Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 35(2): 102154, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38511173

ABSTRACT

Solitary fibrous tumor (SFT) is a rare, non-hereditary soft tissue sarcoma thought to originate from fibroblastic mesenchymal stem cells. The etiology of SFT is thought to be due to an environmental intrachromosomal gene fusion between NGFI-A-binding protein 2 (NAB2) and signal transducer and activator protein 6 (STAT6) genes on chromosome 12, wherein the activation domain of STAT6 is fused with the DNA-binding domain of NAB2 resulting in the oncogenesis of SFT. All NAB2-STAT6 fusion variations discovered in SFTs contain the C-terminal of STAT6 transcript, and thus can serve as target site for antisense oligonucleotides (ASOs)-based therapies. Indeed, our in vitro studies show the STAT6 3' untranslated region (UTR)-targeting ASO (ASO 993523) was able to reduce expression of NAB2-STAT6 fusion transcripts in multiple SFT cell models with high efficiency (half-maximal inhibitory concentration: 116-300 nM). Encouragingly, in vivo treatment of SFT patient-derived xenograft mouse models with ASO 993523 resulted in acceptable tolerability profiles, reduced expression of NAB2-STAT6 fusion transcripts in xenograft tissues (21.9%), and, importantly, reduced tumor growth (32.4% decrease in tumor volume compared with the untreated control). Taken together, our study established ASO 993523 as a potential agent for the treatment of SFTs.

2.
Cancers (Basel) ; 15(12)2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37370737

ABSTRACT

Solitary fibrous tumor (SFT) is a rare soft-tissue sarcoma. This nonhereditary cancer is the result of an environmental intrachromosomal gene fusion between NAB2 and STAT6 on chromosome 12, which fuses the activation domain of STAT6 with the repression domain of NAB2. Currently there is not an approved chemotherapy regimen for SFTs. The best response on available pharmaceuticals is a partial response or stable disease for several months. The purpose of this study is to investigate the potential of RNA-based therapies for the treatment of SFTs. Specifically, in vitro SFT cell models were engineered to harbor the characteristic NAB2-STAT6 fusion using the CRISPR/SpCas9 system. Cell migration as well as multiple cancer-related signaling pathways were increased in the engineered cells as compared to the fusion-absent parent cells. The SFT cell models were then used for evaluating the targeting efficacies of NAB2-STAT6 fusion-specific antisense oligonucleotides (ASOs) and CRISPR/CasRx systems. Our results showed that fusion specific ASO treatments caused a 58% reduction in expression of fusion transcripts and a 22% reduction in cell proliferation after 72 h in vitro. Similarly, the AAV2-mediated CRISPR/CasRx system led to a 59% reduction in fusion transcript expressions in vitro, and a 55% reduction in xenograft growth after 29 days ex vivo.

4.
J Control Release ; 357: 511-530, 2023 05.
Article in English | MEDLINE | ID: mdl-37040842

ABSTRACT

Many diseases affecting the central nervous system (CNS) are deadly but less understood, leading to impaired mental and motor capabilities and poor patient prospects. Gene therapy is a promising therapeutic modality for correcting many genetic disorders, expanding in breadth and scope with further advances. This review summarizes the candidate CNS disorders for gene therapy, mechanisms of gene therapy, and recent clinical advances and limitations of gene therapy in CNS disorders. We highlight that improving delivery across CNS barriers, safety, monitoring techniques, and multiplexing therapies are predominant factors in advancing long-term outcomes from gene therapy.


Subject(s)
Central Nervous System Diseases , Genetic Vectors , Humans , Genetic Vectors/genetics , Central Nervous System , Genetic Therapy/methods , Central Nervous System Diseases/genetics , Central Nervous System Diseases/therapy
5.
Front Med Technol ; 4: 1008540, 2022.
Article in English | MEDLINE | ID: mdl-36523426

ABSTRACT

Despite advancements in early detection and treatment, atherosclerosis remains the leading cause of death across all cardiovascular diseases (CVD). Biomechanical analysis of atherosclerotic lesions has the potential to reveal biomechanically instable or rupture-prone regions. Treatment decisions rarely consider the biomechanics of the stenosed lesion due in-part to difficulties in obtaining this information in a clinical setting. Previous 3D FEA approaches have incompletely incorporated the complex curvature of arterial geometry, material heterogeneity, and use of patient-specific data. To address these limitations and clinical need, herein we present a user-friendly fully automated program to reconstruct and simulate the wall mechanics of patient-specific atherosclerotic coronary arteries. The program enables 3D reconstruction from patient-specific data with heterogenous tissue assignment and complex arterial curvature. Eleven arteries with coronary artery disease (CAD) underwent baseline and 6-month follow-up angiographic and virtual histology-intravascular ultrasound (VH-IVUS) imaging. VH-IVUS images were processed to remove background noise, extract VH plaque material data, and luminal and outer contours. Angiography data was used to orient the artery profiles along the 3D centerlines. The resulting surface mesh is then resampled for uniformity and tetrahedralized to generate the volumetric mesh using TetGen. A mesh convergence study revealed edge lengths between 0.04 mm and 0.2 mm produced constituent volumes that were largely unchanged, hence, to save computational resources, a value of 0.2 mm was used throughout. Materials are assigned and finite element analysis (FEA) is then performed to determine stresses and strains across the artery wall. In a representative artery, the highest average effective stress was in calcium elements with 235 kPa while necrotic elements had the lowest average stress, reaching as low as 0.79 kPa. After applying nodal smoothening, the maximum effective stress across 11 arteries remained below 288 kPa, implying biomechanically stable plaques. Indeed, all atherosclerotic plaques remained unruptured at the 6-month longitudinal follow up diagnosis. These results suggest our automated analysis may facilitate assessment of atherosclerotic plaque stability.

6.
Ann Biomed Eng ; 49(12): 3425-3437, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34734362

ABSTRACT

Macrophage to foam cell transition and their accumulation in the arterial intima are the key events that trigger atherosclerosis, a multifactorial inflammatory disease. Previous studies have linked arterial stiffness and cardiovascular disease and have highlighted the use of arterial stiffness as a potential early-stage marker. Yet the relationship between arterial stiffness and atherosclerosis in terms of macrophage function is poorly understood. Thus, it is pertinent to understand the mechanobiology of macrophages to clarify their role in plaque advancement. We explore how substrate stiffness affects proliferation of macrophages and foam cells, traction forces exerted by macrophages and uptake of native and oxidized low-density lipoproteins. We demonstrate that stiffness influences foam cell proliferation under both naïve and inflammatory conditions. Naïve foam cells proliferated faster on the 4 kPa polyacrylamide gel and glass whereas under inflammatory conditions, maximum proliferation was recorded on glass. Macrophage and foam cell traction forces were positively correlated to the substrate stiffness. Furthermore, the influence of stiffness was demonstrated on the uptake of lipoproteins on macrophages treated with lipopolysaccharide + interferon gamma. Cells on softer 1 kPa substrates had a significantly higher uptake of low-density lipoproteins and oxidized low-density lipoproteins compared to stiffer substrates. The results herein indicate that macrophage function is modulated by stiffness and help better understand ways in which macrophages and foam cells could contribute to the development and progression of atherosclerotic plaque.


Subject(s)
Cell Proliferation , Foam Cells/metabolism , Inflammation/physiopathology , Lipoproteins/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic/physiopathology , Vascular Stiffness/physiology , Cells, Cultured , Humans , Inflammation/metabolism , Plaque, Atherosclerotic/metabolism , Tunica Intima/metabolism
7.
Nano Lett ; 21(22): 9805-9815, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34516144

ABSTRACT

The blood-brain barrier (BBB) is highly selective and acts as the interface between the central nervous system and circulation. While the BBB is critical for maintaining brain homeostasis, it represents a formidable challenge for drug delivery. Here we synthesized gold nanoparticles (AuNPs) for targeting the tight junction specifically and demonstrated that transcranial picosecond laser stimulation of these AuNPs post intravenous injection increases the BBB permeability. The BBB permeability change can be graded by laser intensity, is entirely reversible, and involves increased paracellular diffusion. BBB modulation does not lead to significant disruption in the spontaneous vasomotion or the structure of the neurovascular unit. This strategy allows the entry of immunoglobulins and viral gene therapy vectors, as well as cargo-laden liposomes. We anticipate this nanotechnology to be useful for tissue regions that are accessible to light or fiberoptic application and to open new avenues for drug screening and therapeutic interventions in the central nervous system.


Subject(s)
Metal Nanoparticles , Nanoparticles , Biological Transport , Blood-Brain Barrier , Gold/chemistry , Lasers
8.
Sci Rep ; 11(1): 12680, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135399

ABSTRACT

Numerical simulations of coupled hemodynamics and leukocyte transport and adhesion inside coronary arteries have been performed. Realistic artery geometries have been obtained for a set of four patients from intravascular ultrasound and angiography images. The numerical model computes unsteady three-dimensional blood hemodynamics and leukocyte concentration in the blood. Wall-shear stress dependent leukocyte adhesion is also computed through agent-based modeling rules, fully coupled to the hemodynamics and leukocyte transport. Numerical results have a good correlation with clinical data. Regions where high adhesion is predicted by the simulations coincide to a good approximation with artery segments presenting plaque increase, as documented by clinical data from baseline and six-month follow-up exam of the same artery. In addition, it is observed that the artery geometry and, in particular, the tortuosity of the centerline are a primary factor in determining the spatial distribution of wall-shear stress, and of the resulting leukocyte adhesion patterns. Although further work is required to overcome the limitations of the present model and ultimately quantify plaque growth in the simulations, these results are encouraging towards establishing a predictive methodology for atherosclerosis progress.


Subject(s)
Cell Adhesion , Coronary Artery Disease/physiopathology , Coronary Vessels/physiopathology , Hemodynamics , Leukocytes/physiology , Models, Cardiovascular , Coronary Angiography , Coronary Artery Disease/diagnostic imaging , Coronary Circulation , Coronary Vessels/diagnostic imaging , Humans , Stress, Mechanical , Ultrasonography
9.
Tissue Eng Part C Methods ; 25(11): 641-654, 2019 11.
Article in English | MEDLINE | ID: mdl-31392930

ABSTRACT

Over the past two decades, the increase in prevalence of cardiovascular diseases and the limited availability of autologous blood vessels and saphenous vein grafts have motivated the development of tissue-engineered vascular grafts (TEVGs). However, compliance mismatch and poor mechanical properties of the TEVGs remain as two major issues that need to be addressed. Researchers have investigated the role of various culture conditions and mechanical conditioning in deposition and orientation of collagen fibers, which are the key structural components in the vascular wall; however, the intrinsic complexity of mechanobiological interactions demands implementing new engineering approaches that allow researchers to investigate various scenarios more efficiently. In this study, we utilized a coupled agent-based finite element analysis (AB-FEA) modeling approach to study the effect of various loading modes (uniaxial, biaxial, and equibiaxial), boundary conditions, stretch magnitudes, and growth factor concentrations on growth and remodeling of smooth muscle cell-populated TEVGs, with specific focus on collagen deposition and orientation. Our simulations (12 weeks of culture) showed that biaxial cyclic loading (and not uniaxial or equibiaxial) leads to alignment of collagen fibers in the physiological directions. Moreover, axial boundary conditions of the TEVG act as determinants of fiber orientations. Decreasing the serum concentration, from 10% to 5% or 1%, significantly decreased the growth and remodeling speed, but only affected the fiber orientation in the 1% serum case. In conclusion, in silico tissue engineering has the potential to evolve the future of tissue engineering, as it will allow researchers to conceptualize various interactions and investigate numerous scenarios with great speed. In this study, we were able to predict the orientation of collagen fibers in TEVGs using a coupled AB-FEA model in less than 8 h. Impact Statement Tissue-engineered vascular grafts (TEVGs) hold potential to replace the current gold standard of vascular grafting, saphenous vein grafts. However, developing TEVGs that mimic the mechanical performance of the native tissue remains a challenging task. We developed a computational model of the grafts' remodeling processes and studied the effects of various loading mechanisms and culture conditions on collagen fiber orientation, which is a key factor in mechanical performance of the grafts. We were able to predict the fiber orientations accurately and show that biaxial loading and axial boundary conditions are important factors in collagen fiber organization.


Subject(s)
Computer Simulation , Finite Element Analysis , Systems Analysis , Tissue Engineering/methods , Animals , Blood Vessel Prosthesis , Collagen/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Stress, Mechanical , Weight-Bearing
10.
World Neurosurg ; 123: 68-75, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30503286

ABSTRACT

BACKGROUND: Hemangiopericytomas (HPCs) are rare vascular tumors that resemble meningiomas on imaging and have a high rate of local recurrence and metastases. There remains a paucity of data to guide management decisions of intraspinal dissemination of HPCs in the literature, and none specifically related to anaplastic HPCs. CASE DESCRIPTION: We report a case of a 34-year-old woman with locally and distantly recurrent anaplastic HPC (World Health Organization grade III). She initially presented with tinnitus in her right ear. A well-circumscribed, contrast-enhancing lesion was identified in the right cerebellopontine angle. Treatment consisted of subtotal resection and postoperative radiation therapy (RT) to a dose of 60 Gy in 30 fractions. After a 3-year disease-free interval, 7 lesions recurred intra- and extracranially. The extracranial lesions were drop metastases of the original HPC through the cerebrospinal fluid into the spinal canal. Of note, fluorodeoxyglucose positron emission tomography (PET)/computed tomography scan was not sensitive enough to detect these new lesions. The intracranial recurrence was on the edge of the prior radiotherapy field, representing a marginal failure having received less than 50 Gy. The intracranial recurrences were treated with salvage gamma knife stereotactic radiosurgery (SRS) with local control. She underwent intradural extramedullary hemilaminectomy of a thoracic spine metastasis followed by fractionated proton beam therapy (PBT) with a boost to unresected lesions. Within a few months of PBT, she became pregnant. Pregnancy did not affect recurrence or ameliorate tumor growth. CONCLUSIONS: This case report discusses the role genetics, adjuvant RT, SRS, magnetic resonance imaging, and PET scan played in this unique clinical scenario of anaplastic HPC.


Subject(s)
Brain Neoplasms/pathology , Hemangiopericytoma/pathology , Spinal Cord Neoplasms/secondary , Adult , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery , Female , Hemangiopericytoma/diagnostic imaging , Hemangiopericytoma/radiotherapy , Hemangiopericytoma/surgery , Humans , Magnetic Resonance Imaging , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/radiotherapy , Neoplasm Recurrence, Local/surgery , Neurosurgical Procedures , Positron-Emission Tomography , Radiosurgery , Spinal Cord Neoplasms/diagnostic imaging , Treatment Outcome
11.
J Biomech ; 76: 84-93, 2018 07 25.
Article in English | MEDLINE | ID: mdl-29914741

ABSTRACT

Atherosclerosis, an artery disease, is currently the leading cause of death in the United States in both men and women. The first step in the development of atherosclerosis involves leukocyte adhesion to the arterial endothelium. It is broadly accepted that blood flow, more specifically wall shear stress (WSS), plays an important role in leukocyte capture and subsequent development of an atherosclerotic plaque. What is less known is how instantaneous WSS, which can vary by up to 5 Pa over one cardiac cycle, influences leukocyte capture. In this paper we use direct numerical simulations (DNS), performed using an in-house code, to illustrate that leukocyte capture is different whether as a function of instantaneous or time-averaged blood flow. Specifically, a stenotic plaque is modeled using a computational fluid dynamics (CFD) solver through fully three-dimensional Navier-Stokes equations and the immersed boundary method. Pulsatile triphasic inflow is used to simulate the cardiac cycle. The CFD is coupled with an agent-based leukocyte capture model to assess the impact of instantaneous hemodynamics on stenosis growth. The computed wall shear stress agrees well with the results obtained with a commercial software, as well as with theoretical results in the healthy region of the artery. The analysis emphasizes the importance of the instantaneous flow conditions in evaluating the leukocyte rate of capture. That is, the capture rate computed from mean flow field is generally underpredicted compared to the actual rate of capture. Thus, in order to obtain a reliable estimate, the flow unsteadiness during a cardiac cycle should be taken into account.


Subject(s)
Leukocytes/cytology , Pulsatile Flow , Arteries/physiopathology , Humans , Hydrodynamics , Models, Cardiovascular , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/physiopathology , Software , Stress, Mechanical
12.
Am J Respir Cell Mol Biol ; 58(2): 216-231, 2018 02.
Article in English | MEDLINE | ID: mdl-28915065

ABSTRACT

Hypoxia has long been implicated in the pathogenesis of fibrotic diseases. Aberrantly activated myofibroblasts are the primary pathological driver of fibrotic progression, yet how various microenvironmental influences, such as hypoxia, contribute to their sustained activation and differentiation is poorly understood. As a defining feature of hypoxia is its impact on cellular metabolism, we sought to investigate how hypoxia-induced metabolic reprogramming affects myofibroblast differentiation and fibrotic progression, and to test the preclinical efficacy of targeting glycolytic metabolism for the treatment of pulmonary fibrosis. Bleomycin-induced pulmonary fibrotic progression was evaluated in two independent, fibroblast-specific, promoter-driven, hypoxia-inducible factor (Hif) 1A knockout mouse models and in glycolytic inhibitor, dichloroacetate-treated mice. Genetic and pharmacological approaches were used to explicate the role of metabolic reprogramming in myofibroblast differentiation. Hypoxia significantly enhanced transforming growth factor-ß-induced myofibroblast differentiation through HIF-1α, whereas overexpression of the critical HIF-1α-mediated glycolytic switch, pyruvate dehydrogenase kinase 1 (PDK1) was sufficient to activate glycolysis and potentiate myofibroblast differentiation, even in the absence of HIF-1α. Inhibition of the HIF-1α/PDK1 axis by genomic deletion of Hif1A or pharmacological inhibition of PDK1 significantly attenuated bleomycin-induced pulmonary fibrosis. Our findings suggest that HIF-1α/PDK1-mediated glycolytic reprogramming is a critical metabolic alteration that acts to promote myofibroblast differentiation and fibrotic progression, and demonstrate that targeting glycolytic metabolism may prove to be a potential therapeutic strategy for the treatment of pulmonary fibrosis.


Subject(s)
Cell Hypoxia/physiology , Dichloroacetic Acid/pharmacology , Glycolysis/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pulmonary Fibrosis/pathology , Animals , Bleomycin , Cell Line , Humans , Lung/pathology , Mice , Mice, Knockout , Myofibroblasts/cytology , Myofibroblasts/pathology , Protein Serine-Threonine Kinases/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA Interference , RNA, Small Interfering/genetics
13.
Biomech Model Mechanobiol ; 17(1): 87-101, 2018 02.
Article in English | MEDLINE | ID: mdl-28823079

ABSTRACT

A coupled agent-based model (ABM) and finite element analysis (FEA) computational framework is developed to study the interplay of bio-chemo-mechanical factors in blood vessels and their role in maintaining homeostasis. The agent-based model implements the power of REPAST Simphony libraries and adapts its environment for biological simulations. Coupling a continuum-level model (FEA) to a cellular-level model (ABM) has enabled this computational framework to capture the response of blood vessels to increased or decreased levels of growth factors, proteases and other signaling molecules (on the micro scale) as well as altered blood pressure. Performance of the model is assessed by simulating porcine left anterior descending artery under normotensive conditions and transient increases in blood pressure and by analyzing sensitivity of the model to variations in the rule parameters of the ABM. These simulations proved that the model is stable under normotensive conditions and can recover from transient increases in blood pressure. Sensitivity studies revealed that the model is most sensitive to variations in the concentration of growth factors that affect cellular proliferation and regulate extracellular matrix composition (mainly collagen).


Subject(s)
Arteries/growth & development , Models, Cardiovascular , Vascular Remodeling , Adventitia/physiology , Animals , Biomechanical Phenomena , Blood Pressure , Extracellular Matrix/metabolism , Finite Element Analysis , Mice , Stress, Mechanical , Sus scrofa , Systems Analysis , User-Computer Interface
14.
PLoS Comput Biol ; 13(5): e1005523, 2017 05.
Article in English | MEDLINE | ID: mdl-28542193

ABSTRACT

A vast amount of work has been dedicated to the effects of hemodynamics and cytokines on leukocyte adhesion and trans-endothelial migration (TEM) and subsequent accumulation of leukocyte-derived foam cells in the artery wall. However, a comprehensive mechanobiological model to capture these spatiotemporal events and predict the growth and remodeling of an atherosclerotic artery is still lacking. Here, we present a multiscale model of leukocyte TEM and plaque evolution in the left anterior descending (LAD) coronary artery. The approach integrates cellular behaviors via agent-based modeling (ABM) and hemodynamic effects via computational fluid dynamics (CFD). In this computational framework, the ABM implements the diffusion kinetics of key biological proteins, namely Low Density Lipoprotein (LDL), Tissue Necrosis Factor alpha (TNF-α), Interlukin-10 (IL-10) and Interlukin-1 beta (IL-1ß), to predict chemotactic driven leukocyte migration into and within the artery wall. The ABM also considers wall shear stress (WSS) dependent leukocyte TEM and compensatory arterial remodeling obeying Glagov's phenomenon. Interestingly, using fully developed steady blood flow does not result in a representative number of leukocyte TEM as compared to pulsatile flow, whereas passing WSS at peak systole of the pulsatile flow waveform does. Moreover, using the model, we have found leukocyte TEM increases monotonically with decreases in luminal volume. At critical plaque shapes the WSS changes rapidly resulting in sudden increases in leukocyte TEM suggesting lumen volumes that will give rise to rapid plaque growth rates if left untreated. Overall this multi-scale and multi-physics approach appropriately captures and integrates the spatiotemporal events occurring at the cellular level in order to predict leukocyte transmigration and plaque evolution.


Subject(s)
Atherosclerosis/metabolism , Leukocytes/physiology , Models, Cardiovascular , Plaque, Atherosclerotic/metabolism , Transendothelial and Transepithelial Migration/physiology , Animals , Computational Biology , Computer Simulation , Cytokines/metabolism , Hemodynamics , Humans , Mice
15.
Mol Cell Biol ; 36(22): 2824-2837, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27573018

ABSTRACT

Tissue cells respond to changes in tensional forces with proliferation or death through the control of RhoA. However, the response coupling mechanisms that link force with RhoA activation are poorly understood. We found that tension applied to fibronectin-coated microbeads caused recruitment of all three isoforms of the Shc adapter (p66Shc, p52Shc, and p46Shc) to adhesion complexes. The Shc PTB domain was necessary and sufficient for this recruitment, and screening studies revealed the direct interactions with the FERM domain of focal adhesion kinase (FAK) that were required for Shc translocation to adhesion complexes. The FAK/p66Shc complex specifically bound and activated the Rho guanyl exchange factors (GEFs) p115-RhoGEF and GEF-H1, leading to tension-induced RhoA activation. In contrast, the FAK/p52Shc complex bound SOS1 but not the Rho GEFs to mediate tension-induced Ras activation. Nuclear translocation and activation of the YAP/TAZ transcription factors on firm substrates required the FAK/p66Shc/Rho GEF complex, and both proliferation on firm substrates and anoikis in suspension required signaling through p66Shc and its associated Rho GEFs. These studies reveal the binary and exclusive assignment of p66Shc and p52Shc to tension-induced Rho or Ras signals, respectively, and suggest an integrated role for the two Shc isoforms in coordinating the cellular response to mechanical stimuli.

16.
Eur Biophys J ; 45(4): 301-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26613613

ABSTRACT

Macrophages play a key role in atherosclerosis, cancer, and in the response to implanted medical devices. In each of these situations, the mechanical environment of a macrophage can vary from soft to stiff. However, how stiffness affects macrophage behavior remains uncertain. Using substrates of varying stiffness, we show macrophage phenotype and function depends on substrate stiffness. Notably, the cell area increases slightly from a sphere after 18 h on substrates mimicking healthy arterial stiffness (1-5 kPa), whereas macrophages on stiffer substrates (280 kPa-70 GPa) increased in area by nearly eight-fold. Macrophage migration is random regardless of substrate stiffness. The total average track speed was 7.8 ± 0.5 µm/h, with macrophages traveling fastest on the 280-kPa substrate (12.0 ± 0.5 µm/h) and slowest on the 3-kPa substrate (5.0 ± 0.4 µm/h). In addition F-actin organization in macrophages depends on substrate stiffness. On soft substrates, F-actin is spread uniformly throughout the cytoplasm, whereas on stiff substrates F-actin is functionalized into stress fibers. The proliferation rate of macrophages was faster on stiff substrates. Cells plated on the 280-kPa gel had a significantly shorter doubling time than those plated on the softer substrate. However, the ability of macrophages to phagocytose 1-µm particles did not depend on substrate stiffness. In conclusion, the results herein show macrophages are mechanosensitive; they respond to changes in stiffness by modifying their area, migration speed, actin organization, and proliferation rate. These results are important to understanding how macrophages respond in complex mechanical environments such as an atherosclerotic plaque.


Subject(s)
Elasticity , Macrophages/cytology , Monocytes/cytology , Biomechanical Phenomena , Cell Movement , Cell Proliferation , Cytoskeleton/metabolism , Humans , Phagocytosis
17.
PLoS One ; 8(4): e61377, 2013.
Article in English | MEDLINE | ID: mdl-23626676

ABSTRACT

Transmigration through the endothelium is a key step in the immune response. In our recent work, the mechanical properties of the subendothelial matrix and biophysical state of the endothelium have been identified as key modulators of leukocyte trans-endothelial migration. Here, we demonstrated that neutrophil contractile forces and cytoskeletal dynamics also play an active biophysical role during transmigration through endothelial cell-cell junctions. Using our previously-established model for leukocyte transmigration, we first discovered that >93% of human neutrophils preferentially exploit the paracellular mode of transmigration in our in vitro model, and that is independent of subendothelial matrix stiffness. We demonstrated that inhibition of actin polymerization or depolymerization completely blocks transmigration, thus establishing a critical role for neutrophil actin dynamics in transmigration. Next, inhibition of neutrophil myosin II-mediated contractile forces renders 44% of neutrophils incapable of retracting their trailing edge under the endothelium for several minutes after the majority of the neutrophil transmigrates. Meanwhile, inhibition of neutrophil contractile forces or stabilization of microtubules doubles the time to complete transmigration for the first neutrophils to cross the endothelium. Notably, the time to complete transmigration is significantly reduced for subsequent neutrophils that cross through the same path as a previous neutrophil and is less dependent on neutrophil contractile forces and microtubule dynamics. These results suggest that the first neutrophil induces a gap in endothelial cell-cell adhesions, which "opens the door" in the endothelium and facilitates transmigration of subsequent neutrophils through the same hole. Collectively, this work demonstrates that neutrophils play an active biophysical role during the transmigration step of the immune response.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Movement/immunology , Microtubules/metabolism , Neutrophils/immunology , Transendothelial and Transepithelial Migration/immunology , Actin Cytoskeleton/ultrastructure , Actins/metabolism , Cell Adhesion , Cells, Cultured , Endothelium/cytology , Genes, Reporter , Green Fluorescent Proteins , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intercellular Junctions , Leukocyte Count , Microtubules/ultrastructure , Myosin Type II/metabolism , Neutrophils/cytology , Tumor Necrosis Factor-alpha/pharmacology
18.
Acta Biomater ; 8(11): 3974-81, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22796654

ABSTRACT

Tissue engineering strategies based on multipotent stem cells (MSCs) hold significant promise for the repair or replacement of damaged smooth muscle tissue. To design scaffolds which specifically induce MSC smooth muscle lineage progression requires a deeper understanding of the relative influence of various microenvironmental signals on myogenesis. For instance, MSC myogenic differentiation has been shown to be promoted by increases in active RhoA and FAK, both of which can be induced via increased cell-substrate stress. Separate studies have demonstrated MSC myogenesis to be enhanced by uniaxial cell alignment. The goal of the present study was to compare the impact of increased peak cell-substrate stresses vs. increased uniaxial cell alignment on MSC myogenic differentiation. To this end, MSC fate decisions were compared within two distinct multicellular "forms". A "stripe" multicellular pattern was designed to induce uniaxial cell alignment. In contrast, a second multicellular pattern was designed with "loops" or curves, which altered cell directionality while simultaneously generating regional peak stresses significantly above that intrinsic to the "stripe" form. As anticipated, the higher peak stress levels of the "loop" pattern were associated with increased fractions of active RhoA and active FAK. In contrast, two markers of early smooth muscle lineage progression, myocardin and SM-α-actin, were significantly elevated in the "stripe" pattern relative to the "loop" pattern. These results indicate that scaffolds which promote uniaxial MSC alignment may be more inductive of myogenic differentiation than those associated with increased peak, cell-substrate stress but in which cell directionality varies.


Subject(s)
Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Muscle Development , Stress, Mechanical , Actins/genetics , Actins/metabolism , Animals , Biomarkers/metabolism , Cadherins/metabolism , Cell Lineage , Elastic Modulus , Enzyme-Linked Immunosorbent Assay , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation , Hedgehog Proteins/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Mice , Multipotent Stem Cells/enzymology , NIH 3T3 Cells , Transcription Factors/metabolism , rhoA GTP-Binding Protein/metabolism
19.
Tissue Eng Part A ; 18(15-16): 1710-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22519299

ABSTRACT

Growth factors have been shown to be potent mediators of osteogenesis. However, their use in tissue-engineered scaffolds not only can be costly but also can induce undesired responses in surrounding tissues. Thus, the ability to specifically induce osteogenic differentiation in the absence of exogenous growth factors through manipulation of scaffold material properties would be desirable for bone regeneration. Previous research indicates that addition of inorganic or hydrophobic components to organic, hydrophilic scaffolds can enhance multipotent stem cell (MSC) osteogenesis. However, the combined impact of scaffold inorganic content and hydrophobicity on MSC behavior has not been systematically explored, particularly in three-dimensional (3D) culture systems. The aim of the present study was therefore to examine the effects of simultaneous increases in scaffold hydrophobicity and inorganic content on MSC osteogenic fate decisions in a 3D culture environment toward the development of intrinsically osteoinductive scaffolds. Mouse 10T½ MSCs were encapsulated in a series of novel scaffolds composed of varying levels of hydrophobic, inorganic poly(dimethylsiloxane) (PDMS) and hydrophilic, organic poly(ethylene glycol) (PEG). After 21 days of culture, increased levels of osteoblast markers, runx2 and osteocalcin, were observed in scaffolds with increased PDMS content. Bone extracellular matrix (ECM) molecules, collagen I and calcium phosphate, were also elevated in formulations with higher PDMS:PEG ratios. Importantly, this osteogenic response appeared to be specific in that markers for chondrocytic, smooth muscle cell, and adipocytic lineages were not similarly affected by variations in scaffold PDMS content. As anticipated, the increase in scaffold hydrophobicity accompanying increasing PDMS levels was associated with elevated scaffold serum protein adsorption. Thus, scaffold inorganic content combined with alterations in adsorbed serum proteins may underlie the observed cell behavior.


Subject(s)
Dimethylpolysiloxanes/pharmacology , Hydrogels/chemistry , Osteogenesis/drug effects , Polyethylene Glycols/pharmacology , Adsorption/drug effects , Animals , Biomarkers/metabolism , Blood Proteins/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Collagen/metabolism , Dimethylpolysiloxanes/chemistry , Elastic Modulus/drug effects , Elastin/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Glycosaminoglycans/metabolism , Hydrophobic and Hydrophilic Interactions/drug effects , Materials Testing , Mice , Particle Size , Phenotype , Polyethylene Glycols/chemistry
20.
Ann Biomed Eng ; 39(11): 2669-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21809144

ABSTRACT

There is a need to develop multiscale models of vascular adaptations to understand tissue-level manifestations of cellular level mechanisms. Continuum-based biomechanical models are well suited for relating blood pressures and flows to stress-mediated changes in geometry and properties, but less so for describing underlying mechanobiological processes. Discrete stochastic agent-based models are well suited for representing biological processes at a cellular level, but not for describing tissue-level mechanical changes. We present here a conceptually new approach to facilitate the coupling of continuum and agent-based models. Because of ubiquitous limitations in both the tissue- and cell-level data from which one derives constitutive relations for continuum models and rule-sets for agent-based models, we suggest that model verification should enforce congruency across scales. That is, multiscale model parameters initially determined from data sets representing different scales should be refined, when possible, to ensure that common outputs are consistent. Potential advantages of this approach are illustrated by comparing simulated aortic responses to a sustained increase in blood pressure predicted by continuum and agent-based models both before and after instituting a genetic algorithm to refine 16 objectively bounded model parameters. We show that congruency-based parameter refinement not only yielded increased consistency across scales, it also yielded predictions that are closer to in vivo observations.


Subject(s)
Arteries/physiology , Models, Cardiovascular , Muscle, Smooth, Vascular/physiology , Algorithms , Animals , Arteries/cytology , Biomechanical Phenomena/physiology , Blood Pressure/physiology , Cell Physiological Phenomena , Computer Simulation , Mice , Muscle Development/physiology , Muscle, Smooth, Vascular/cytology
SELECTION OF CITATIONS
SEARCH DETAIL