Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Mol Pharm ; 2024 Oct 14.
Article in English | MEDLINE | ID: mdl-39400078

ABSTRACT

The blood-brain barrier (BBB) is a highly restrictive barrier at the interface between the brain and the vascular system. Even under BBB dysfunction, it is extremely difficult to deliver therapies across the barrier, limiting the options for treatment of neurological injuries and disorders. To circumvent these challenges, there is interest in developing therapies that directly engage with the damaged BBB to restore its function. Previous studies revealed that poloxamer 188 (P188), a water-soluble triblock copolymer of poly(ethylene oxide) (PEO) and poly(propylene oxide) (PPO), partially mitigated BBB dysfunction in vivo. In the context of stabilization of the damaged BBB, the mechanism of action of PEO-PPO block copolymers is unknown, and there has been minimal exploration of polymers beyond P188. In this study, a human-based in vitro BBB model under oxidative stress was used to investigate polymer-BBB interactions since oxidative stress is closely linked with BBB dysfunction in many neurological injuries and disorders. PEO-PPO block copolymers of varied numbers of chemically distinct blocks, PEO block length, and functionality of the end group of the PPO block were assessed for their efficacy in improving key physiological readouts associated with BBB dysfunction. While treatment with P188 did not mitigate damage in the in vitro BBB model, treatment with three diblock copolymers improved barrier integrity under oxidative stress to a similar extent. Of the considered variations in the block copolymer design, the reduction in the number of chemically distinct blocks had the strongest influence on therapeutic function. The demonstrated efficacy of three alternative PEO-PPO diblock copolymers in this work reveals the potential of these polymers as a class of therapeutics that directly treat the damaged BBB, expanding the options for treatment of neurological injuries and disorders.

2.
Biophys J ; 120(4): 687-698, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33453275

ABSTRACT

Multistability and natural biological variability can result in significant heterogeneity within a cell population, leading to challenges in understanding and modulating cell behavior. Energy landscapes can offer qualitatively intuitive visualizations of cell phenotype and facilitate a more quantitative understanding of cellular dynamics, but current methods for landscape generation are mathematically involved and often require specific system properties (e.g., ergodicity or independent gene/protein probability distributions) that do not always hold. Here, we present a simple kinetic Monte Carlo-based method for landscape generation from a system of ordinary differential equations using only simulation trajectories initialized throughout the phase space of interest. The resulting landscape produces three quantitative features relevant to understanding cell behavior: stability (reflected by the depth or potential of landscape valleys), velocity (representing average directional movement on the landscape), and variance in velocity (indicative of landscape positions with heterogeneous movements). We applied this method to a genetic toggle switch, a core decision-making network in binary cellular responses, to elucidate effects of biologically relevant intrinsic and extrinsic cues. Intrinsic noise, such as stochasticity in transcription-translation and differences in cell cycle position, manifests through changes in valley width and position, reflecting increased population heterogeneity and more probabilistic cell fate transitions. The landscapes also capture the effect of an external inducer, revealing a quantitative correlation between the rate of cell fate transition and the energy barrier above a threshold inducer concentration determined by the permissivity of the valley. Further, in tracking dynamically changing landscapes under time-varying external cues, we unexpectedly found that an oscillatory inducer input can modulate cell fate heterogeneity and lead to periodic cell fate transitions entrained to the input frequency, depending on the intrinsic degradation rate of the switch. The landscape generation approach outlined herein is generalizable to other network topologies and may provide new quantitative insights into their dynamics.


Subject(s)
Gene Regulatory Networks , Cell Cycle , Cell Differentiation , Computer Simulation , Kinetics
3.
Biotechnol Bioeng ; 117(10): 3066-3080, 2020 10.
Article in English | MEDLINE | ID: mdl-32589792

ABSTRACT

Around 20-30% of ovarian cancer patients exhibit chemoresistance, but there are currently no methods to predict whether a patient will respond to chemotherapy. Here, we discovered that chemoresistant ovarian cancer cells exhibit enhanced survival in a quiescent state upon experiencing the stress of physical confinement. When immobilized in stiff silica gels, most ovarian cancer cells die within days, but surviving cells exhibit hallmarks of single-cell dormancy. Upon extraction from gels, the cells resume proliferation but demonstrate enhanced viability upon reimmobilization, indicating that initial immobilization selects for cells with a higher propensity to enter dormancy. RNA-seq analysis of the extracted cells shows they have signaling responses similar to cells surviving cisplatin treatment, and in comparison to chemoresistant patient cohorts, they share differentially expressed genes that are associated with platinum-resistance pathways. Furthermore, these extracted cells demonstrate greater resistance to cisplatin and paclitaxel, despite being proliferative. In contrast, serum starvation and hypoxia could not effectively select for chemoresistant cells upon removal of the environmental stress. These findings demonstrate that ovarian cancer chemoresistance and the ability to enter dormancy are linked, and immobilization rapidly distinguishes chemoresistant cells. This platform could be suitable for mechanistic studies, drug development, or as a clinical diagnostic tool.


Subject(s)
Biological Assay/methods , Cell Survival , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Silica Gel/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Signal Transduction
4.
Langmuir ; 36(13): 3393-3403, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32216370

ABSTRACT

Maintaining the integrity of cell membranes is indispensable for cellular viability. Poloxamer 188 (P188), a poly(ethylene oxide)-b-poly(propylene oxide)-b-poly(ethylene oxide) (PEO-PPO-PEO) triblock copolymer with a number-average molecular weight of 8700 g/mol and containing 80% by mass PEO, protects cell membranes from various external injuries and has the potential to be used as a therapeutic agent in diverse applications. The membrane protection mechanism associated with P188 is intimately connected with how this block copolymer interacts with the lipid bilayer, the main component of a cell membrane. Here, we report the distribution of P188 in a model lipid bilayer comprising 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (POPC) using neutron reflectivity (NR) and atomic force microscopy (AFM). We also investigated the association of a PEO homopolymer (PEO8.4K; Mn = 8400 g/mol) that does not protect living cell membranes. These experiments were conducted following incubation of a 4.5 mmol/L polymer solution in a buffer that mimics physiological conditions with supported POPC bilayer membranes followed by washing with the aqueous medium. In contrast to previous reports, which dealt with P188 and PEO in salt-free solutions, both P188 and PEO8.4K penetrate into the inner portion of the lipid bilayer as revealed by NR, with approximately 30% by volume occupancy across the membrane without loss of bilayer structural integrity. These results indicate that PEO is the chemical moiety that principally drives P188 binding to bilayer membranes. No defects or phase-separated domains were observed in either P188- or PEO8.4K-incubated lipid bilayers when examined by AFM, indicating that polymer chains mingle homogeneously with lipid molecules in the bilayer. Remarkably, the breakthrough force required for penetration of the AFM tip through the bilayer membrane is unaffected by the presence of the large amount of P188 and PEO8.4K.


Subject(s)
Lipid Bilayers , Propylene Glycols , Polyethylene Glycols , Polymers
5.
Biotechnol Bioeng ; 116(9): 2353-2363, 2019 09.
Article in English | MEDLINE | ID: mdl-31038193

ABSTRACT

Local delivery of viral vectors can enhance the efficacy of therapies by selectively affecting necessary tissues and reducing the required vector dose. Pluronic F127 is a thermosensitive polymer that undergoes a solution-gelation (sol-gel) transition as temperature increases and can deliver vectors without damaging them. While pluronics can be spread over large areas, such as the surface of an organ, before gelation, they lack sufficient adhesivity to remain attached to some tissues, such as the surface of the heart or mucosal surfaces. Here, we utilized blends of pluronic F127 and polycarbophil (PCB), a mucoadhesive agent, to provide the necessary adhesivity for local delivery of viral vectors to the cardiac muscle. The effects of PCB concentration on adhesive properties, sol-gel temperature transition and cytocompatibility were evaluated. Rheological studies showed that PCB decreased the sol-gel transition temperature at concentrations >1% and increased the adhesive properties of the gel. Furthermore, these gels were able to deliver viral vectors and transduce cells in vitro and in vivo in a neonatal mouse apical resection model. These gels could be a useful platform for delivering viral vectors over the surface of organs where increased adhesivity is required.


Subject(s)
Acrylic Resins , Gene Transfer Techniques , Genetic Vectors , Myocardium/metabolism , Poloxamer , Tissue Adhesives , Viruses , Acrylic Resins/chemistry , Acrylic Resins/pharmacology , Cells, Cultured , Fibroblasts/metabolism , Humans , Materials Testing , Poloxamer/chemistry , Poloxamer/pharmacology , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology
6.
Int J Hyperthermia ; 36(1): 130-138, 2019.
Article in English | MEDLINE | ID: mdl-30676126

ABSTRACT

Focal thermal therapy (Heat), cryosurgery (Cryo) and irreversible electroporation (IRE) are increasingly used to treat cancer. However, local recurrence and systemic spread are persistent negative outcomes. Nevertheless, emerging work with immunotherapies (i.e., checkpoint blockade or dendritic cell (DC) vaccination) in concert with focal therapies may improve outcomes. To understand the role of focal therapy in priming the immune system for immunotherapy, an in vitro model of T cell response after exposure to B16 melanoma cell lysates after lethal exposures was designed. Exposure included: Heat (50 °C, 30 min), Cryo (-80 °C, 30 min) and IRE (1250 V/cm, 99 pulses, 50 µs pulses with 1 Hz intervals). After viability assessment (CCK-8 assay), cell lysates were collected and assessed for protein release (BCA assay), protein denaturation (FTIR-spectroscopy), TRP-2 antigen release (western blot), and T cell activation (antigen-specific CD8 T cell proliferation). Results showed IRE released the most protein and antigen (TRP-2), followed by Cryo and Heat. In contrast, Cryo released the most native (not denatured) protein, compared to IRE and Heat. Finally, IRE dramatically outperformed both Cryo and Heat in T cell activation while Cryo modestly outperformed Heat. This study demonstrates that despite all focal therapies ability to destroy cells, the 'quantity' (i.e., amount) and 'quality' (i.e., molecular state) of tumor protein (including antigen) released can dramatically change the ensuing priming of the immune system. This suggests protein-based metrics whereby focal therapies can be designed to prime the immune system in concert with immunotherapies to eventually achieve improved and durable cancer treatment in vivo.


Subject(s)
Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Animals , Humans , Mice , Mice, Transgenic
7.
Proc Natl Acad Sci U S A ; 109(27): E1848-57, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22645348

ABSTRACT

Human pluripotent stem cells (hPSCs) offer the potential to generate large numbers of functional cardiomyocytes from clonal and patient-specific cell sources. Here we show that temporal modulation of Wnt signaling is both essential and sufficient for efficient cardiac induction in hPSCs under defined, growth factor-free conditions. shRNA knockdown of ß-catenin during the initial stage of hPSC differentiation fully blocked cardiomyocyte specification, whereas glycogen synthase kinase 3 inhibition at this point enhanced cardiomyocyte generation. Furthermore, sequential treatment of hPSCs with glycogen synthase kinase 3 inhibitors followed by inducible expression of ß-catenin shRNA or chemical inhibitors of Wnt signaling produced a high yield of virtually (up to 98%) pure functional human cardiomyocytes from multiple hPSC lines. The robust ability to generate functional cardiomyocytes under defined, growth factor-free conditions solely by genetic or chemically mediated manipulation of a single developmental pathway should facilitate scalable production of cardiac cells suitable for research and regenerative applications.


Subject(s)
Cell Culture Techniques/methods , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Cell Differentiation/physiology , Culture Media/pharmacology , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , RNA, Small Interfering/genetics , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
8.
Sci Rep ; 14(1): 18204, 2024 08 06.
Article in English | MEDLINE | ID: mdl-39107470

ABSTRACT

A limited number of accessible and representative models of human trophoblast cells currently exist for the study of placentation. Current stem cell models involve either a transition through a naïve stem cell state or precise dynamic control of multiple growth factors and small-molecule cues. Here, we demonstrated that a simple five-day treatment of human induced pluripotent stem cells with two small molecules, retinoic acid (RA) and Wnt agonist CHIR 99021 (CHIR), resulted in rapid, synergistic upregulation of CDX2. Transcriptomic analysis of RA + CHIR-treated cells showed high similarity to primary trophectoderm cells. Multipotency was verified via further differentiation towards cells with syncytiotrophoblast or extravillous trophoblast features. RA + CHIR-treated cells were also assessed for the established criteria defining a trophoblast cell model, and they possess all the features necessary to be considered valid. Collectively, our data demonstrate a facile, scalable method for generating functional trophoblast-like cells in vitro to better understand the placenta.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Tretinoin , Trophoblasts , Humans , Trophoblasts/drug effects , Trophoblasts/metabolism , Trophoblasts/cytology , Tretinoin/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Cell Differentiation/drug effects , Pyridines/pharmacology , Female , CDX2 Transcription Factor/metabolism , CDX2 Transcription Factor/genetics , Pyrimidines/pharmacology , Pregnancy , Models, Biological , Cells, Cultured
9.
NPJ Syst Biol Appl ; 10(1): 111, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358384

ABSTRACT

Energy landscapes can provide intuitive depictions of population heterogeneity and dynamics. However, it is unclear whether individual cell behavior, hypothesized to be determined by initial position and noise, is faithfully recapitulated. Using the p21-/Cdk2-dependent quiescence-proliferation decision in breast cancer dormancy as a testbed, we examined single-cell dynamics on the landscape when perturbed by hypoxia, a dormancy-inducing stress. Combining trajectory-based energy landscape generation with single-cell time-lapse microscopy, we found that a combination of initial position and velocity on a p21/Cdk2 landscape, but not position alone, was required to explain the observed cell fate heterogeneity under hypoxia. This is likely due to additional cell state information such as epigenetic features and/or other species encoded in velocity but missing in instantaneous position determined by p21 and Cdk2 levels alone. Here, velocity dependence manifested as inertia: cells with higher cell cycle velocities prior to hypoxia continued progressing along the cell cycle under hypoxia, resisting the change in landscape towards cell cycle exit. Such inertial effects may markedly influence cell fate trajectories in tumors and other dynamically changing microenvironments where cell state transitions are governed by coordination across several biochemical species.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase 2 , Humans , Cell Proliferation/physiology , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cell Cycle/physiology , Cell Line, Tumor , Breast Neoplasms , Female
10.
J Alzheimers Dis ; 99(s2): S281-S297, 2024.
Article in English | MEDLINE | ID: mdl-38393902

ABSTRACT

Background: A strong body of evidence suggests that cerebrovascular pathologies augment the onset and progression of Alzheimer's disease (AD). One distinctive aspect of this cerebrovascular dysfunction is the degeneration of brain pericytes-often overlooked supporting cells of blood-brain barrier endothelium. Objective: The current study investigates the influence of pericytes on gene and protein expressions in the blood-brain barrier endothelium, which is expected to facilitate the identification of pathophysiological pathways that are triggered by pericyte loss and lead to blood-brain barrier dysfunction in AD. Methods: Bioinformatics analysis was conducted on the RNA-Seq expression counts matrix (GSE144474), which compared solo-cultured human blood-brain barrier endothelial cells against endothelial cells co-cultured with human brain pericytes in a non-contact model. We constructed a similar cell culture model to verify protein expression using western blots. Results: The insulin resistance and ferroptosis pathways were found to be enriched. Western blots of the insulin receptor and heme oxygenase expressions were consistent with those observed in RNA-Seq data. Additionally, we observed more than 5-fold upregulation of several genes associated with neuroprotection, including insulin-like growth factor 2 and brain-derived neurotrophic factor. Conclusions: Results suggest that pericyte influence on blood-brain barrier endothelial gene expression confers protection from insulin resistance, iron accumulation, oxidative stress, and amyloid deposition. Since these are conditions associated with AD pathophysiology, they imply mechanisms by which pericyte degeneration could contribute to disease progression.


Subject(s)
Alzheimer Disease , Blood-Brain Barrier , Endothelial Cells , Pericytes , Pericytes/metabolism , Pericytes/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Endothelial Cells/metabolism , Coculture Techniques , Brain/metabolism , Brain/pathology , Cells, Cultured , Receptor, Insulin/metabolism , Receptor, Insulin/genetics , Gene Expression Regulation , Insulin Resistance/physiology
11.
bioRxiv ; 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37961090

ABSTRACT

Entrainment to an external stimulus enables a synchronized oscillatory response across a population of cells, increasing coherent responses by reducing cell-to-cell heterogeneity. It is unclear whether the property of entrainability extends to systems where responses are intrinsic to the individual cell, rather than dependent on coherence across a population of cells. Using a combination of mathematical modeling, time-lapse fluorescence microscopy, and single-cell tracking, we demonstrated that p53 oscillations triggered by DNA double-strand breaks (DSBs) can be entrained with a periodic damage stimulus, despite such synchrony not known to function in effective DNA damage responses. Surprisingly, p53 oscillations were experimentally entrained over a wider range of DSB frequencies than predicted by an established computational model for the system. We determined that recapitulating the increased range of entrainment frequencies required, non-intuitively, a less robust oscillator and wider steady-state valley on the energy landscape. Further, we show that p53 entrainment can lead to altered expression dynamics of downstream targets responsible for cell fate in a manner dependent on target mRNA stability. Overall, this study demonstrates that entrainment can occur in a biological oscillator despite the apparent lack of an evolutionary advantage conferred through synchronized responses and highlights the potential of externally entraining p53 dynamics to reduce cellular variability and synchronize cell-fate responses for therapeutic outcomes.

12.
Trends Biotechnol ; 41(10): 1314-1326, 2023 10.
Article in English | MEDLINE | ID: mdl-37142447

ABSTRACT

Cell therapy offers the potential for curative treatment of cancers. Although T cells have been the predominantly used cell type, natural killer (NK) cells have attracted great attention owing to their ability to kill cancer cells and because they are naturally suitable for allogeneic applications. Upon stimulation by cytokines or activation by a target cell, NK cells proliferate and expand their population. These cytotoxic NK cells can be cryopreserved and used as an off-the-shelf medicine. The production process for NK cells thus differs from that of autologous cell therapies. We briefly outline key biological features of NK cells, review the manufacturing technologies for protein biologics, and discuss their adaptation for developing robust NK cell biomanufacturing processes.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Humans , Killer Cells, Natural/metabolism , Immunotherapy , Neoplasms/therapy , Neoplasms/metabolism , T-Lymphocytes
13.
bioRxiv ; 2023 May 24.
Article in English | MEDLINE | ID: mdl-37292599

ABSTRACT

Energy landscapes can provide intuitive depictions of population heterogeneity and dynamics. However, it is unclear whether individual cell behavior, hypothesized to be determined by initial position and noise, is faithfully recapitulated. Using the p21-/Cdk2-dependent quiescence-proliferation decision in breast cancer dormancy as a testbed, we examined single-cell dynamics on the landscape when perturbed by hypoxia, a dormancy-inducing stress. Combining trajectory-based energy landscape generation with single-cell time-lapse microscopy, we found that initial position on a p21/Cdk2 landscape did not fully explain the observed cell-fate heterogeneity under hypoxia. Instead, cells with higher cell state velocities prior to hypoxia, influenced by epigenetic parameters, tended to remain proliferative under hypoxia. Thus, the fate decision on this landscape is significantly influenced by "inertia", a velocity-dependent ability to resist directional changes despite reshaping of the underlying landscape, superseding positional effects. Such inertial effects may markedly influence cell-fate trajectories in tumors and other dynamically changing microenvironments.

14.
Int J Pharm ; 635: 122744, 2023 Mar 25.
Article in English | MEDLINE | ID: mdl-36804522

ABSTRACT

When exposed to an external electric field, lipid bilayer membranes are subject to increased permeability through the generation of pores. Combining this phenomenon, known as electroporation, with liposomal drug delivery offers the added benefit of on-demand release of the liposomal cargo. In previous studies, the maximum percent drug release when exposing liposomes to a pulsed electric field has not surpassed 30%, indicating most of the drug is still retained in the liposomes. Here we showed that by modulating the fluidity of the liposome membrane through appropriate selection of the primary lipid, as well as the addition of other fluidity modulating components such as cholesterol and biotinylated lipid, the electroporation-induced percent release could be increased to over 50%. In addition to improved induced release from liposomes in suspension, biomaterial scaffold-bound liposomes were developed. Electroporation-induced protein release from this solid phase was verified after performing further optimization of the liposome formulation to achieve increased stability at physiological temperatures. Collectively, this work advances the ability to achieve efficient electroporation-induced liposomal drug delivery, which has the potential to be used in concert with other clinical applications of electroporation, such as gene electrotransfer and irreversible electroporation (IRE), in order to synergistically increase treatment efficacy.


Subject(s)
Drug Delivery Systems , Liposomes , Drug Liberation , Lipid Bilayers , Electroporation , Suspensions
15.
Biotechnol Appl Biochem ; 59(2): 88-96, 2012.
Article in English | MEDLINE | ID: mdl-23586789

ABSTRACT

Human embryonic stem cells (hESCs) hold potential in the field of tissue engineering, given their capacity for both limitless self-renewal and differentiation to any adult cell type. However, several limitations, including the ability to expand undifferentiated cells and efficiently direct differentiation at scales needed for commercial cell production, prevent realizing the potential of hESCs in tissue engineering. Numerous studies have illustrated that three-dimensional (3D) culture systems provide microenvironmental cues that affect hESC pluripotency and differentiation fates, but little is known about how 3D culture affects cell expansion. Here, we have used a 3D microwell array to model the differences in hESC growth kinetics and metabolism in two-dimensional (2D) versus 3D cultures. Our results demonstrated that 3D microwell culture reduced hESC size and proliferative capacity, and impacted cell cycle dynamics, lengthening the G1 phase and shortening the G2/M phase of the cell cycle. However, glucose and lactate metabolism were similar in 2D and 3D cultures. Elucidating the effects of 3D culture on growth and metabolism of hESCs may facilitate efforts for developing integrated, scalable cell expansion and differentiation processes with these cells.


Subject(s)
Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Embryonic Stem Cells/physiology , Bioengineering , Cell Cycle/physiology , Cell Growth Processes/physiology , Cell Line , Cell Size , Collagen/chemistry , Drug Combinations , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Glucose/metabolism , Humans , Kinetics , Lactic Acid/metabolism , Laminin/chemistry , Proteoglycans/chemistry
16.
Cell Mol Bioeng ; 15(1): 99-114, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35096187

ABSTRACT

INTRODUCTION: In vivo, breast cancer cells spend on average 3-7 days adhered to the endothelial cells inside the vascular lumen before entering the brain. IL-1ß is one of the highly upregulated molecules in brain-seeking triple negative breast cancer (TNBC) cells. In this study, the effect of IL-1ß on the blood-brain barrier (BBB) and astrocytes and its role in transmigration of TNBC cells were evaluated. METHODS: The effect of IL-1ß on transendothelial electrical resistance, gene and protein expression of human induced pluripotent stem cell-derived brain-specific microvascular endothelial-like cells (iBMECs) was studied. Transport of IL-1ß across the iBMEC layer was investigated and the effect of IL-1ß treatment of astrocytes on their cytokine and chemokine secretome was evaluated with a cytokine membrane array. Using BBB-on-a-chip devices, transmigration of MDA-MB-231 cells and their brain-seeking variant (231BR) across the iBMECs was studied, and the effect of an IL-1ß neutralizing antibody on TNBC cell transmigration was investigated. RESULTS: We showed that IL-1ß reduces BBB integrity and induces endothelial-to-mesenchymal transition in iBMECs. IL-1ß crosses the iBMEC layer and induces secretion of multiple chemokines by astrocytes, which can enhance TNBC cell transmigration across the BBB. Transmigration assays in a BBB-on-a-chip device showed that 231BR cells have a higher rate of transmigration across the iBMECs compared to MDA-MB-231 cells, and IL-1ß pretreatment of BBB-on-a-chip devices increases the number of transmigrated MDA-MB-231 cells. Finally, we demonstrated that neutralizing IL-1ß reduces the rate of 231BR cell transmigration. CONCLUSION: IL-1ß plays a significant role in transmigration of brain-seeking TNBC cells across the BBB. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s12195-021-00710-y.

17.
Nat Commun ; 13(1): 4152, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35858965

ABSTRACT

Site-specific recombination (SSR) is an important tool in synthetic biology, but its applications are limited by the inability to predictably tune SSR reaction rates. Facile rate manipulation could be achieved by modifying the DNA substrate sequence; however, this approach lacks rational design principles. Here, we develop an integrated experimental and computational method to engineer the DNA attachment sequence attP for predictably modulating the inversion reaction mediated by the recombinase Bxb1. After developing a qPCR method to measure SSR reaction rate, we design, select, and sequence attP libraries to inform a machine-learning model that computes Bxb1 inversion rate as a function of attP sequence. We use this model to predict reaction rates of attP variants in vitro and demonstrate their utility in gene circuit design in Escherichia coli. Our high-throughput, model-guided approach for rationally tuning SSR reaction rates enhances our understanding of recombinase function and expands the synthetic biology toolbox.


Subject(s)
Bacteriophages , Recombination, Genetic , Bacteriophages/genetics , Base Sequence , DNA/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Integrases/genetics , Integrases/metabolism , Recombinases/genetics , Recombinases/metabolism
18.
Biomacromolecules ; 12(6): 1998-2007, 2011 Jun 13.
Article in English | MEDLINE | ID: mdl-21504222

ABSTRACT

We report an approach to the fabrication and selective functionalization of amine-reactive polymer multilayers on the surfaces of 3-D polyurethane-based microwell cell culture arrays. "Reactive" layer-by-layer assembly of multilayers using branched polyethyleneimine (BPEI) and the azlactone-functionalized polymer poly(2-vinyl-4,4'-dimethylazlactone) (PVDMA) yielded film-coated microwell arrays that could be chemically functionalized postfabrication by treatment with different amine-functionalized macromolecules or small molecule primary amines. Treatment of film-coated arrays with the small molecule amine d-glucamine resulted in microwell surfaces that resisted the adhesion and proliferation of mammalian fibroblast cells in vitro. These and other experiments demonstrated that it was possible to functionalize different structural features of these arrays in a spatially resolved manner to create dual-functionalized substrates (e.g., to create arrays having either (i) azlactone-functionalized wells, with regions between the wells functionalized with glucamine or (ii) substrates with spatially resolved regions of two different cationic polymers). In particular, spatial control over glucamine functionalization yielded 3-D substrates that could be used to confine cell attachment and growth to microwells for periods of up to 28 days and support the 3-D culture of arrays of cuboidal cell clusters. These approaches to dual functionalization could prove useful for the long-term culture and maintenance of cell types for which the presentation of specific and chemically well-defined 3-D culture environments is required for control over cell growth, differentiation, and other important behaviors. More generally, our approach provides methods for the straightforward chemical functionalization of otherwise unreactive topographically patterned substrates that could prove to be useful in a range of other fundamental and applied contexts.


Subject(s)
Amines/chemistry , Coated Materials, Biocompatible/chemistry , Lactones/chemistry , Polyethyleneimine/chemistry , Polyurethanes/chemistry , Amines/metabolism , Cell Adhesion , Cell Culture Techniques/methods , Cell Proliferation , Coated Materials, Biocompatible/metabolism , Fluorescence , HEK293 Cells , Humans , Lactones/metabolism , Polyethyleneimine/metabolism , Surface Properties
19.
J Biomed Mater Res B Appl Biomater ; 109(12): 2117-2130, 2021 12.
Article in English | MEDLINE | ID: mdl-33983681

ABSTRACT

The principal cause of cancer deaths is the residual disease, which eventually results in metastases. Certain metastases are induced by disseminated dormancy-capable single cancer cells that can reside within the body undetected for months to years. Awakening of the dormant cells starts a cascade resulting in the patient's demise. Despite its established clinical significance, dormancy research and its clinical translation have been hindered by lack of in vitro models that can identify, isolate, and analyze dormancy-capable cells. We have previously shown that immobilization of cells in a stiff microenvironment induces dormancy in dormancy-capable cell lines. In this communication, we present a novel biomaterial and an in vitro immobilization method to isolate, analyze, and efficiently recover dormancy-capable cancer cells. MCF-7, MDA-MB-231, and MDA-MB-468 cells were individually coated with agarose using a microfluidic flow-focusing device. Coated cells were then immobilized in a rigid and porous silica gel. Dormancy induction by this process was validated by decreased Ki-67 expression, increased p38/ERK activity ratio, and reduced expression of CDK-2, cyclins D1, and E1. We showed that we can reliably and repeatedly induce dormancy in dormancy-capable MCF-7 cells and enhance the dormancy-capable sub-population in MDA-MB-231 cells. As expected, dormancy-resistant MDA-MB-468 cells did not survive immobilization. The dormant cells could be awakened on demand, by digesting the agarose gel in situ, and efficiently recovered by magnetically separating the silica gel, making the cells available for downstream analysis and testing. The awakened cells were shown to regain motility immediately, proliferating, and migrating normally.


Subject(s)
Biocompatible Materials , Neoplasms , Humans , MCF-7 Cells , Neoplasms/metabolism , Sepharose/pharmacology , Silicon Dioxide/pharmacology
20.
Adv Healthc Mater ; 10(13): e2001368, 2021 07.
Article in English | MEDLINE | ID: mdl-34050609

ABSTRACT

Advanced stage ovarian cancer is challenging to treat due to widespread seeding of tumor spheroids throughout the mesothelial lining of the peritoneal cavity. In this work, a therapeutic strategy using graphene nanoribbons (GNR) functionalized with 4-arm polyethylene glycol (PEG) and chlorin e6 (Ce6), a sonosensitizer, to target metastatic ovarian cancer spheroids is reported. GNR-PEG-Ce6 adsorbs onto the spheroids and disrupts their adhesion to extracellular matrix proteins or LP-9 mesothelial cells. Furthermore, for spheroids that do adhere, GNR-PEG-Ce6 delays spheroid disaggregation and spreading as well as mesothelial clearance, key metastatic processes following adhesion. Owing to the sonodynamic effects of Ce6 and its localized delivery via the biomaterial, GNR-PEG-Ce6 can kill ovarian cancer spheroids adhered to LP-9 cell monolayers when combined with mild ultrasound irradiation. The interaction with GNR-PEG-Ce6 also loosens cell-cell adhesions within the spheroids, rendering them more susceptible to treatment with the chemotherapeutic agents cisplatin and paclitaxel, which typically have difficulty in penetrating ovarian cancer spheroids. Thus, this material can facilitate effective chemotherapeutic and sonodynamic combination therapies. Finally, the adhesion inhibiting and sonodynamic effects of GNR-PEG-Ce6 are also validated with tumor spheroids derived from the ascites fluid of ovarian cancer patients, providing evidence of the translational potential of this biomaterial approach.


Subject(s)
Graphite , Nanotubes, Carbon , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Humans , Ovarian Neoplasms/therapy , Spheroids, Cellular
SELECTION OF CITATIONS
SEARCH DETAIL