Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
2.
J Vis Exp ; (190)2022 12 23.
Article in English | MEDLINE | ID: mdl-36622034

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is prevalent worldwide, accounting for 90% of all esophageal cancer cases each year, and is the deadliest of all human squamous cell carcinomas. Despite recent progress in defining the molecular changes accompanying ESCC initiation and development, patient prognosis remains poor. The functional annotation of these molecular changes is the necessary next step and requires models that both capture the molecular features of ESCC and can be readily and inexpensively manipulated for functional annotation. Mice treated with the tobacco smoke mimetic 4-nitroquinoline 1-oxide (4NQO) predictably form ESCC and esophageal preneoplasia. Of note, 4NQO lesions also arise in the oral cavity, most commonly in the tongue, as well as the forestomach, which all share the stratified squamous epithelium. However, these mice cannot be simply manipulated for functional hypothesis testing, as generating isogenic mouse models is time- and resource-intensive. Herein, we overcome this limitation by generating single cell-derived three-dimensional (3D) organoids from mice treated with 4NQO to characterize murine ESCC or preneoplastic cells ex vivo. These organoids capture the salient features of ESCC and esophageal preneoplasia, can be cheaply and quickly leveraged to form isogenic models, and can be utilized for syngeneic transplantation experiments. We demonstrate how to generate 3D organoids from normal, preneoplastic, and SCC murine esophageal tissue and maintain and cryopreserve these organoids. The applications of these versatile organoids are broad and include the utilization of genetically engineered mice and further characterization by flow cytometry or immunohistochemistry, the generation of isogeneic organoid lines using CRISPR technologies, and drug screening or syngeneic transplantation. We believe that the widespread adoption of the techniques demonstrated in this protocol will accelerate progress in this field to combat the severe burden of ESCC.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Mice , Animals , Esophageal Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , Squamous Cell Carcinoma of Head and Neck , Organoids/metabolism , Cell Line, Tumor , Cell Proliferation
3.
Sci Rep ; 11(1): 13429, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34183700

ABSTRACT

Here it is investigated the effect of the antiferromagnet Cr2O3 on the magnetic properties of ferromagnetic Fe72Ga28 thin films. Sputtered Fe72Ga28 layers have their magnetization in the sample plane with a magnetic fluctuation that gives rise to magnetic ripple. In order to turn its magnetization into the out of plane (OOP) direction, it has been magnetically coupled with Cr2O3 that has magnetic moments along the c-axis, that is the perpendicular direction when properly aligned. Cr2O3 has been obtained from Cr oxidation, whereas Fe72Ga28 has been deposited on top of it by sputtering in the ballistic regime. Although a uniaxial in-plane magnetic anisotropy is expected for Fe72Ga28 thickness above 100 nm, the interfacial coupling with Cr2O3 prevents this anisotropy. The formation of stripe domains in Fe72Ga28 above a critical thickness reveals the enhancement of the out of plane component of the Fe72Ga28 magnetization with respect to uncoupled layers. Due to the interface coupling, the Fe72Ga28 magnetization turns into the out-of-plane direction as its thickness is gradually reduced, and a perpendicular magnetic anisotropy of 3·106 erg·cm-3 is inferred from experimental results. Eventually, the coupling between Cr2O3 and Fe72Ga28 promotes an exchange-bias effect that has been well fitted by means of the random field model.

4.
Acta Derm Venereol ; 100(10): adv00157, 2020 May 28.
Article in English | MEDLINE | ID: mdl-32449780

ABSTRACT

Although recent therapeutic developments raise hope, melanoma remains a devastating disease with a need for new treatment targets. In other tumours prohormone convertases have been shown to be pro-tumourigenic as they are involved in processing preforms of matrix-metalloproteinases, growth factors and adhesion molecules. The aim of this study was to look for new treatment options for melanoma, by investigating the role of the prohormone convertase Paired basic Amino acid-Cleaving Enzyme 4 (PACE4/PCSK6) in melanoma cell lines and human melanoma tissue. PACE4-transfected A375 melanoma cells displayed significantly increased proliferation, MMP-2 production, gelatinase activity and migratory capacity in vitro compared with sham-transfected cells. In vivo, elevated PACE4 expression resulted in significantly increased tumour growth on immunodeficient mice. In the majority of 45 human primary melanomas and melanoma metastases ex vivo PACE4 immunoreactivity was detectable, while it was absent in in situ melanomas. These results indicate PACE4 as a regulator of melanoma cell aggressiveness.


Subject(s)
Melanoma/enzymology , Proprotein Convertases/metabolism , Serine Endopeptidases/metabolism , Skin Neoplasms/enzymology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Mice, Hairless , Mice, SCID , Molecular Targeted Therapy , Neoplasm Invasiveness , Proprotein Convertases/antagonists & inhibitors , Proprotein Convertases/genetics , Serine Endopeptidases/genetics , Serine Proteinase Inhibitors/therapeutic use , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Burden
6.
Biochem Pharmacol ; 164: 336-341, 2019 06.
Article in English | MEDLINE | ID: mdl-30953636

ABSTRACT

Hypercholesterolemia represents a leading cause in the development of atherosclerotic plaques, increasing the risk for ACVS. It actually counts as a major cause of cardiovascular disease etiopathogenesis. The causes of hypercholesterolemia are multifactorial, spanning from genetic constitution, age, sex, to sedentary lifestyle and diets rich in sugars and lipids. Although dietary restriction in saturated fats, increased exercise, and other modification in lifestyle represent a first-line approach to treat very initial stages in hypercholesterolemia, most patients will require the addition of pharmacological agents. Pharmacological approaches include inhibition of cholesterol synthesis, decreased fat absorption from the GI tract, and increased degradation of FA. These strategies present a series of side effects, low therapeutic efficiency in some patients, and reduced tolerability. One of the major goals in treatment for hypercholesterolemia is to decrease the levels of low density lipoproteins (LDL), while maintaining those of high density lipoproteins (HDL). LDL particles contain about 80% of lipids, most of it cholesterol and cholesteryl esters, and 20% of the ApoB-100 protein. LDL carries cholesterol to the tissues, to be incorporated to biological membranes, or to be transformed to steroids. Excess of LDL translates into increased levels of circulating cholesterol particles and accumulation in certain tissues, especially vascular tissue, initiating a fatty streak, which may evolve to an atheroma, causing a series of cardiovascular problems, including impaired circulation, high blood pressure, increased cardiac workload, and coronary artery disease. It is essential to prevent LDL accumulation into the bloodstream to avoid the formation of these fatty streaks and the initiation of a cascade that will lead to the development of atherosclerosis. In healthy individuals. Under physiological conditions, LDL is effectively removed from circulation through receptor-mediated endocytosis. LDL clearance involves binding to its receptor, LDLR, which enables the internalization of the LDL particle and drives its degradation in lysosomes. Once the LDL particle is degraded, the free receptor recycles to the plasma membrane, and captures new LDL particles. Adequate levels of LDLR are essential to remove the excess of cholesterol-laden LDL. Proprotein convertase, subtilysin kexin type 9 (PCSK-9), expressed in liver and intestine, binds to LDLR, and internalized. Once inside the cell, PCSK-9 catalyzes the proteolysis of LDLR, preventing its recycling to the cell surface, and effectively decreasing the number of LDLR, notoriously decreasing the ability to clear LDL from circulation. Levels of PCSK-9 varies with age, gender, and levels of insulin, glucose, and triglycerides. Loss-of-function mutations in PCSK-9 gene invariably translates into lower levels of LDL, and decreased risk of developing coronary artery disease. Conversely, increased activity or expression of this enzyme leads to hypercholesterolemia. Inhibition of PCSK9 has proven to be successful in decreasing LDL levels and risk of the development of hypercholesterolemia with its associated higher risk for ASCVD. Patient with gain-of-function mutations in the PCSK9 undoubtedly benefit from therapies based on PCSK-9 inhibitors. However, millions of patients show statin intolerance, or cannot be efficiently controlled by statins alone- the most prevalent therapy for hypeprcholesterolemia. This commentary will evaluate the possibilities, caveats and future directions in the treatment of hypercholesterolemia, and therapies with combination of drugs.


Subject(s)
Anticholesteremic Agents/therapeutic use , Cholesterol, LDL/antagonists & inhibitors , Cholesterol, LDL/blood , Hypercholesterolemia/blood , Hypercholesterolemia/drug therapy , Animals , Anticholesteremic Agents/pharmacology , Atherosclerosis/blood , Atherosclerosis/drug therapy , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipid Metabolism/drug effects , Lipid Metabolism/physiology
8.
Oncotarget ; 8(9): 14941-14956, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28122332

ABSTRACT

Homeobox genes play a critical role in embryonic development, but they have also been implicated in cancer through mechanisms that are largely unknown. While not expressed during normal T-cell development, homeobox transcription factor genes can be reactivated via recurrent chromosomal rearrangements in human T-cell acute leukemia/lymphoma (T-ALL), a malignancy often associated with activated Notch and Akt signaling. To address how epigenetic reprogramming via an activated homeobox gene might contribute to T-lymphomagenesis, we investigated a transgenic mouse model with thymocyte-specific overexpression of the Dlx5 homeobox gene. We demonstrate for the first time that Dlx5 induces T-cell lymphomas with high penetrance. Integrated ChIP-seq and mRNA microarray analyses identified Notch1/3 and Irs2 as direct transcriptional targets of Dlx5, a gene signature unique to lymphomas from Lck-Dlx5 mice as compared to T-cell lymphomas from Lck-MyrAkt2 mice, which were previously reported by our group. Moreover, promoter/enhancer studies confirmed that Dlx5 directly transactivates Notch expression. Notch1/3 expression and Irs2-induced Akt signaling were upregulated throughout early stages of T-cell development, which promoted cell survival during ß-selection of T lymphocytes. Dlx5 was required for tumor maintenance via its activation of Notch and Akt, as tumor cells were highly sensitive to Notch and Akt inhibitors. Together, these findings provide unbiased genetic and mechanistic evidence that Dlx5 acts as an oncogene when aberrantly expressed in T cells, and that it is a novel discovery that Notch is a direct target of Dlx5. These experimental findings provide mechanistic insights about how reactivation of the Dlx5 gene can drive T-ALL by aberrant epigenetic reprogramming of the T-cell genome.


Subject(s)
Gene Expression Regulation, Neoplastic , Homeodomain Proteins/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Lymphoma, T-Cell/pathology , Proto-Oncogene Proteins c-akt/physiology , Receptor, Notch1/genetics , Animals , Apoptosis , Cell Proliferation , Humans , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic , Signal Transduction , Transcriptional Activation , Tumor Cells, Cultured
9.
Oncotarget ; 6(40): 42892-904, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26503472

ABSTRACT

The DNA glycosylase gene MBD4 safeguards genomic stability at CpG sites and is frequently mutated at coding poly-A tracks in mismatch repair (MMR)-defective colorectal tumors (CRC). Mbd4 biallelic inactivation in mice provided conflicting results as to its role in tumorigenesis. Thus, it is unclear whether MBD4 alterations are only secondary to MMR defects without functional consequences or can contribute to the mutator phenotype. We investigated MBD4 variants in a large series of hereditary/familial and sporadic CRC cases. Whereas MBD4 frameshifts were only detected in tumors, missense variants were found in both normal and tumor DNA. In CRC with double-MBD4/MMR and single-MBD4 variants, transition mutation frequency was increased, indicating that MBD4 defects may affect the mutational landscape independently of MMR defect. Mbd4-deficient mice showed reduced survival when combined with Mlh1-/- genotype. Taken together, these data suggest that MBD4 inactivation may contribute to tumorigenesis, acting as a modifier of MMR-deficient cancer phenotype.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , DNA Mismatch Repair/genetics , Endodeoxyribonucleases/genetics , Animals , DNA Mutational Analysis , Female , Humans , Male , Mice , Mice, Knockout , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , Polymerase Chain Reaction
10.
Cancer Discov ; 5(3): 288-303, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25582697

ABSTRACT

UNLABELLED: Protein arginine methyltransferase 5 (PRMT5) has been implicated as a key modulator of lymphomagenesis. Whether PRMT5 has overt oncogenic function in the context of leukemia/lymphoma and whether it represents a therapeutic target remains to be established. We demonstrate that inactivation of PRMT5 inhibits colony-forming activity by multiple oncogenic drivers, including cyclin D1, c-MYC, NOTCH1, and MLL-AF9. Furthermore, we demonstrate that PRMT5 overexpression specifically cooperates with cyclin D1 to drive lymphomagenesis in a mouse model, revealing inherent neoplastic activity. Molecular analysis of lymphomas revealed that arginine methylation of p53 selectively suppresses expression of crucial proapoptotic and antiproliferative target genes, thereby sustaining tumor cell self-renewal and proliferation and bypassing the need for the acquisition of inactivating p53 mutations. Critically, analysis of human tumor specimens reveals a strong correlation between cyclin D1 overexpression and p53 methylation, supporting the biomedical relevance of this pathway. SIGNIFICANCE: We have identified and functionally validated a crucial role for PRMT5 for the inhibition of p53-dependent tumor suppression in response to oncogenic insults. The requisite role for PRMT5 in the context of multiple lymphoma/leukemia oncogenic drivers suggests a molecular rationale for therapeutic development.


Subject(s)
Cell Transformation, Neoplastic/genetics , Lymphoma/genetics , Protein-Arginine N-Methyltransferases/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Animals , Apoptosis/genetics , Arginine/metabolism , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Enzyme Activation , Gene Expression Profiling , Humans , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Lymphoma/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Methylation , Mice , Mutation , Oncogenes , Phosphorylation , Protein-Arginine N-Methyltransferases/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Nanotechnology ; 25(10): 105702, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24532090

ABSTRACT

Porous films of Co/CoO magnetic nanoparticles have been obtained by inert gas condensation and partially oxidized in situ in the deposition chamber. These nanoparticle films were subjected to thermal treatments in high vacuum and the chemical and structural changes monitored by x-ray diffraction, transmission electron microscopy, transport and magnetic measurements (with a focus on the exchange-bias phenomenon), which evidence that for vacuum annealing temperatures above 360 °C, most of the CoO phase is reduced to metallic Co without requiring the presence of an external reducing agent (e.g., H2) or a plasma. Additionally, there is a certain degree of particle coalescence resulting in the formation of greater nanoparticles as the annealing temperature increases. This yields a smaller proportion of CoO compared to metallic Co and a reduction of the Co/CoO interface density, pinpointed by a drastic decrease of the exchange-bias field. The crucial roles of the vacuum level and the surface-to-volume ratio are evidenced by magnetic measurements, highlighting the potential of magnetometry as a probe for the reduction/oxidation of composite nanostructures.

12.
Head Neck ; 36(8): 1113-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23804308

ABSTRACT

BACKGROUND: Psychosocial functioning is associated with vascular endothelial growth factor (VEGF) in various patient populations. This study examined whether psychosocial functioning in patients with head and neck squamous cell carcinoma (HNSCC) is associated with tumor VEGF expression, a protein that stimulates angiogenesis and is associated with poor prognosis. METHODS: Forty-two newly diagnosed patients completed assessments of psychosocial functioning (ie, depressive symptoms, perceived stress, anxiety, social support) before surgery. Tumor samples were obtained for VEGF analysis and human papillomavirus (HPV)-typing. RESULTS: Poorer psychosocial functioning was associated with greater VEGF expression controlling for disease stage (odds ratio [OR], 4.55; 95% confidence interval [CI], 1.72-12.0; p < .01). When examined by HPV status, the association between psychosocial functioning and VEGF remained significant among patients who were HPV negative (OR, 5.50; 95% CI, 1.68-17.3; p < .01), but not among patients who were HPV positive. CONCLUSION: These findings inform our understanding of the biobehavioral pathways that may contribute to poor outcomes in non-HPV-associated HNSCCs.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Stress, Psychological/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Aged , Anxiety Disorders/metabolism , Depressive Disorder/metabolism , Female , Humans , Male , Middle Aged , Papillomaviridae/isolation & purification , Papillomavirus Infections/diagnosis , Prognosis , Squamous Cell Carcinoma of Head and Neck
13.
Cancer Res ; 72(22): 5966-75, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22983922

ABSTRACT

The RAS genes are the most commonly mutated oncogenes in human cancer and present a particular therapeutic dilemma, as direct targeting of Ras proteins by small molecules has proved difficult. Signaling pathways downstream of Ras, in particular Raf/Mek/Erk and PI3K/Akt/mTOR, are dominated by lipid and protein kinases that provide attractive alternate targets in Ras-driven tumors. As p21-activated kinase 1 (Pak1) has been shown to regulate both these signaling pathways and is itself upregulated in many human cancers, we assessed the role of Pak1 in Ras-driven skin cancer. In human squamous cell carcinoma (SCC), we found a strong positive correlation between advanced stage and grade and PAK1 expression. Using a mouse model of Kras-driven SCC, we showed that deletion of the mouse Pak1 gene led to markedly decreased tumorigenesis and progression, accompanied by near total loss of Erk and Akt activity. Treatment of Kras(G12D) mice with either of two distinct small molecule Pak inhibitors (PF3758309 and FRAX597) caused tumor regression and loss of Erk and Akt activity. Tumor regression was also seen in mice treated with a specific Mek inhibitor, but not with an Akt inhibitor. These findings establish Pak1 as a new target in KRAS-driven tumors and suggest a mechanism of action through the Erk, but not the Akt, signaling pathway.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Proto-Oncogene Proteins/metabolism , Pyridones/pharmacology , Pyrimidines/pharmacology , Skin Neoplasms/enzymology , p21-Activated Kinases/biosynthesis , ras Proteins/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Deletion , Genes, ras , Humans , Immunohistochemistry , Mice , Mice, Knockout , Mice, Transgenic , Neoplasm Grading , Neoplasm Staging , Oncogene Protein v-akt/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/deficiency , p21-Activated Kinases/genetics , ras Proteins/genetics
14.
Neoplasia ; 14(4): 271-82, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22577343

ABSTRACT

Furin, one of the members of the family of proprotein convertases (PCs), ubiquitously expressed as a type I membrane-bound proteinase, activates several proteins that contribute to tumor progression. In vitro studies using cancer cell lines and clinical specimens demonstrated that furin processes important substrates such as insulin-like growth factor 1 receptor (IGF-1R) and transforming growth factor ß, leading to increased tumor growth and progression. Despite the numerous studies associating furin with tumor development, its effects in preclinical models has not been comprehensively studied. In this study, we sought to determine the protumorigenic role of furin in vivo after a two-stage chemical carcinogenesis protocol in transgenic mice in which furin expression was targeted to the epidermal basal layer. We found that processing of the PC substrate IGF-1R and the proliferation rate of mouse epidermis was enhanced in transgenic mice when compared with their WT counterparts. Histopathologic diagnoses of the tumors demonstrated that furin transgenic mice (line F47) developed twice as many squamous carcinomas as the control, WT mice (P < .002). Similarly, tumors cells from transgenic mice were able to process PC substrates more efficiently than tumor cells from WT mice. Furthermore, furin expression resulted in a higher SCC volume in transgenic mice as well as an increase in the percentage of high-grade SCC, including poorly differentiated and spindle cell carcinomas. In conclusion, expression of furin in the basal layer of the epidermis increased tumor development and enhanced tumor growth, supporting the consideration of furin as a potential target for cancer treatment.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Furin/metabolism , Skin Neoplasms/metabolism , Animals , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Female , Furin/genetics , Humans , Immunohistochemistry , Keratinocytes/metabolism , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/genetics , Skin Neoplasms/pathology
15.
Mol Cell Biol ; 31(22): 4513-23, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21911473

ABSTRACT

Skp1-Cul1-F-box (SCF) E3 ubiquitin ligase complexes modulate the accumulation of key cell cycle regulatory proteins. Following the G(1)/S transition, SCF(Fbx4) targets cyclin D1 for proteasomal degradation, a critical event necessary for DNA replication fidelity. Deregulated cyclin D1 drives tumorigenesis, and inactivating mutations in Fbx4 have been identified in human cancer, suggesting that Fbx4 may function as a tumor suppressor. Fbx4(+/-) and Fbx4(-/-) mice succumb to multiple tumor phenotypes, including lymphomas, histiocytic sarcomas and, less frequently, mammary and hepatocellular carcinomas. Tumors and premalignant tissue from Fbx4(+/-) and Fbx4(-/-) mice exhibit elevated cyclin D1, an observation consistent with cyclin D1 as a target of Fbx4. Molecular dissection of the Fbx4 regulatory network in murine embryonic fibroblasts (MEFs) revealed that loss of Fbx4 results in cyclin D1 stabilization and nuclear accumulation throughout cell division. Increased proliferation in early passage primary MEFs is antagonized by DNA damage checkpoint activation, consistent with nuclear cyclin D1-driven genomic instability. Furthermore, Fbx4(-/-) MEFs exhibited increased susceptibility to Ras-dependent transformation in vitro, analogous to tumorigenesis observed in mice. Collectively, these data reveal a requisite role for the SCF(Fbx4) E3 ubiquitin ligase in regulating cyclin D1 accumulation, consistent with tumor suppressive function in vivo.


Subject(s)
Cell Transformation, Neoplastic , Cyclin D1/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Cyclin D1/biosynthesis , Cyclin D1/genetics , DNA Damage , Fibroblasts/metabolism , Gene Knockout Techniques , Mice , Mice, Transgenic , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
16.
Cell ; 146(1): 148-63, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21729786

ABSTRACT

Mechanotransduction is a key determinant of tissue homeostasis and tumor progression. It is driven by intercellular adhesions, cell contractility, and forces generated within the microenvironment and is dependent on extracellular matrix composition, organization, and compliance. We show that caveolin-1 (Cav1) favors cell elongation in three-dimensional cultures and promotes Rho- and force-dependent contraction, matrix alignment, and microenvironment stiffening through regulation of p190RhoGAP. In turn, microenvironment remodeling by Cav1 fibroblasts forces cell elongation. Cav1-deficient mice have disorganized stromal tissue architecture. Stroma associated with human carcinomas and melanoma metastases is enriched in Cav1-expressing carcinoma-associated fibroblasts (CAFs). Cav1 expression in breast CAFs correlates with low survival, and Cav1 depletion in CAFs decreases CAF contractility. Consistently, fibroblast expression of Cav1, through p190RhoGAP regulation, favors directional migration and invasiveness of carcinoma cells in vitro. In vivo, stromal Cav1 remodels peri- and intratumoral microenvironments to facilitate tumor invasion, correlating with increased metastatic potency. Thus, Cav1 modulates tissue responses through force-dependent architectural regulation of the microenvironment.


Subject(s)
Caveolin 1/metabolism , Neoplasm Metastasis/pathology , Neoplasms/pathology , Animals , Cell Movement , Fibroblasts/pathology , Humans , Melanoma/pathology , Mice , Mice, Knockout
17.
Cancer Res ; 71(6): 2250-9, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21406401

ABSTRACT

Antibody drugs are widely used in cancer therapy, but conditions to maximize tumor penetration and efficacy have yet to be fully elucidated. In this study, we investigated the impact of antibody binding affinity on tumor targeting and penetration with affinity variants that recognize the same epitope. Specifically, we compared four derivatives of the C6.5 monoclonal antibody (mAb), which recognizes the same HER2 epitope (monovalent K(D) values ranging from 270 to 0.56 nmol/L). Moderate affinity was associated with the highest tumor accumulation at 24 and 120 hours after intravenous injection, whereas high affinity was found to produce the lowest tumor accumulation. Highest affinity mAbs were confined to the perivascular space of tumors with an average penetration of 20.4 ± 7.5 µm from tumor blood vessels. Conversely, lowest affinity mAbs exhibited a broader distribution pattern with an average penetration of 84.8 ± 12.8 µm. In vitro internalization assays revealed that antibody internalization and catabolism generally increased with affinity, plateauing once the rate of HER2 internalization exceeded the rate of antibody dissociation. Effects of internalization and catabolism on tumor targeting were further examined using antibodies of moderate (C6.5) or high-affinity (trastuzumab), labeled with residualizing ((111)In-labeled) or nonresidualizing ((125)I-labeled) radioisotopes. Significant amounts of antibody of both affinities were degraded by tumors in vivo. Furthermore, moderate- to high-affinity mAbs targeting the same HER2 epitope with monovalent affinity above 23 nmol/L had equal tumor accumulation of residualizing radiolabel over 120 hours. Results indicated equal tumor exposure, suggesting that mAb penetration and retention in tumors reflected affinity-based differences in tumor catabolism. Together, these results suggest that high-density, rapidly internalizing antigens subject high-affinity antibodies to greater internalization and degradation, thereby limiting their penetration of tumors. In contrast, lower-affinity antibodies penetrate tumors more effectively when rates of antibody-antigen dissociation are higher than those of antigen internalization. Together, our findings offer insights into how to optimize the ability of therapeutic antibodies to penetrate tumors.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Ovarian Neoplasms/immunology , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacokinetics , Antibody Affinity/immunology , Antigens, Neoplasm/metabolism , Binding, Competitive , Cell Line, Tumor , Endocytosis/immunology , Epitopes/immunology , Epitopes/metabolism , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunohistochemistry , Indium Radioisotopes , Iodine Radioisotopes , Mice , Mice, SCID , Mutation , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Time Factors , Tissue Distribution , Transplantation, Heterologous
18.
Cancer Prev Res (Phila) ; 4(1): 107-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21205741

ABSTRACT

Squamous cell carcinoma of the head and neck (HNSCC) is the sixth most common type of cancer in the United States. The goal of this study was to evaluate the contribution of estrogens to the development of HNSCCs. Various cell lines derived from early- and late-stage head and neck lesions were used to characterize the expression of estrogen synthesis and metabolism genes, including cytochrome P450 (CYP) 1B1, examine the effect of estrogen on gene expression, and evaluate the role of CYP1B1 and/or estrogen in cell motility, proliferation, and apoptosis. Estrogen metabolism genes (CYP1B1, CYP1A1, catechol-o-methyltransferase, UDP-glucuronosyltransferase 1A1, and glutathione-S-transferase P1) and estrogen receptor (ER) ß were expressed in cell lines derived from both premalignant (MSK-Leuk1) and malignant (HNSCC) lesions. Exposure to estrogen induced CYP1B1 2.3- to 3.6-fold relative to vehicle-treated controls (P = 0.0004) in MSK-Leuk1 cells but not in HNSCC cells. CYP1B1 knockdown by shRNA reduced the migration and proliferation of MSK-Leuk1 cells by 57% and 45%, respectively. Exposure of MSK-Leuk1 cells to estrogen inhibited apoptosis by 26%, whereas supplementation with the antiestrogen fulvestrant restored estrogen-dependent apoptosis. Representation of the estrogen pathway in human head and neck tissues from 128 patients was examined using tissue microarrays. The majority of the samples exhibited immunohistochemical staining for ERß (91.9%), CYP1B1 (99.4%), and 17ß-estradiol (88.4%). CYP1B1 and ERß were elevated in HNSCCs relative to normal epithelium (P = 0.024 and 0.008, respectively). These data provide novel insight into the mechanisms underlying head and neck carcinogenesis and facilitate the identification of new targets for chemopreventive intervention.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Estrogens/pharmacology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Apoptosis/drug effects , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Blotting, Western , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytochrome P-450 CYP1B1 , Estradiol/genetics , Estradiol/metabolism , Estrogen Receptor beta/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunoenzyme Techniques , Leukoplakia/drug therapy , Leukoplakia/metabolism , Leukoplakia/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Tumor Cells, Cultured
19.
Neoplasia ; 12(7): 516-26, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20651981

ABSTRACT

PACE4 is a proprotein convertase (PC) responsible for cleaving and activating proteins that contribute to enhance tumor progression. PACE4 overexpression significantly increased the susceptibility to carcinogenesis, leading to enhanced tumor cell proliferation and premature degradation of the basement membrane. In the present study, we sought to evaluate a novel approach to retard skin tumor progression based on the inhibition of PACE4. We used decanoyl-RVKR-chloromethylketone (CMK), a small-molecule PC inhibitor, for in vitro and in vivo experiments. We found that CMK-dependent blockage of PACE4 activity in skin squamous cell carcinoma cell lines resulted in impaired insulin-like growth factor 1 receptor maturation, diminished its intrinsic tyrosine kinase activity, and decreased tumor cell proliferation. Two-stage skin chemical carcinogenesis experiments, together with topical applications of CMK, demonstrated that this PC inhibitor markedly reduced tumor incidence, tumor multiplicity, and metastasis, pointing to a significant delay in tumor progression in wild-type and PACE4 transgenic mice. These results identify PACE4, together with other PCs, as suitable targets to slow down or block tumor progression, suggesting that PC inhibition is a potential approach for therapy for solid tumors.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Proprotein Convertases/antagonists & inhibitors , Skin Neoplasms/pathology , Skin/pathology , Animals , Antineoplastic Agents/pharmacology , Cell Line , Cell Transformation, Neoplastic/pathology , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Female , Humans , Mice , Mice, Transgenic , Neoplasm Metastasis , Proprotein Convertases/genetics , Skin/drug effects , Xenograft Model Antitumor Assays
20.
PLoS One ; 5(4): e10196, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20419170

ABSTRACT

VILIP-1, a member of the neuronal Ca(2+) sensor protein family, is able to act as a tumor suppressor in carcinoma cells by inhibiting cell proliferation and migration. In order to study the role of VILIP-1 in skin carcinogenesis we generated transgenic mice overexpressing VILIP-1 in epidermis under the control of the bovine keratin K5 promoter (K5-VILIP-1). We studied the susceptibility of FVB wild type and VILIP-1 transgenic mice to chemically mediated carcinogenesis. After 30 weeks of treatment with a two-stage carcinogenesis protocol, all animals showed numerous skin tumors. Nevertheless, K5-VILIP-1 mice showed decreased squamous cell carcinoma (SCC) multiplicity of approximately 49% (p<0.02) with respect to the corresponding SCC multiplicity observed in wild type (WT) mice. In addition, the relative percentage of low-grade cutaneous SCCs grade I (defined by the differentiation pattern according to the Broders grading scale) increased approximately 50% in the K5-VILIP1 mice when compared with SCCs in WT mice. Similar tendency was observed using a complete carcinogenesis protocol for skin carcinogenesis using benzo(a)pyrene (B(a)P). Further studies of tumors and primary epidermal keratinocyte cultures showed that matrix metalloproteinase 9 (MMP-9) levels and cell proliferation decreased in K5-VILIP-1 mice when compared with their wild counterparts. In addition tissue inhibitor of metalloproteinase 1 (TIMP-1) expression was higher in K5-VILIP-1 keratinocytes. These results show that VILIP-1 overexpression decreases the susceptibility to skin carcinogenesis in experimental mouse cancer models, thus supporting its role as a tumor suppressor gene.


Subject(s)
Keratinocytes/cytology , Neurocalcin/physiology , Skin Neoplasms/etiology , Animals , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Disease Susceptibility , Genes, Tumor Suppressor , Matrix Metalloproteinase 9 , Mice , Mice, Transgenic , Neurocalcin/genetics , Skin Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-1 , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...