Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 75(6): 1113-22, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25634210

ABSTRACT

Tumor-initiating cells (TIC) perpetuate tumor growth, enable therapeutic resistance, and drive initiation of successive tumors. Virtually nothing is known about the role of mechanotransductive signaling in controlling TIC tumorigenesis, despite the recognized importance of altered mechanics in tissue dysplasia and the common observation that extracellular matrix (ECM) stiffness strongly regulates cell behavior. To address this open question, we cultured primary human glioblastoma (GBM) TICs on laminin-functionalized ECMs spanning a range of stiffnesses. Surprisingly, we found that these cells were largely insensitive to ECM stiffness cues, evading the inhibition of spreading, migration, and proliferation typically imposed by compliant ECMs. We hypothesized that this insensitivity may result from insufficient generation of myosin-dependent contractile force. Indeed, we found that both pharmacologic and genetic activation of cell contractility through RhoA GTPase, Rho-associated kinase, or myosin light chain kinase restored stiffness-dependent spreading and motility, with TICs adopting the expected rounded and nonmotile phenotype on soft ECMs. Moreover, constitutive activation of RhoA restricted three-dimensional invasion in both spheroid implantation and Transwell paradigms. Orthotopic xenotransplantation studies revealed that control TICs formed tumors with classical GBM histopathology including diffuse infiltration and secondary foci, whereas TICs expressing a constitutively active mutant of RhoA produced circumscribed masses and yielded a 30% enhancement in mean survival time. This is the first direct evidence that manipulation of mechanotransductive signaling can alter the tumor-initiating capacity of GBM TICs, supporting further exploration of these signals as potential therapeutic targets and predictors of tumor-initiating capacity within heterogeneous tumor cell populations.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Myosins/physiology , Neoplastic Stem Cells/physiology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Extracellular Matrix/metabolism , Female , Humans , Mice , Neoplasm Invasiveness , rhoA GTP-Binding Protein/physiology
2.
PLoS One ; 9(7): e101771, 2014.
Article in English | MEDLINE | ID: mdl-25000176

ABSTRACT

The aggressive and rapidly lethal brain tumor glioblastoma (GBM) is associated with profound tissue stiffening and genomic lesions in key members of the epidermal growth factor receptor (EGFR) pathway. Previous studies from our laboratory have shown that increasing microenvironmental stiffness in culture can strongly enhance glioma cell behaviors relevant to tumor progression, including proliferation, yet it has remained unclear whether stiffness and EGFR regulate proliferation through common or independent signaling mechanisms. Here we test the hypothesis that microenvironmental stiffness regulates cell cycle progression and proliferation in GBM tumor cells by altering EGFR-dependent signaling. We began by performing an unbiased reverse phase protein array screen, which revealed that stiffness modulates expression and phosphorylation of a broad range of signals relevant to proliferation, including members of the EGFR pathway. We subsequently found that culturing human GBM tumor cells on progressively stiffer culture substrates both dramatically increases proliferation and facilitates passage through the G1/S checkpoint of the cell cycle, consistent with an EGFR-dependent process. Western Blots showed that increasing microenvironmental stiffness enhances the expression and phosphorylation of EGFR and its downstream effector Akt. Pharmacological loss-of-function studies revealed that the stiffness-sensitivity of proliferation is strongly blunted by inhibition of EGFR, Akt, or PI3 kinase. Finally, we observed that stiffness strongly regulates EGFR clustering, with phosphorylated EGFR condensing into vinculin-positive focal adhesions on stiff substrates and dispersing as microenvironmental stiffness falls to physiological levels. Our findings collectively support a model in which tissue stiffening promotes GBM proliferation by spatially and biochemically amplifying EGFR signaling.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/pathology , Mechanical Phenomena , Signal Transduction , Tumor Microenvironment , Biomechanical Phenomena , Cell Proliferation , Focal Adhesions/metabolism , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Mechanotransduction, Cellular , Nitrogen/metabolism , Phosphorylation , Protein Transport , S Phase Cell Cycle Checkpoints
3.
Stem Cell Res Ther ; 4 Suppl 1: S11, 2013.
Article in English | MEDLINE | ID: mdl-24565274

ABSTRACT

The vast majority of cancer mortalities result from distant metastases. The metastatic microenvironment provides unique protection to ectopic tumors as the primary tumors often respond to specific agents. Although significant interventional progress has been made on primary tumors, the lack of relevant accessible model in vitro systems in which to study metastases has plagued metastatic therapeutic development--particularly among micrometastases. A real-time, all-human model of metastatic seeding and cancer cells that recapitulate metastatic growth and can be probed in real time by a variety of measures and challenges would provide a critical window into the pathophysiology of metastasis and pharmacology of metastatic tumor resistance. To achieve this we are advancing our microscale bioreactor that incorporates human hepatocytes, human nonparenchymal liver cells, and human breast cancer cells to mimic the hepatic niche in three dimensions with functional tissue. This bioreactor is instrumented with oxygen sensors, micropumps capable of generating diurnally varying profiles of nutrients and hormones, while enabling real-time sampling. Since the liver is a major metastatic site for a wide variety of carcinomas and other tumors, this bioreactor uniquely allows us to more accurately recreate the human metastatic microenvironment and probe the paracrine effects between the liver parenchyma and metastatic cells. Further, as the liver is the principal site of xenobiotic metabolism, this reactor will help us investigate the chemotherapeutic response within a metabolically challenged liver microenvironment. This model is anticipated to yield markers of metastatic behavior and pharmacologic metabolism that will enable better clinical monitoring, and will guide the design of clinical studies to understand drug efficacy and safety in cancer therapeutics. This highly instrumented bioreactor format, hosting a growing tumor within a microenvironment and monitoring its responses, is readily transferable to other organs, giving this work impact beyond the liver.


Subject(s)
Breast Neoplasms/pathology , Hepatocytes/cytology , Liver Neoplasms/secondary , Neoplastic Stem Cells/cytology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Bioreactors , Breast Neoplasms/drug therapy , Cell Survival/drug effects , Cytokines/metabolism , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/drug effects , Liver/metabolism , Liver Neoplasms/drug therapy , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism
4.
Biomaterials ; 32(24): 5633-42, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21575987

ABSTRACT

Cells are strongly influenced by the local structure and mechanics of the extracellular matrix (ECM). We recently showed that adding agarose to soft collagen ECMs can mechanically stiffen these hydrogels by two orders of magnitude while limiting 3D cell motility, which we speculated might derive from agarose-mediated inhibition of collagen fiber deformation and remodeling. Here, we directly address this hypothesis by investigating the effects of agarose on cell-collagen interactions at the microscale. Addition of agarose progressively restricts cell spreading, reduces stress fiber and focal adhesion assembly, and inhibits macroscopic gel compaction. While time-of-flight secondary ion mass spectrometry and scanning electron microscopy fail to reveal agarose-induced alterations in collagen ligand presentation, the latter modality shows that agarose strongly impairs cell-directed assembly of large collagen bundles. Agarose-mediated inhibition of cell spreading and cytoarchitecture can be rescued by ß-agarase digestion or by covalently crosslinking the matrix with glutaraldehyde. Based on these results, we argue that cell spreading and motility on collagen requires local matrix stiffening, which can be achieved via cell-mediated fiber remodeling or by chemically crosslinking the fibers. These findings provide new mechanistic insights into the regulatory function of agarose and bear general implications for cell adhesion and motility in fibrous ECMs.


Subject(s)
Collagen/metabolism , Sepharose/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Mass Spectrometry , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Principal Component Analysis
5.
J Clin Invest ; 120(4): 1084-96, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20200449

ABSTRACT

Injury and loss of podocytes are leading factors of glomerular disease and renal failure. The postmitotic podocyte is the primary glomerular target for toxic, immune, metabolic, and oxidant stress, but little is known about how this cell type copes with stress. Recently, autophagy has been identified as a major pathway that delivers damaged proteins and organelles to lysosomes in order to maintain cellular homeostasis. Here we report that podocytes exhibit an unusually high level of constitutive autophagy. Podocyte-specific deletion of autophagy-related 5 (Atg5) led to a glomerulopathy in aging mice that was accompanied by an accumulation of oxidized and ubiquitinated proteins, ER stress, and proteinuria. These changes resulted ultimately in podocyte loss and late-onset glomerulosclerosis. Analysis of pathophysiological conditions indicated that autophagy was substantially increased in glomeruli from mice with induced proteinuria and in glomeruli from patients with acquired proteinuric diseases. Further, mice lacking Atg5 in podocytes exhibited strongly increased susceptibility to models of glomerular disease. These findings highlight the importance of induced autophagy as a key homeostatic mechanism to maintain podocyte integrity. We postulate that constitutive and induced autophagy is a major protective mechanism against podocyte aging and glomerular injury, representing a putative target to ameliorate human glomerular disease and aging-related loss of renal function.


Subject(s)
Aging/physiology , Autophagy , Kidney Diseases/etiology , Kidney Glomerulus/pathology , Podocytes/physiology , Animals , Autophagy-Related Protein 5 , Cells, Cultured , Disease Susceptibility , Homeostasis , Humans , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/physiology , Proteasome Endopeptidase Complex/physiology , Proteinuria/etiology , Stress, Physiological , Ubiquitination
6.
Biomaterials ; 31(7): 1875-84, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19926126

ABSTRACT

The study of how cell behavior is controlled by the biophysical properties of the extracellular matrix (ECM) is limited in part by the lack of three-dimensional (3D) scaffolds that combine the biofunctionality of native ECM proteins with the tunability of synthetic materials. Here, we introduce a biomaterial platform in which the biophysical properties of collagen I are progressively altered by adding agarose. We find that agarose increases the elasticity of 3D collagen ECMs over two orders of magnitude with modest effect on collagen fiber organization. Surprisingly, increasing the agarose content slows and eventually stops invasion of glioma cells in a 3D spheroid model. Electron microscopy reveals that agarose forms a dense meshwork between the collagen fibers, which we postulate slows invasion by structurally coupling and reinforcing the collagen fibers and introducing steric barriers to motility. This is supported by time lapse imaging of individual glioma cells and multicellular spheroids, which shows that addition of agarose promotes amoeboid motility and restricts cell-mediated remodeling of individual collagen fibers. Our results are consistent with a model in which agarose shifts ECM dissipation of cell-induced stresses from non-affine deformation of individual collagen fibers to bulk-affine deformation of a continuum network.


Subject(s)
Cell Culture Techniques/methods , Extracellular Matrix/metabolism , Fibrillar Collagens/pharmacology , Sepharose/pharmacology , Biomechanical Phenomena/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Elasticity/drug effects , Extracellular Matrix/drug effects , Gels , Glioma/pathology , Humans , Mesoderm/drug effects , Mesoderm/pathology , Microscopy, Electron, Scanning , Neoplasm Invasiveness , Porosity/drug effects , Stress, Mechanical , Time Factors
7.
Cancer Res ; 69(10): 4167-74, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19435897

ABSTRACT

Glioblastoma multiforme (GBM) is a malignant astrocytoma of the central nervous system associated with a median survival time of 15 months, even with aggressive therapy. This rapid progression is due in part to diffuse infiltration of single tumor cells into the brain parenchyma, which is thought to involve aberrant interactions between tumor cells and the extracellular matrix (ECM). Here, we test the hypothesis that mechanical cues from the ECM contribute to key tumor cell properties relevant to invasion. We cultured a series of glioma cell lines (U373-MG, U87-MG, U251-MG, SNB19, C6) on fibronectin-coated polymeric ECM substrates of defined mechanical rigidity and investigated the role of ECM rigidity in regulating tumor cell structure, migration, and proliferation. On highly rigid ECMs, tumor cells spread extensively, form prominent stress fibers and mature focal adhesions, and migrate rapidly. As ECM rigidity is lowered to values comparable with normal brain tissue, tumor cells appear rounded and fail to productively migrate. Remarkably, cell proliferation is also strongly regulated by ECM rigidity, with cells dividing much more rapidly on rigid than on compliant ECMs. Pharmacologic inhibition of nonmuscle myosin II-based contractility blunts this rigidity-sensitivity and rescues cell motility on highly compliant substrates. Collectively, our results provide support for a novel model in which ECM rigidity provides a transformative, microenvironmental cue that acts through actomyosin contractility to regulate the invasive properties of GBM tumor cells.


Subject(s)
Actomyosin/physiology , Extracellular Matrix/pathology , Extracellular Matrix/ultrastructure , Glioma/pathology , Glioma/physiopathology , Animals , Cell Adhesion/physiology , Cell Division/physiology , Cell Line, Tumor , Cell Movement/physiology , Cues , Fibronectins/physiology , Glioblastoma/pathology , Glioblastoma/physiopathology , Humans , Muscle Contraction , Neoplasm Invasiveness , Stress, Mechanical
8.
Appl Opt ; 46(3): 421-7, 2007 Jan 20.
Article in English | MEDLINE | ID: mdl-17228390

ABSTRACT

Mechanical drift is a long-standing problem in optical microscopy that occurs in all three dimensions. This drift increasingly limits the resolution of advanced surface-coupled, single-molecule experiments. We overcame this drift and achieved atomic-scale stabilization (0.1 nm) of an optical microscope in 3D. This was accomplished by measuring the position of a fiducial mark coupled to the microscope cover slip using back-focal-plane (BFP) detection and correcting for the drift using a piezoelectric stage. Several significant factors contributed to this experimental realization, including (i) dramatically reducing the low frequency noise in BFP detection, (ii) increasing the sensitivity of BFP detection to vertical motion, and (iii) fabricating a regular array of nanometer-sized fiducial marks that were firmly coupled to the cover slip. With these improvements, we achieved short-term (1 s) stabilities of 0.11, 0.10, and 0.09 nm (rms) and long-term (100 s) stabilities of 0.17, 0.12, and 0.35 nm (rms) in x, y, and z, respectively, as measured by an independent detection laser.


Subject(s)
Micromanipulation , Microscopy/instrumentation , Algorithms , Equipment Design , Image Interpretation, Computer-Assisted , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Lasers , Microscopy/methods , Sensitivity and Specificity , Software , Subtraction Technique , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...