent system behavior. We develop a computational approach for altering myosin isoform parameters and their collective organization, and support predictions with in vitro experiments of motility assays with α-actinins as molecular force sensors. The computational approach models variations in single myosin molecular structure, system organization, and force stimuli to predict system behavior for filament velocity, energy consumption, and robustness. Robustness is the range of forces where a filament is expected to have continuous velocity and depends on used myosin system energy. Myosin systems are shown to have highly nonlinear behavior across force conditions that may be exploited at a systems level by combining slow and fast myosin isoforms heterogeneously. Results suggest some heterogeneous systems have lower energy use near stall conditions and greater energy consumption when unloaded, therefore promoting robustness. These heterogeneous system capabilities are unique in comparison with homogenous systems and potentially advantageous for high performance bionanotechnologies. Findings open doors at the intersections of mechanics and biology, particularly for understanding and treating myosin-related diseases and developing approaches for motor molecule-based technologies.


Subject(s)
Computational Biology , Models, Theoretical , Muscle Contraction/physiology , Muscles/physiology , Myosins/metabolism , Actins/metabolism , Actomyosin/metabolism , Biomechanical Phenomena/physiology , Humans
12.
Lab Invest ; 99(1): 138-145, 2019 01.
Article in English | MEDLINE | ID: mdl-30310180

ABSTRACT

Vascular leakage, protein exudation, and edema formation are events commonly triggered by inflammation and facilitated by gaps that form between adjacent endothelial cells (ECs) of the vasculature. In such paracellular gap formation, the role of EC contraction is widely implicated, and even therapeutically targeted. However, related measurement approaches remain slow, tedious, and complex to perform. Here, we have developed a multiplexed, high-throughput screen to simultaneously quantify paracellular gaps, EC contractile forces, and to visualize F-actin stress fibers, and VE-cadherin. As proof-of-principle, we examined barrier-protective mechanisms of the Rho-associated kinase inhibitor, Y-27632, and the canonical agonist of the Tie2 receptor, Angiopoietin-1 (Angpt-1). Y-27632 reduced EC contraction and actin stress fiber formation, whereas Angpt-1 did not. Yet both agents reduced thrombin-, LPS-, and TNFα-induced paracellular gap formation. This unexpected result suggests that Angpt-1 can achieve barrier defense without reducing EC contraction, a mechanism that has not been previously described. This insight was enabled by the multiplex nature of the force-based platform. The high-throughput format we describe should accelerate both mechanistic studies and the screening of pharmacological modulators of endothelial barrier function.


Subject(s)
Actin Cytoskeleton/physiology , Endothelial Cells/physiology , High-Throughput Screening Assays/methods , Amides , Angiopoietin-1 , Antigens, CD/metabolism , Cadherins/metabolism , Endothelium, Vascular/physiology , Humans , Intercellular Junctions/physiology , Microscopy, Fluorescence , Permeability , Primary Cell Culture , Pyridines
13.
Biophys J ; 114(9): 2194-2199, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29742412

ABSTRACT

Actomyosin contractility is an essential element of many aspects of cellular biology and manifests as traction forces that cells exert on their surroundings. The central role of these forces makes them a novel principal therapeutic target in diverse diseases. This requires accurate and higher-capacity measurements of traction forces; however, existing methods are largely low throughput, limiting their utility in broader applications. To address this need, we employ Fourier-transform traction force microscopy in a parallelized 96-well format, which we refer to as contractile force screening. Critically, rather than the frequently employed hydrogel polyacrylamide, we fabricate these plates using polydimethylsiloxane rubber. Key to this approach is that the polydimethylsiloxane used is very compliant, with a lower-bound Young's modulus of ∼0.4 kPa. We subdivide these monolithic substrates spatially into biochemically independent wells, creating a uniform multiwell platform for traction force screening. We demonstrate the utility and versatility of this platform by quantifying the compound and dose-dependent contractility responses of human airway smooth muscle cells and retinal pigment epithelial cells. By directly quantifying the endpoint of therapeutic intent, airway-smooth-muscle contractile force, this approach fills an important methodological void in current screening approaches for bronchodilator drug discovery, and, more generally, in measuring contractile response for a broad range of cell types and pathologies.


Subject(s)
Dimethylpolysiloxanes/chemistry , Elastomers/chemistry , Mechanical Phenomena , Nylons/chemistry , Myocytes, Smooth Muscle/cytology
14.
Proc Natl Acad Sci U S A ; 112(21): 6619-24, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25918384

ABSTRACT

The actin cytoskeleton is a key element of cell structure and movement whose properties are determined by a host of accessory proteins. Actin cross-linking proteins create a connected network from individual actin filaments, and though the mechanical effects of cross-linker binding affinity on actin networks have been investigated in reconstituted systems, their impact on cellular forces is unknown. Here we show that the binding affinity of the actin cross-linker α-actinin 4 (ACTN4) in cells modulates cytoplasmic mobility, cellular movement, and traction forces. Using fluorescence recovery after photobleaching, we show that an ACTN4 mutation that causes human kidney disease roughly triples the wild-type binding affinity of ACTN4 to F-actin in cells, increasing the dissociation time from 29 ± 13 to 86 ± 29 s. This increased affinity creates a less dynamic cytoplasm, as demonstrated by reduced intracellular microsphere movement, and an approximate halving of cell speed. Surprisingly, these less motile cells generate larger forces. Using traction force microscopy, we show that increased binding affinity of ACTN4 increases the average contractile stress (from 1.8 ± 0.7 to 4.7 ± 0.5 kPa), and the average strain energy (0.4 ± 0.2 to 2.1 ± 0.4 pJ). We speculate that these changes may be explained by an increased solid-like nature of the cytoskeleton, where myosin activity is more partitioned into tension and less is dissipated through filament sliding. These findings demonstrate the impact of cross-linker point mutations on cell dynamics and forces, and suggest mechanisms by which such physical defects lead to human disease.


Subject(s)
Actinin/physiology , Actinin/chemistry , Actinin/genetics , Actins/metabolism , Amino Acid Substitution , Binding Sites/genetics , Biomechanical Phenomena , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Cross-Linking Reagents , Fluorescence Recovery After Photobleaching , HeLa Cells , Humans , Kinetics , Microscopy, Confocal , Models, Biological , Mutagenesis, Site-Directed , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Blood ; 125(5): 860-8, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25411426

ABSTRACT

Bone marrow megakaryocytes produce platelets by extending long cytoplasmic protrusions, designated proplatelets, into sinusoidal blood vessels. Although microtubules are known to regulate platelet production, the underlying mechanism of proplatelet elongation has yet to be resolved. Here we report that proplatelet formation is a process that can be divided into repetitive phases (extension, pause, and retraction), as revealed by differential interference contrast and fluorescence loss after photoconversion time-lapse microscopy. Furthermore, we show that microtubule sliding drives proplatelet elongation and is dependent on cytoplasmic dynein under static and physiological shear stress by using fluorescence recovery after photobleaching in proplatelets with fluorescence-tagged ß1-tubulin. A refined understanding of the specific mechanisms regulating platelet production will yield strategies to treat patients with thrombocythemia or thrombocytopenia.


Subject(s)
Blood Platelets/metabolism , Cytoplasmic Dyneins/metabolism , Megakaryocytes/metabolism , Microtubules/metabolism , Tubulin/metabolism , Animals , Blood Platelets/cytology , Cell Differentiation , Cytoplasm/metabolism , Cytoplasmic Dyneins/genetics , Fluorescence Recovery After Photobleaching , Gene Expression , Mechanotransduction, Cellular , Megakaryocytes/cytology , Mice , Microscopy, Interference , Microtubules/chemistry , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stress, Mechanical , Thrombopoiesis/genetics , Tubulin/genetics
16.
Biochim Biophys Acta ; 1853(11 Pt B): 3126-31, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26119327

ABSTRACT

During development neuronal cells traverse substantial distances across the developing tissue. In the mature organism, however, they are bound to the confines of the nervous system. Likewise metastatic cancer cells have the potential to establish auxiliary tumor sites in remote tissues or entirely different organs. The epithelial-mesenchymal transition is the transformation of proliferative cancer cells into a highly invasive state, which facilitates the crossing of tissue boundaries and migration across various environments. This review contributes a first look into the parallels and contrasts between physical aspects of neuronal and metastatic cancer cells.


Subject(s)
Cell Proliferation , Epithelial-Mesenchymal Transition , Neoplasms , Neurons , Animals , Humans , Neoplasms/chemistry , Neoplasms/metabolism , Neurons/chemistry , Neurons/metabolism
17.
Blood ; 124(12): 1857-67, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25606631

ABSTRACT

Platelet transfusions total >2.17 million apheresis-equivalent units per year in the United States and are derived entirely from human donors, despite clinically significant immunogenicity, associated risk of sepsis, and inventory shortages due to high demand and 5-day shelf life. To take advantage of known physiological drivers of thrombopoiesis, we have developed a microfluidic human platelet bioreactor that recapitulates bone marrow stiffness, extracellular matrix composition,micro-channel size, hemodynamic vascular shear stress, and endothelial cell contacts, and it supports high-resolution live-cell microscopy and quantification of platelet production. Physiological shear stresses triggered proplatelet initiation, reproduced ex vivo bone marrow proplatelet production, and generated functional platelets. Modeling human bone marrow composition and hemodynamics in vitro obviates risks associated with platelet procurement and storage to help meet growing transfusion needs.


Subject(s)
Bioreactors , Blood Platelets , Microfluidic Analytical Techniques , Animals , Biomimetic Materials , Blood Platelets/cytology , Blood Platelets/physiology , Equipment Design , Humans , Megakaryocytes/cytology , Megakaryocytes/physiology , Mice , Models, Biological , Platelet Transfusion , Thrombopoiesis
18.
Nucleus ; 15(1): 2374854, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38951951

ABSTRACT

The nucleus not only is a repository for DNA but also a center of cellular and nuclear mechanotransduction. From nuclear deformation to the interplay between mechanosensing components and genetic control, the nucleus is poised at the nexus of mechanical forces and cellular function. Understanding the stresses acting on the nucleus, its mechanical properties, and their effects on gene expression is therefore crucial to appreciate its mechanosensitive function. In this review, we examine many elements of nuclear mechanotransduction, and discuss the repercussions on the health of cells and states of illness. By describing the processes that underlie nuclear mechanosensation and analyzing its effects on gene regulation, the review endeavors to open new avenues for studying nuclear mechanics in physiology and diseases.


Subject(s)
Cell Nucleus , Mechanotransduction, Cellular , Humans , Cell Nucleus/metabolism , Animals , Gene Expression Regulation
19.
Biophys J ; 105(7): 1562-8, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24094397

ABSTRACT

The mechanical properties of a cell determine many aspects of its behavior, and these mechanics are largely determined by the cytoskeleton. Although the contribution of actin filaments and microtubules to the mechanics of cells has been investigated in great detail, relatively little is known about the contribution of the third major cytoskeletal component, intermediate filaments (IFs). To determine the role of vimentin IF (VIF) in modulating intracellular and cortical mechanics, we carried out studies using mouse embryonic fibroblasts (mEFs) derived from wild-type or vimentin(-/-) mice. The VIFs contribute little to cortical stiffness but are critical for regulating intracellular mechanics. Active microrheology measurements using optical tweezers in living cells reveal that the presence of VIFs doubles the value of the cytoplasmic shear modulus to ∼10 Pa. The higher levels of cytoplasmic stiffness appear to stabilize organelles in the cell, as measured by tracking endogenous vesicle movement. These studies show that VIFs both increase the mechanical integrity of cells and localize intracellular components.


Subject(s)
Cytoplasm/metabolism , Cytoskeleton/metabolism , Fibroblasts/metabolism , Vimentin/metabolism , Animals , Cytoplasmic Vesicles/metabolism , Cytoskeleton/ultrastructure , Fibroblasts/ultrastructure , Mice , Mice, Knockout , Optical Tweezers , Protein Transport , Rheology , Shear Strength , Vimentin/genetics
20.
bioRxiv ; 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37503095

ABSTRACT

The role of morphogenetic forces in cell fate specification is an area of intense interest. Our prior studies suggested that the development of high cell-cell tension in human embryonic stem cells (hESC) colonies permits the Src-mediated phosphorylation of junctional ß-catenin that accelerates its release to potentiate Wnt-dependent signaling critical for initiating mesoderm specification. Using an ectopically expressed nonphosphorylatable mutant of ß-catenin (Y654F), we now provide direct evidence that impeding tension-dependent Src-mediated ß-catenin phosphorylation impedes the expression of Brachyury (T) and the epithelial-to-mesenchymal transition (EMT) necessary for mesoderm specification. Addition of exogenous Wnt3a or inhibiting GSK3ß activity rescued mesoderm expression, emphasizing the importance of force dependent Wnt signaling in regulating mechanomorphogenesis. Our work provides a framework for understanding tension-dependent ß-catenin/Wnt signaling in the self-organization of tissues during developmental processes including gastrulation.

SELECTION OF CITATIONS
SEARCH DETAIL