Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Mar 13.
Article in English | MEDLINE | ID: mdl-36993714

ABSTRACT

Somatic cell fate is an outcome set by the activities of specific transcription factors and the chromatin landscape and is maintained by gene silencing of alternate cell fates through physical interactions with the nuclear scaffold. Here, we evaluate the role of the nuclear scaffold as a guardian of cell fate in human fibroblasts by comparing the effects of transient loss (knockdown) and mutation (progeria) of functional Lamin A/C, a core component of the nuclear scaffold. We observed that Lamin A/C deficiency or mutation disrupts nuclear morphology, heterochromatin levels, and increases access to DNA in lamina-associated domains. Changes in Lamin A/C were also found to impact the mechanical properties of the nucleus when measured by a microfluidic cellular squeezing device. We also show that transient loss of Lamin A/C accelerates the kinetics of cellular reprogramming to pluripotency through opening of previously silenced heterochromatin domains while genetic mutation of Lamin A/C into progerin induces a senescent phenotype that inhibits the induction of reprogramming genes. Our results highlight the physical role of the nuclear scaffold in safeguarding cellular fate.

2.
PLoS One ; 18(3): e0282563, 2023.
Article in English | MEDLINE | ID: mdl-36888609

ABSTRACT

Droplet-based microfluidic devices have been used to achieve homogeneous cell encapsulation, but cells sediment in a solution, leading to heterogeneous products. In this technical note, we describe automated and programmable agitation device to maintain colloidal suspensions of cells. We demonstrate that the agitation device can be interfaced with a syringe pump for microfluidic applications. Agitation profiles of the device were predictable and corresponded to device settings. The device maintains the concentration of cells in an alginate solution over time without implicating cell viability. This device replaces manual agitation, and hence is suitable for applications that require slow perfusion for a longer period of time in a scalable manner.


Subject(s)
Microfluidics , Syringes , Perfusion , Cell Survival , Magnetic Phenomena
3.
Adv Sci (Weinh) ; 10(3): e2206014, 2023 01.
Article in English | MEDLINE | ID: mdl-36453581

ABSTRACT

Various signals in tissue microenvironments are often unevenly distributed around cells. Cellular responses to asymmetric cell-matrix adhesion in a 3D space remain generally unclear and are to be studied at the single-cell resolution. Here, the authors developed a droplet-based microfluidic approach to manufacture a pure population of single cells in a microscale layer of compartmentalized 3D hydrogel matrices with a tunable spatial presentation of ligands at the subcellular level. Cells elongate with an asymmetric presentation of the integrin adhesion ligand Arg-Gly-Asp (RGD), while cells expand isotropically with a symmetric presentation of RGD. Membrane tension is higher on the side of single cells interacting with RGD than on the side without RGD. Finite element analysis shows that a non-uniform isotropic cell volume expansion model is sufficient to recapitulate the experimental results. At a longer timescale, asymmetric ligand presentation commits mesenchymal stem cells to the osteogenic lineage. Cdc42 is an essential mediator of cell polarization and lineage specification in response to asymmetric cell-matrix adhesion. This study highlights the utility of precisely controlling 3D ligand presentation around single cells to direct cell polarity for regenerative engineering and medicine.


Subject(s)
Cell Encapsulation , Cell Polarity , Ligands , Hydrogels , Oligopeptides
4.
Proc Natl Acad Sci U S A ; 119(15): e2121098119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35377803

ABSTRACT

The pathogenesis of lung fibrosis involves hyperactivation of innate and adaptive immune pathways that release inflammatory cytokines and growth factors such as tumor growth factor (TGF)ß1 and induce aberrant extracellular matrix protein production. During the genesis of pulmonary fibrosis, resident alveolar macrophages are replaced by a population of newly arrived monocyte-derived interstitial macrophages that subsequently transition into alveolar macrophages (Mo-AMs). These transitioning cells initiate fibrosis by releasing profibrotic cytokines and remodeling the matrix. Here, we describe a strategy for leveraging the up-regulation of the mannose receptor CD206 in interstitial macrophages and Mo-AM to treat lung fibrosis. We engineered mannosylated albumin nanoparticles, which were found to be internalized by fibrogenic CD206+ monocyte derived macrophages (Mo-Macs). Mannosylated albumin nanoparticles incorporating TGFß1 small-interfering RNA (siRNA) targeted the profibrotic subpopulation of CD206+ macrophages and prevented lung fibrosis. The findings point to the potential utility of mannosylated albumin nanoparticles in delivering TGFß-siRNA into CD206+ profibrotic macrophages as an antilung fibrosis strategy.


Subject(s)
Lymphotoxin-alpha , Macrophages, Alveolar , Nanoparticles , Pulmonary Fibrosis , RNA, Small Interfering , Animals , Bleomycin/pharmacology , Disease Models, Animal , Lymphotoxin-alpha/genetics , Macrophages, Alveolar/immunology , Mannose Receptor , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/therapy , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
5.
Nat Biomed Eng ; 6(1): 54-66, 2022 01.
Article in English | MEDLINE | ID: mdl-34083763

ABSTRACT

The precise understanding and control of microenvironmental cues could be used to optimize the efficacy of cell therapeutics. Here, we show that mesenchymal stromal cells (MSCs) singly coated with a soft conformal gel presenting defined chemomechanical cues promote matrix remodelling by secreting soluble interstitial collagenases in response to the presence of tumour necrosis factor alpha (TNF-α). In mice with fibrotic lung injury, treatment with the coated MSCs maintained normal collagen levels, fibre density and microelasticity in lung tissue, and the continuous presentation of recombinant TNF-α in the gel facilitated the reversal of aberrant tissue remodelling by the cells when inflammation subsided in the host. Gel coatings with predefined chemomechanical cues could be used to tailor cells with specific mechanisms of action for desired therapeutic outcomes.


Subject(s)
Choristoma , Mesenchymal Stem Cells , Tissue Engineering , Animals , Chemotaxis , Choristoma/pathology , Collagen , Gels , Mice , Tissue Engineering/methods , Tumor Necrosis Factor-alpha
6.
ACS Nano ; 15(11): 17439-17452, 2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34677951

ABSTRACT

Extracellular vesicles (EVs) are cell-secreted particles with broad potential to treat tissue injuries by delivering cargo to program target cells. However, improving the yield of functional EVs on a per cell basis remains challenging due to an incomplete understanding of how microenvironmental cues regulate EV secretion at the nanoscale. We show that mesenchymal stromal cells (MSCs) seeded on engineered hydrogels that mimic the elasticity of soft tissues with a lower integrin ligand density secrete ∼10-fold more EVs per cell than MSCs seeded on a rigid plastic substrate, without compromising their therapeutic activity or cargo to resolve acute lung injury in mice. Mechanistically, intracellular CD63+ multivesicular bodies (MVBs) transport faster within MSCs on softer hydrogels, leading to an increased frequency of MVB fusion with the plasma membrane to secrete more EVs. Actin-related protein 2/3 complex but not myosin-II limits MVB transport and EV secretion from MSCs on hydrogels. The results provide a rational basis for biomaterial design to improve EV secretion while maintaining their functionality.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Animals , Mice , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Cell Communication , Biological Transport , Hydrogels/pharmacology , Hydrogels/metabolism
7.
Acta Biomater ; 133: 126-138, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34365041

ABSTRACT

Hydrogels have been used to design synthetic matrices that capture salient features of matrix microenvironments to study and control cellular functions. Recent advances in understanding of both extracellular matrix biology and biomaterial design have shown that biophysical cues are powerful mediators of cell biology, especially that of mesenchymal stromal cells (MSCs). MSCs have been tested in many clinical trials because of their ability to modulate immune cells in different pathological conditions. While roles of biophysical cues in MSC biology have been studied in the context of multilineage differentiation, their significance in regulating immunomodulatory functions of MSCs is just beginning to be elucidated. This review first describes design principles behind how biophysical cues in native microenvironments influence the ability of MSCs to regulate immune cell production and functions. We will then discuss how biophysical cues can be leveraged to optimize cell isolation, priming, and delivery, which can help improve the success of MSC therapy for immunomodulation. Finally, a perspective is presented on how implementing biophysical cues in MSC potency assay can be important in predicting clinical outcomes. STATEMENT OF SIGNIFICANCE: Stromal cells of mesenchymal origin are known to direct immune cell functions in vivo by secreting paracrine mediators. This property has been leveraged in developing mesenchymal stromal cell (MSC)-based therapeutics by adoptive transfer to treat immunological rejection and tissue injuries, which have been tested in over one thousand clinical trials to date, but with mixed success. Advances in biomaterial design have enabled precise control of biophysical cues based on how stromal cells interact with the extracellular matrix in microenvironments in situ. Investigators have begun to use this approach to understand how different matrix biophysical parameters, such as fiber orientation, porosity, dimensionality, and viscoelasticity impact stromal cell-mediated immunomodulation. The insights gained from this effort can potentially be used to precisely define the microenvironmental cues for isolation, priming, and delivery of MSCs, which can be tailored based on different disease indications for optimal therapeutic outcomes.


Subject(s)
Mesenchymal Stem Cells , Cell Differentiation , Cues , Extracellular Matrix , Immunomodulation
8.
Adv Sci (Weinh) ; 7(20): 2001066, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33101850

ABSTRACT

Advances in engineered hydrogels reveal how cells sense and respond to 3D biophysical cues. However, most studies rely on interfacing a population of cells in a tissue-scale bulk hydrogel, an approach that overlooks the heterogeneity of local matrix deposition around individual cells. A droplet microfluidic technique to deposit a defined amount of 3D hydrogel matrices around single cells independently of material composition, elasticity, and stress relaxation times is developed. Mesenchymal stem cells (MSCs) undergo isotropic volume expansion more rapidly in thinner gels that present an Arg-Gly-Asp integrin ligand. Mathematical modeling and experiments show that MSCs experience higher membrane tension as they expand in thinner gels. Furthermore, thinner gels facilitate osteogenic differentiation of MSCs. By modulating ion channels, it is shown that isotropic volume expansion of single cells predicts intracellular tension and stem cell fate. The results suggest the utility of precise microscale gel deposition to control single cell functions.

9.
Adv Biosyst ; 4(11): e2000012, 2020 11.
Article in English | MEDLINE | ID: mdl-33053274

ABSTRACT

The extracellular matrix varies considerably in mechanical properties at the microscale. It remains unclear how cells respond to these properties, in part, due to lack of tools to create precisely defined microenvironments in a discrete manner. Here, freeform stereolithography is leveraged to control the placement and elastic modulus of individual hydrogel microposts that serve as discrete matrix signals to interface with cells. Mesenchymal stromal cells (MSCs) located in the interstitial spaces between microposts above a base layer are analyzed. Cell volume is higher when MSCs interact with more microposts. MSCs show higher strain energy when they interact simultaneously with 4-kPa and 20-kPa microposts than with mechanically homogeneous micropost arrays. MSCs are sensitive to pharmacological inhibition of Rho-associated protein kinase in 4-kPa arrays, but resistant when presented together with 20-kPa arrays. Yes-associated protein (YAP) activity increases with higher cell volume and elastic modulus of microposts. Surprisingly, YAP activity becomes less variable with higher cell volume and decreases with higher average force and strain energy per post when MSCs interact with both 4-kPa and 20-kPa microposts simultaneously. Together, these results describe a material system for systematically investigating how the placement and intrinsic properties of discrete matrix signals impact cell volume and mechanotransduction.


Subject(s)
Cell Size , Cytological Techniques/instrumentation , Hydrogels/chemistry , Mechanotransduction, Cellular/physiology , Cells, Cultured , Elastic Modulus , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Stereolithography
10.
Sci Adv ; 6(15): eaaw0158, 2020 04.
Article in English | MEDLINE | ID: mdl-32284989

ABSTRACT

Mesenchymal stromal cells (MSCs) modulate immune cells to ameliorate multiple inflammatory pathologies. Biophysical signals that regulate this process are poorly defined. By engineering hydrogels with tunable biophysical parameters relevant to bone marrow where MSCs naturally reside, we show that soft extracellular matrix maximizes the ability of MSCs to produce paracrine factors that have been implicated in monocyte production and chemotaxis upon inflammatory stimulation by tumor necrosis factor-α (TNFα). Soft matrix increases clustering of TNF receptors, thereby enhancing NF-κB activation and downstream gene expression. Actin polymerization and lipid rafts, but not myosin-II contractility, regulate mechanosensitive activation of MSCs by TNFα. We functionally demonstrate that human MSCs primed with TNFα in soft matrix enhance production of human monocytes in marrow of xenografted mice and increase trafficking of monocytes via CCL2. The results suggest the importance of biophysical signaling in tuning inflammatory activation of stromal cells to control the innate immune system.


Subject(s)
Extracellular Matrix/metabolism , Inflammation/metabolism , Mesenchymal Stem Cells/metabolism , Monocytes/metabolism , Actins/metabolism , Animals , Biomarkers , Cell Movement/immunology , Cells, Cultured , Chemotaxis , Cytokines/metabolism , Humans , Inflammation/etiology , Inflammation/pathology , Inflammation Mediators/metabolism , Lipid Metabolism , Mice , Monocytes/immunology , NF-kappa B/metabolism , Protein Binding , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
11.
Proc Natl Acad Sci U S A ; 116(31): 15392-15397, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31311862

ABSTRACT

Mesenchymal stem cell (MSC) therapies demonstrate particular promise in ameliorating diseases of immune dysregulation but are hampered by short in vivo cell persistence and inconsistencies in phenotype. Here, we demonstrate that biomaterial encapsulation into alginate using a microfluidic device could substantially increase in vivo MSC persistence after intravenous (i.v.) injection. A combination of cell cluster formation and subsequent cross-linking with polylysine led to an increase in injected MSC half-life by more than an order of magnitude. These modifications extended persistence even in the presence of innate and adaptive immunity-mediated clearance. Licensing of encapsulated MSCs with inflammatory cytokine pretransplantation increased expression of immunomodulatory-associated genes, and licensed encapsulates promoted repopulation of recipient blood and bone marrow with allogeneic donor cells after sublethal irradiation by a ∼2-fold increase. The ability of microgel encapsulation to sustain MSC survival and increase overall immunomodulatory capacity may be applicable for improving MSC therapies in general.


Subject(s)
Cell Encapsulation , Immunomodulation , Mesenchymal Stem Cells/cytology , Alginates/chemistry , Animals , Cells, Cultured , Gene Expression Regulation , Hematopoiesis/genetics , Immunity , Immunomodulation/genetics , Mice, Inbred BALB C , Time Factors , Transplantation, Homologous
12.
APL Bioeng ; 2(3): 031802, 2018 Sep.
Article in English | MEDLINE | ID: mdl-31069313

ABSTRACT

It is increasingly appreciated that physical forces play important roles in cancer biology, in terms of progression, invasiveness, and drug resistance. Clinical progress in treating hematological malignancy and in developing cancer immunotherapy highlights the role of the hematopoietic system as a key model in devising new therapeutic strategies against cancer. Understanding mechanobiology of the hematopoietic system in the context of cancer will thus yield valuable fundamental insights that can information about novel cancer therapeutics. In this perspective, biophysical insights related to blood cancer are defined and detailed. The interactions with immune cells relevant to immunotherapy against cancer are considered and expounded, followed by speculation of potential regulatory roles of mesenchymal stromal cells (MSCs) in this complex network. Finally, a perspective is presented as to how insights from these complex interactions between matrices, blood cancer cells, immune cells, and MSCs can be leveraged to influence and engineer the treatment of blood cancers in the clinic.

13.
J Biomech ; 55: 113-120, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28314562

ABSTRACT

Deep tissue pressure ulcers, a serious clinical challenge originating in the muscle layer, are hardly detectable at the beginning. The challenge apparently occurs in aged subjects more frequently. As the ulcer propagates to the skin surface, it becomes very difficult to manage and can lead to fatal complications. Preventive measures are thus highly desirable. Although the complex pathological mechanisms have not been fully understood, prolonged and excessive physical challenges and oxidative stress are believed to be involved in the ulcer development. Previous reports have demonstrated that oxidative stress could compromise the mechanical properties of muscle cells, making them easier to be damaged when physical challenges are introduced. In this study, we used senescence accelerated (SAMP8) mice and its control breed (SAMR1) to examine the protective effects of intermittent vibration on aged and control muscle tissues during prolonged epidermal compression under 100mmHg for 6h. Results showed that an application of 35Hz, 0.25g intermittent vibration during compression decreased the compression-induced muscle breakdown in SAMP8 mice, as indicated histologically in terms of number of interstitial nuclei. The fact that no significant difference in muscle damage could be established in the corresponding groups in SAMR1 mice suggests that SAMR1 mice could better accommodate the compression insult than SAMP8 mice. Compression-induced oxidative damage was successfully curbed using intermittent vibration in SAMP8 mice, as indicated by 8-OHdG. A possible explanation is that the anti-oxidative defense could be maintained with intermittent vibration during compression. This was supported by the expression level of PGC-1-alpha, catalase, Gpx-1 and SOD1. Our data suggested intermittent vibration could serve as a preventive measure for deep tissue ulcer, particularly in aged subjects.


Subject(s)
Aging/metabolism , Muscles/metabolism , Oxidative Stress , Stress, Mechanical , Vibration , Animals , Mice , Muscles/cytology , Muscles/physiology , Pressure
14.
Ann Biomed Eng ; 45(4): 1083-1092, 2017 04.
Article in English | MEDLINE | ID: mdl-27650939

ABSTRACT

High oxidative stress can occur during ischemic reperfusion and chronic inflammation. It has been hypothesized that such oxidative challenges could contribute to clinical risks such as deep tissue pressure ulcers. Skeletal muscles can be challenged by inflammation-induced or reperfusion-induced oxidative stress. Oxidative stress reportedly can lower the compressive damage threshold of skeletal muscles cells, causing actin filament depolymerization, and reduce membrane sealing ability. Skeletal muscles thus become easier to be damaged by mechanical loading under prolonged oxidative exposure. In this study, we investigated the preventive effect of poloxamer 188 (P188) on skeletal muscle cells against extrinsic oxidative challenges (H2O2). It was found that with 1 mM P188 pre-treatment for 1 h, skeletal muscle cells could maintain their compressive damage threshold. The actin polymerization dynamics largely remained stable in term of the expression of cofilin, thymosin beta 4 and profilin. Laser photoporation demonstrated that membrane sealing ability was preserved even as the cells were challenged by H2O2. These findings suggest that P188 pre-treatment can help skeletal muscle cells retain their normal mechanical integrity in oxidative environments, adding a potential clinical use of P188 against the combined challenge of mechanical-oxidative stresses. Such effect may help to prevent deep tissue ulcer development.


Subject(s)
Gene Expression Regulation/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/biosynthesis , Oxidative Stress/drug effects , Poloxamer/pharmacology , Animals , Cell Line , Compressive Strength/drug effects , Hydrogen Peroxide/pharmacology , Mice , Muscle Fibers, Skeletal/pathology
15.
Ann Biomed Eng ; 43(5): 1178-88, 2015 May.
Article in English | MEDLINE | ID: mdl-25371376

ABSTRACT

Skeletal muscles often experience oxidative stress in anaerobic metabolism and ischemia-reperfusion. This paper reports how oxidative stress affects the stiffness of cultured murine myotubes and their actin filaments polymerization dynamics. H2O2 was applied as an extrinsic oxidant to C2C12 myotubes. Atomic force microscopy results showed that short exposures to H2O2 apparently increased the stiffness of myotubes, but that long exposures made the cells softer. The turning point seemed to take place somewhere between 1 and 2 h of H2O2 exposure. We found that the stiffness change was probably due to actin filaments being favored for depolymerization after prolong H2O2 treatments, especially when the exposure duration exceeded 1 h and the exposure concentration reached 1.0 mM. Such depolymerization effect was associated with the down-regulation of thymosin beta 4, as well as the up-regulation of both cofilin2 and profilin1 after prolong H2O2 treatments.


Subject(s)
Actins/metabolism , Hydrogen Peroxide/pharmacology , Muscle Fibers, Skeletal/drug effects , Actins/genetics , Animals , Cell Line , Cofilin 2/genetics , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , Polymerization , Profilins/genetics , Reactive Oxygen Species/metabolism , Thymosin/genetics
16.
PLoS One ; 8(3): e59477, 2013.
Article in English | MEDLINE | ID: mdl-23555679

ABSTRACT

PML protein plays important roles in regulating cellular homeostasis. It forms PML nuclear bodies (PML-NBs) that act like nuclear relay stations and participate in many cellular functions. In this study, we have examined the proteome of mouse embryonic fibroblasts (MEFs) derived from normal (PML(+/+)) and PML knockout (PML(-/-)) mice. The aim was to identify proteins that were differentially expressed when MEFs were incapable of producing PML. Using comparative proteomics, total protein were extracted from PML(-/-) and PML(+/+) MEFs, resolved by two dimensional electrophoresis (2-DE) gels and the differentially expressed proteins identified by LC-ESI-MS/MS. Nine proteins (PML, NDRG1, CACYBP, CFL1, RSU1, TRIO, CTRO, ANXA4 and UBE2M) were determined to be down-regulated in PML(-/-) MEFs. In contrast, ten proteins (CIAPIN1, FAM50A, SUMO2 HSPB1 NSFL1C, PCBP2, YWHAG, STMN1, TPD52L2 and PDAP1) were found up-regulated. Many of these differentially expressed proteins play crucial roles in cell adhesion, migration, morphology and cytokinesis. The protein profiles explain why PML(-/-) and PML(+/+) MEFs were morphologically different. In addition, we demonstrated PML(-/-) MEFs were less adhesive, proliferated more extensively and migrated significantly slower than PML(+/+) MEFs. NDRG1, a protein that was down-regulated in PML(-/-) MEFs, was selected for further investigation. We determined that silencing NDRG1expression in PML(+/+) MEFs increased cell proliferation and inhibited PML expression. Since NDRG expression was suppressed in PML(-/-) MEFs, this may explain why these cells proliferate more extensively than PML(+/+) MEFs. Furthermore, silencing NDRG1expression also impaired TGF-ß1 signaling by inhibiting SMAD3 phosphorylation.


Subject(s)
Cell Movement , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Adhesion , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Chemotaxis , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Deletion , Gene Expression Regulation , Gene Silencing , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Proteome , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics , Transforming Growth Factor beta1/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...