Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bioeng Transl Med ; 7(2): e10283, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35600639

RESUMO

Induced neural stem cells (iNSCs) have emerged as a promising therapeutic platform for glioblastoma (GBM). iNSCs have the innate ability to home to tumor foci, making them ideal carriers for antitumor payloads. However, the in vivo persistence of iNSCs limits their therapeutic potential. We hypothesized that by encapsulating iNSCs in the FDA-approved, hemostatic matrix FLOSEAL®, we could increase their persistence and, as a result, therapeutic durability. Encapsulated iNSCs persisted for 95 days, whereas iNSCs injected into the brain parenchyma persisted only 2 weeks in mice. Two orthotopic GBM tumor models were used to test the efficacy of encapsulated iNSCs. In the GBM8 tumor model, mice that received therapeutic iNSCs encapsulated in FLOSEAL® survived 30 to 60 days longer than mice that received nonencapsulated cells. However, the U87 tumor model showed no significant differences in survival between these two groups, likely due to the more solid and dense nature of the tumor. Interestingly, the interaction of iNSCs with FLOSEAL® appears to downregulate some markers of proliferation, anti-apoptosis, migration, and therapy which could also play a role in treatment efficacy and durability. Our results demonstrate that while FLOSEAL® significantly improves iNSC persistence, this alone is insufficient to enhance therapeutic durability.

2.
Mol Cancer Ther ; 20(11): 2291-2301, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34433662

RESUMO

Converting human fibroblasts into personalized induced neural stem cells (hiNSC) that actively seek out tumors and deliver cytotoxic agents is a promising approach for treating cancer. Herein, we provide the first evidence that intravenously-infused hiNSCs secreting cytotoxic agent home to and suppress the growth of non-small cell lung cancer (NSCLC) and triple-negative breast cancer (TNBC). Migration of hiNSCs to NSCLC and TNBC in vitro was investigated using time-lapse motion analysis, which showed directional movement of hiNSCs to both tumor cell lines. In vivo, migration of intravenous hiNSCs to orthotopic NSCLC or TNBC tumors was determined using bioluminescent imaging (BLI) and immunofluorescent post-mortem tissue analysis, which indicated that hiNSCs colocalized with tumors within 3 days of intravenous administration and persisted through 14 days. In vitro, efficacy of hiNSCs releasing cytotoxic TRAIL (hiNSC-TRAIL) was monitored using kinetic imaging of co-cultures, in which hiNSC-TRAIL therapy induced rapid killing of both NSCLC and TNBC. Efficacy was determined in vivo by infusing hiNSC-TRAIL or control cells intravenously into mice bearing orthotopic NSCLC or TNBC and tracking changes in tumor volume using BLI. Mice treated with intravenous hiNSC-TRAIL showed a 70% or 72% reduction in NSCLC or TNBC tumor volume compared with controls within 14 or 21 days, respectively. Safety was assessed by hematology, blood chemistry, and histology, and no significant changes in these safety parameters was observed through 28 days. These results indicate that intravenous hiNSCs-TRAIL seek out and kill NSCLC and TNBC tumors, suggesting a potential new strategy for treating aggressive peripheral cancers.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Células-Tronco Neurais/transplante , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Humanos , Camundongos
3.
Bioeng Transl Med ; 6(1): e10171, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33532581

RESUMO

In this study, we take an important step toward clinical translation by generating the first canine-induced neural stem cells (iNSCs). We explore key aspects of scale-up, persistence, and safety of personalized iNSC therapy in autologous canine surgery models. iNSCs are a promising new approach to treat aggressive cancers of the brain, including the deadly glioblastoma. Created by direct transdifferentiation of fibroblasts, iNSCs are known to migrate through the brain, track down invasive cancer foci, and deliver anticancer payloads that significantly reduce tumor burden and extend survival of tumor-bearing mice. Here, skin biopsies were collected from canines and converted into the first personalized canine iNSCs engineered to carry TNFα-related apoptosis-inducing ligand (TRAIL) and thymidine kinase (TK), as well as magnetic resonance imaging (MRI) contrast agents for in vivo tracking. Time-lapse analysis showed canine iNSCs efficiently migrate to human tumor cells, and cell viability assays showed both TRAIL and TK monotherapy markedly reduced tumor growth. Using intraoperative navigation and two delivery methods to closely mimic human therapy, canines received autologous iNSCs either within postsurgical cavities in a biocompatible matrix or via a catheter placed in the lateral ventricle. Both strategies were well tolerated, and serial MRI showed hypointense regions at the implant sites that remained stable through 86 days postimplant. Serial fluid sample testing following iNSC delivery showed the bimodal personalized therapy was well tolerated, with no iNSC-induced abnormal tissue pathology. Overall, this study lays an important foundation as this promising personalized cell therapy advances toward human patient testing.

4.
J Control Release ; 324: 1-16, 2020 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-32407745

RESUMO

Glioblastoma multiforme (GBM) is impossible to fully remove surgically and almost always recurs at the borders of the resection cavity, while systemic delivery of therapeutic drug levels to the brain tumour is limited by the blood-brain barrier. This research describes the development of a novel formulation of Irinotecan-loaded Drug Eluting Seeds (iDES) for insertion into the margin of the GBM resection cavity to provide a sustained high local dose with reduced systemic toxicities. We used primary GBM cells from both the tumour core and Brain Around the Tumour tissue from recurrent GBM patients to demonstrate that irinotecan is more effective than temozolomide. Irinotecan had a 75% response rate, while only 50% responded to temozolomide. With temozolomide the cell viability was never below 80% whereas irinotecan achieved cell viabilities of less than 44%. The iDES were manufactured using a hot melt extrusion process with accurate irinotecan drug loadings and the same cytotoxicity as unformulated irinotecan. The iDES released irinotecan in a sustained fashion for up to 7 days. However, only the 30, 40 and 50% w/w loaded iDES formulations released the 300 to 1000 µg of irinotecan needed to be effective in vivo. The 30 and 40% w/w iDES formulations containing 10% plasticizer and either 60 or 50% PLGA prolonged survival from 27 to 70 days in a GBM xenograft mouse resection model with no sign of tumour recurrence. The 30% w/w iDES formulations showed equivalent toxicity to a placebo in non-tumour bearing mice. This innovative drug delivery approach could transform the treatment of recurrent GBM patients by improving survival and reducing toxicity.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Preparações Farmacêuticas , Animais , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Humanos , Irinotecano , Camundongos , Temozolomida
5.
Mol Ther ; 28(4): 1056-1067, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32109370

RESUMO

Pre-clinical and clinical studies have shown that engineered tumoricidal neural stem cells (tNSCs) are a promising treatment strategy for the aggressive brain cancer glioblastoma (GBM). Yet, stabilizing human tNSCs within the surgical cavity following GBM resection is a significant challenge. As a critical step toward advancing engineered human NSC therapy for GBM, we used a preclinical variant of the clinically utilized NSC line HB1.F3.CD and mouse models of human GBM resection/recurrence to identify a polymeric scaffold capable of maximizing the transplant, persistence, and tumor kill of NSC therapy for post-surgical GBM. Using kinetic bioluminescence imaging, we found that tNSCs delivered into the mouse surgical cavity wall by direct injection persisted only 3 days. We found that delivery of tNSCs into the cavity on nanofibrous electrospun poly-l-lactic acid scaffolds extended tNSC persistence to 8 days. Modifications to fiber surface coating, diameter, and morphology of the scaffold failed to significantly extend tNSC persistence in the cavity. In contrast, tNSCs delivered into the post-operative cavity on gelatin matrices (GEMs) persisted 8-fold longer as compared to direct injection. GEMs remained permissive to tumor-tropic homing, as tNSCs migrated off the scaffolds and into invasive tumor foci both in vitro and in vivo. To mirror envisioned human brain tumor trials, we engineered tNSCs to express the prodrug/enzyme thymidine kinase (tNSCstk) and transplanted the therapeutic cells in the post-operative cavity of mice bearing resected orthotopic patient-derived GBM xenografts. Following administration of the prodrug ganciclovir, residual tumor volumes in mice receiving GEM/tNSCs were reduced by 10-fold at day 35, and median survival was extended from 31 to 46 days. Taken together, these data begin to define design parameters for effective scaffold/tNSC composites and suggest a new approach to maximizing the efficacy of tNSC therapy in human patient trials.


Assuntos
Neoplasias Encefálicas/terapia , Ganciclovir/administração & dosagem , Glioblastoma/terapia , Células-Tronco Neurais/transplante , Timidina Quinase/metabolismo , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Linhagem Celular Tumoral , Terapia Combinada , Ganciclovir/farmacologia , Glioblastoma/patologia , Glioblastoma/cirurgia , Humanos , Medições Luminescentes , Camundongos , Células-Tronco Neurais/metabolismo , Poliésteres/química , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacologia , Alicerces Teciduais/química , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
6.
PLoS One ; 13(7): e0198596, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29990322

RESUMO

BACKGROUND: Cytotoxic neural stem cells (NSCs) have emerged as a promising treatment for Medulloblastoma (MB), the most common malignant primary pediatric brain tumor. The lack of accurate pre-clinical models incorporating surgical resection and tumor recurrence limits advancement in post-surgical MB treatments. Using cell lines from two of the 5 distinct MB molecular sub-groups, in this study, we developed an image-guided mouse model of MB surgical resection and investigate intra-cavity NSC therapy for post-operative MB. METHODS: Using D283 and Daoy human MB cells engineered to express multi-modality optical reporters, we created the first image-guided resection model of orthotopic MB. Brain-derived NSCs and novel induced NSCs (iNSCs) generated from pediatric skin were engineered to express the pro-drug/enzyme therapy thymidine kinase/ganciclovir, seeded into the post-operative cavity, and used to investigate intra-cavity therapy for post-surgical MB. RESULTS: We found that surgery reduced MB volumes by 92%, and the rate of post-operative MB regrowth increased 3-fold compared to pre-resection growth. Real-time imaging showed NSCs rapidly homed to MB, migrating 1.6-fold faster and 2-fold farther in the presence of tumors, and co-localized with MB present in the contra-lateral hemisphere. Seeding of cytotoxic NSCs into the post-operative surgical cavity decreased MB volumes 15-fold and extended median survival 133%. As an initial step towards novel autologous therapy in human MB patients, we found skin-derived iNSCs homed to MB cells, while intra-cavity iNSC therapy suppressed post-surgical tumor growth and prolonged survival of MB-bearing mice by 123%. CONCLUSIONS: We report a novel image-guided model of MB resection/recurrence and provide new evidence of cytotoxic NSCs/iNSCs delivered into the surgical cavity effectively target residual MB foci.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Meduloblastoma/terapia , Recidiva Local de Neoplasia/prevenção & controle , Células-Tronco Neurais/transplante , Cirurgia Assistida por Computador/métodos , Animais , Encéfalo/patologia , Encéfalo/cirurgia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Diferenciação Celular , Movimento Celular , Modelos Animais de Doenças , Terapia Enzimática/métodos , Células Epiteliais/citologia , Ganciclovir/farmacologia , Humanos , Injeções Intralesionais , Meduloblastoma/mortalidade , Meduloblastoma/patologia , Meduloblastoma/cirurgia , Camundongos , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/cirurgia , Células-Tronco Neurais/citologia , Pró-Fármacos/farmacologia , Pele/citologia , Análise de Sobrevida , Timidina Quinase/genética , Timidina Quinase/metabolismo
7.
Methods Mol Biol ; 1831: 49-58, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30051424

RESUMO

Engineered stem cells have recently entered clinical trials as therapeutic agents for treating glioblastoma foci that remain after primary brain tumor resection. However, efficient delivery of anti-cancer mesenchymal stem cells (MSCs) into the resection cavity remains a potential obstacle to therapeutic efficacy in humans. Direct injection quickly leads to significant stem cell loss and poor tumor killing. Recent reports have shown that biodegradable scaffolds improve MSC persistence and restore therapeutic potential. Here, we describe a method for the delivery of therapeutic MSCs on biodegradable fibrin scaffolds into the resection cavity to treat postoperative brain cancer.


Assuntos
Materiais Biocompatíveis/farmacologia , Neoplasias Encefálicas/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Alicerces Teciduais/química , Animais , Neoplasias Encefálicas/cirurgia , Morte Celular , Linhagem Celular Tumoral , Humanos , Camundongos Nus
8.
J Vis Exp ; (137)2018 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-30059037

RESUMO

Glioblastoma (GBM), the most common and aggressive primary brain cancer, carries a life expectancy of 12-15 months. The short life expectancy is due in part to the inability of the current treatment, consisting of surgical resection followed by radiation and chemotherapy, to eliminate invasive tumor foci. Treatment of these foci may be improved with tumoricidal human mesenchymal stem cells (MSCs). MSCs exhibit potent tumor tropism and can be engineered to express therapeutic proteins that kill tumor cells. Advancements in preclinical models indicate that surgical resection induces premature MSC loss and reduces therapeutic efficacy. Efficacy of MSC treatment can be improved by seeding MSCs on a biodegradable poly(lactic acid) (PLA) scaffold. MSC delivery into the surgical resection cavity on a PLA scaffold restores cell retention, persistence, and tumor killing. To study the effects of MSC-seeded PLA implantation on GBM, an accurate preclinical model is needed. Here we provide a preclinical surgical protocol for image-guided tumor resection of GBM in immune-deficient mice followed by MSC-seeded scaffold implantation. MSCs are engineered with lentiviral constructs to constitutively express and secrete therapeutic TNFα-related apoptosis-inducing ligand (TRAIL) as well as green fluorescent protein (GFP) to allow fluorescent tracking. Similarly, the U87 tumor cells are engineered to express mCherry and firefly luciferase, providing dual fluorescent/luminescent tracking. While currently used for investigating stem cell mediated delivery of therapeutics, this protocol could be modified to investigate the impact of surgical resection on other GBM interventions.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/cirurgia , Glioblastoma/diagnóstico por imagem , Glioblastoma/cirurgia , Células-Tronco/metabolismo , Alicerces Teciduais/química , Animais , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Células-Tronco/patologia
9.
Mol Pharm ; 15(3): 1309-1318, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29342360

RESUMO

The primary cause of mortality for glioblastoma (GBM) is local tumor recurrence following standard-of-care therapies, including surgical resection. With most tumors recurring near the site of surgical resection, local delivery of chemotherapy at the time of surgery is a promising strategy. Herein drug-loaded polymer scaffolds with two distinct degradation profiles were fabricated to investigate the effect of local drug delivery rate on GBM recurrence following surgical resection. The novel biopolymer, acetalated dextran (Ace-DEX), was compared with commercially available polyester, poly(l-lactide) (PLA). Steady-state doxorubicin (DXR) release from Ace-DEX scaffolds was found to be faster when compared with scaffolds composed of PLA, in vitro. This increased drug release rate translated to improved therapeutic outcomes in a novel surgical model of orthotopic glioblastoma resection and recurrence. Mice treated with DXR-loaded Ace-DEX scaffolds (Ace-DEX/10DXR) resulted in 57% long-term survival out to study completion at 120 days compared with 20% survival following treatment with DXR-loaded PLA scaffolds (PLA/10DXR). Additionally, all mice treated with PLA/10DXR scaffolds exhibited disease progression by day 38, as defined by a 5-fold growth in tumor bioluminescent signal. In contrast, 57% of mice treated with Ace-DEX/10DXR scaffolds displayed a reduction in tumor burden, with 43% exhibiting complete remission. These results underscore the importance of polymer choice and drug release rate when evaluating local drug delivery strategies to improve prognosis for GBM patients undergoing tumor resection.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Glioblastoma/tratamento farmacológico , Recidiva Local de Neoplasia/prevenção & controle , Acetais/química , Animais , Antibióticos Antineoplásicos/farmacocinética , Encéfalo/patologia , Encéfalo/cirurgia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/farmacocinética , Dextranos/química , Progressão da Doença , Doxorrubicina/farmacocinética , Liberação Controlada de Fármacos , Glioblastoma/patologia , Glioblastoma/cirurgia , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Mol Biol Cell ; 28(14): 1894-1900, 2017 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-28450452

RESUMO

A number of innovative methods exist to measure cell-matrix adhesive forces, but they have yet to accurately describe and quantify the intricate interplay of a cell and its fibrous extracellular matrix (ECM). In cardiovascular pathologies, such as aortic aneurysm, new knowledge on the involvement of cell-matrix forces could lead to elucidation of disease mechanisms. To better understand this dynamics, we measured primary human aortic single smooth muscle cell (SMC) forces using nanonet force microscopy in both inside-out (I-O intrinsic contractility) and outside-in (O-I external perturbation) modes. For SMC populations, we measured the I-O and O-I forces to be 12.9 ± 1.0 and 57.9 ± 2.5 nN, respectively. Exposure of cells to oxidative stress conditions caused a force decrease of 57 and 48% in I-O and O-I modes, respectively, and an increase in migration rate by 2.5-fold. Finally, in O-I mode, we cyclically perturbed cells at constant strain of varying duration to simulate in vivo conditions of the cardiac cycle and found that I-O forces decrease with increasing duration and O-I forces decreased by half at shorter cycle times. Thus our findings highlight the need to study forces exerted and felt by cells simultaneously to comprehensively understand force modulation in cardiovascular disease.


Assuntos
Aorta/citologia , Microscopia de Força Atômica/métodos , Miócitos de Músculo Liso/citologia , Fenômenos Biomecânicos/fisiologia , Células Cultivadas , Matriz Extracelular , Humanos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo
11.
Biophys J ; 111(1): 197-207, 2016 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-27410747

RESUMO

The influence of physical forces exerted by or felt by cells on cell shape, migration, and cytoskeleton arrangement is now widely acknowledged and hypothesized to occur due to modulation of cellular inside-out forces in response to changes in the external fibrous environment (outside-in). Our previous work using the non-electrospinning Spinneret-based Tunable Engineered Parameters' suspended fibers has revealed that cells are able to sense and respond to changes in fiber curvature and structural stiffness as evidenced by alterations to focal adhesion cluster lengths. Here, we present the development and application of a suspended nanonet platform for measuring C2C12 mouse myoblast forces attached to fibers of three diameters (250, 400, and 800 nm) representing a wide range of structural stiffness (3-50 nN/µm). The nanonet force microscopy platform measures cell adhesion forces in response to symmetric and asymmetric external perturbation in single and cyclic modes. We find that contractility-based, inside-out forces are evenly distributed at the edges of the cell, and that forces are dependent on fiber structural stiffness. Additionally, external perturbation in symmetric and asymmetric modes biases cell-fiber failure location without affecting the outside-in forces of cell-fiber adhesion. We then extend the platform to measure forces of (1) cell-cell junctions, (2) single cells undergoing cyclic perturbation in the presence of drugs, and (3) cancerous single-cells transitioning from a blebbing to a pseudopodial morphology.


Assuntos
Fenômenos Mecânicos , Microscopia de Força Atômica , Mioblastos/citologia , Nanotecnologia , Animais , Fenômenos Biomecânicos , Adesão Celular , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , Junções Intercelulares/metabolismo , Camundongos , Nanofibras
12.
Methods ; 99: 37-43, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26314280

RESUMO

Cancers of the brain remain one of the greatest medical challenges. Traditional surgery and chemo-radiation therapy are unable to eradicate diffuse cancer cells and tumor recurrence is nearly inevitable. In contrast to traditional regenerative medicine applications, engineered neural stem cells (NSCs) are emerging as a promising new therapeutic strategy for cancer therapy. The tumor-homing properties allow NSCs to access both primary and invasive tumor foci, creating a novel delivery platform. NSCs engineered with a wide array of cytotoxic agents have been found to significantly reduce tumor volumes and markedly extend survival in preclinical models. With the recent launch of new clinical trials, the potential to successfully manage cancer in human patients with cytotoxic NSC therapy is moving closer to becoming a reality.


Assuntos
Neoplasias/terapia , Células-Tronco Neurais/transplante , Animais , Técnicas de Cultura de Células , Engenharia Celular , Movimento Celular , Reprogramação Celular , Humanos , Células-Tronco Neurais/fisiologia
13.
Biofabrication ; 6(4): 045006, 2014 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-25264874

RESUMO

The role of physical forces in disease onset and progression is widely accepted and this knowledge presents an alternative route to investigating disease models. Recently, numerous force measurement techniques have been developed to probe single and multi-cell behavior. While these methods have yielded fundamental insights, they are yet unable to capture the fibrous extra-cellular matrix biophysical interactions, involving parameters of curvature, structural stiffness (N m(-1)), alignment and hierarchy, which have been shown to play key roles in disease and developmental biology. Using a highly aggressive glioma model (DBTRG-05MG), we present a platform technology to quantify single cell force modulation (both inside-out and outside-in) with and without the presence of a cytoskeleton altering drug (cytochalasin D) using suspended and aligned fiber networks (nanonets) beginning to represent the aligned glioma environment. The nanonets fused in crisscross patterns were manufactured using the non-electrospinning spinneret based tunable engineering parameters technique. We demonstrate the ability to measure contractile single cell forces exerted by glioma cells attached to and migrating along the fiber axis (inside-out). This is followed by a study of force response of glioma cells attached to two parallel fibers using a probe deflecting the leading fiber (outside-in). The forces are calculated using beam deflection within the elastic limit. Our data shows that cytochalasin D compromises the spreading area of single glioma cells, eventually decreasing their 'inside-out' contractile forces, and 'outside-in' force response to external strain. Most notably, for the first time, we demonstrate the feasibility of using physiologically relevant aligned fiber networks as ultra-sensitive force (∼nanoNewtons) probes for investigating drug response and efficacy in disease models at the single cell resolution.


Assuntos
Citocalasina D/farmacologia , Citoesqueleto , Nanofibras/química , Análise de Célula Única/métodos , Fenômenos Biomecânicos/fisiologia , Linhagem Celular Tumoral , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/fisiologia , Humanos , Microscopia , Imagem com Lapso de Tempo
14.
Integr Biol (Camb) ; 5(8): 1036-44, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23817451

RESUMO

Investigating the mechanistic influence of the tumor microenvironment on cancer cell migration and membrane blebbing is crucial in the understanding and eventual arrest of cancer metastasis. In this study, we investigate the effect of suspended and aligned nanofibers on the glioma cytoskeleton, cell shape, migration and plasma membrane blebbing dynamics using a non-electrospinning fiber-manufacturing platform. Cells attached in repeatable shapes of spindle on single fibers, rectangular on two parallel fibers and polygonal on intersecting fibers. Structural stiffness (N m(-1)) of aligned and suspended nanofibers (average diameter: 400 nm, length: 4, 6, and 10 mm) was found to significantly alter the migration speed with higher migration on lower stiffness fibers. For cells attached to fibers and exhibiting blebbing, an increase in cellular spread area resulted in both reduced bleb count and bleb size with an overall increase in cell migration speed. Blebs no longer appeared past a critical cellular spread area of approximately 1400 µm(2). Our results highlighting the influence of the mechanistic environment on the invasion dynamics of glioma cells add to the understanding of how biophysical components influence glioma cell migration and blebbing dynamics.


Assuntos
Neoplasias Encefálicas/metabolismo , Movimento Celular/fisiologia , Forma Celular , Glioma/metabolismo , Nanofibras/química , Neoplasias Encefálicas/patologia , Adesão Celular , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Glioma/patologia , Humanos , Metástase Neoplásica
15.
Acta Biomater ; 9(7): 7169-77, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23567946

RESUMO

In the body, cells dynamically respond to chemical and mechanical cues from the extracellular matrix (ECM), yet precise mechanisms by which biophysical parameters (stiffness, topography and alignment) affect cell behavior remain unclear. Here, highly aligned and suspended multilayer polystyrene (PS) nanofiber scaffolds are used to study biophysical influences on focal adhesion complex (FAC) arrangement and associated migration behavior of mouse C2C12 cells arranged in specific shapes: spindle, parallel and polygonal. Furthermore, the role of cytoskeletal-altering drugs including blebbistatin, nocodazole and cytochalasin-D on FAC formation and migratory behavior is investigated. For the first time, this work reports that cells on suspended fiber networks, including cells with administered drugs, elongated along the fiber axes and developed longer (∼ 4×) and more concentrated FAC clusters compared to cells on flat PS control substrates. Additionally, substrate designs which topographically restrict sites of cell attachment and align adhesions were found to promote higher migration speeds (spindle: 52µmh(-1), parallel: 39µmh(-1), polygonal: 25µmh(-1), flat: 32µmh(-1)). This work demonstrates that suspended fiber topography-induced concentration of FACs along fiber axes generates increased migration potential as opposed to flat surfaces, which diffuse and randomly orient adhesions.


Assuntos
Matriz Extracelular/química , Mecanotransdução Celular/fisiologia , Mioblastos/citologia , Mioblastos/fisiologia , Nanofibras/química , Poliestirenos/química , Alicerces Teciduais , Animais , Materiais Biomiméticos/química , Adesão Celular/fisiologia , Linhagem Celular , Movimento Celular/fisiologia , Tamanho Celular , Desenho de Equipamento , Análise de Falha de Equipamento , Adesões Focais/fisiologia , Teste de Materiais , Camundongos , Nanofibras/ultraestrutura , Propriedades de Superfície
16.
Biomed Sci Instrum ; 48: 202-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22846284

RESUMO

According to the Centers for Disease Control and Prevention and the Institute of Medicine of the National Academies, insufficient sleep has become a public health epidemic. Approximately 50-70 million adults (20 years or older) suffer from some disorder of sleep and wakefulness, hindering daily functioning and adversely affecting health and longevity. Melatonin, a naturally produced hormone which plays a role in sleep-wake regulation, is currently offered as an over-the-counter sleep aid. However, the effects of melatonin on the sleep-wake cycle are incompletely understood. The goal of this modeling study was to incorporate the effects of exogenous melatonin administration into a mathematical model of the human sleep-wake switch. The model developed herein adds a simple kinetic model of the MT1 melatonin receptor to an existing model which simulates the interactions of different neuronal groups thought to be involved in sleep-wake regulation. Preliminary results were obtained by simulating the effects of an exogenous melatonin dose typical of over-the-counter sleep aids. The model predicted an increase in homeostatic sleep drive and a resulting alteration in circadian rhythm consistent with experimental results. The time of melatonin administration was also observed to have a strong influence on the sleep-wake effects elicited, which is also consistent with prior experimental findings.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...