Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Lung Cancer ; 111: 79-83, 2017 09.
Article in English | MEDLINE | ID: mdl-28838404

ABSTRACT

BACKGROUND: ALK and ROS1 gene rearrangements are distinct molecular subsets of non-small-cell lung cancer (NSCLC), and they are strong predictive biomarkers of response to ALK/ROS1 inhibitors, such as crizotinib. Thus, it is clinically important to develop an effective screening strategy to detect patients who will benefit from such treatment. In this study, we aimed to validate analytical performance of Vysis ALK/ROS1 Dual Break Apart Probe Kit (RUO) in NSCLC. METHODS: Study population composed of three patient cohorts with histologically confirmed lung adenocarcinoma (patients with ALK rearrangement, patients with ROS1 rearrangement and patients with wild-type ALK and ROS1). Specimens consisted of 12 ALK-positive, 8 ROS1-positive and 21 ALK/ROS1-wild type formalin-fixed paraffin-embedded samples obtained from surgical resection or excisional biopsy. ALK rearrangement was previously assessed by Vysis ALK Break Apart FISH Probe Kit (Abbott Molecular, Abbot Park, IL, USA) and ROS1 rearrangement was previously assessed by ZytoLight® SPEC ROS1 Break Apart Probe (ZytoVision, GmbH). All specimens were re-evaluated by Vysis ALK/ROS1 Dual Break Apart Probe Kit. FISH images were scanned on BioView AllegroPlus system and interpreted via BioView SoloWeb remotely. RESULTS: For a total of 41 patient samples, the concordance of the results by Vysis ALK/ROS1 Dual Break Apart Probe Kit was evaluated and compared to the known ALK and ROS1 rearrangement status of the specimen. Of the 12 ALK-positive cases, hybridization with Vysis ALK/ROS1 Dual Break Apart Probe Kit was successful in 10 cases (success rate 10/12, 83%) and of these 10 cases, all showed ALK rearrangement (100% concordance with the results of Vysis ALK Break Apart FISH Probe Kit). Two of the ALK+ cases were excluded due to weak ROS1 signals that could not be enumerated. Of the 8 ROS1-positive cases, 6 cases were successfully evaluated using Vysis ALK/ROS1 Dual Break Apart Probe Kit. The success rate was 75% (6/8), and of these 6 cases, all showed ROS1 rearrangement, giving a 100% concordance with ZytoLight® SPEC ROS1 Break Apart Probe. Two of the cases were excluded due to weak ROS1 gold signal or high background. In the cohort of 21 wild-type cases, the success rate using Vysis ALK/ROS1 Dual Break Apart FISH Probe Kit was 85% (18/21) and the concordance with ALK and ROS1 probe kit was 100% (18/18). CONCLUSION: Vysis ALK/ROS1 Dual Break Apart Probe Kit (RUO) can detect ALK and ROS1 rearrangement simultaneously in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Gene Rearrangement , In Situ Hybridization, Fluorescence , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adult , Aged , Anaplastic Lymphoma Kinase , Biomarkers, Tumor , DNA Probes , Female , Genotype , Humans , In Situ Hybridization, Fluorescence/methods , Male , Middle Aged , Mutation , Neoplasm Staging , Oncogene Proteins, Fusion/genetics , Reagent Kits, Diagnostic , Reproducibility of Results
2.
Arch Suicide Res ; 15(3): 185-94, 2011.
Article in English | MEDLINE | ID: mdl-21827309

ABSTRACT

Although there has been extensive research into the epidemiology and prevention of suicide, there continues to be a paucity of research on non-fatal suicides, in particular persons not treated in hospitals following a suicide attempt. In this study, we analyzed call data from the Illinois Poison Center from 2002 to 2007, which primarily comprises of non-fatal hospitalized and non-hospitalized attempts. We analyzed 43,057 calls by persons suspected of attempting suicide. The three most common groups of substances used were analgesics, antidepressants, and sedative/hypnotics/antipsychotics. The Poisson regression model showed significant declines in calls for suspected suicides during periods of holidays and vacations, and was more pronounced among youths. This study provides a current and detailed description of substances used primarily in non-fatal suicide attempts.


Subject(s)
Central Nervous System Agents/poisoning , Poison Control Centers/standards , Poisoning/epidemiology , Suicide, Attempted/statistics & numerical data , Analgesics/poisoning , Antidepressive Agents/poisoning , Antipsychotic Agents/poisoning , Female , Humans , Hypnotics and Sedatives/poisoning , Male , Poisson Distribution , Regression Analysis , United States/epidemiology
3.
J Biol Chem ; 286(40): 34777-87, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21840991

ABSTRACT

Interactions between polymorphonuclear neutrophils (PMNs) and tumor cells have been reported to facilitate the adhesion and subsequent extravasation of tumor cells through the endothelium under blood flow, both of which are mediated by binding ß(2)-integrin to intercellular adhesion molecule 1 (ICAM-1). Here the adhesions between human WM9 metastatic melanoma cells, PMNs, and human pulmonary microvascular endothelial cells (HPMECs) were quantified by a gas-driven micropipette aspiration technique (GDMAT). Our data indicated that the cellular binding affinity of PMN-WM9 pair was 3.9-fold higher than that of the PMN-HPMEC pair. However, the effective binding affinities per molecular pair were comparable between the two cell pairs no matter whether WM9 cells or HPMECs were quiescent or cytokine-activated, indicating that the stronger adhesion between PMN-WM9 pair is mainly attributed to the high expression of ICAM-1 on WM9 cells. These results proposed an alternative mechanism, where WM9 melanoma cells adhere first with PMNs near vessel-wall regions and then bind to endothelial cells via PMNs under blood flow. In contrast, the adhesions between human MDA-MB-231 metastatic breast carcinoma cells and PMNs showed a comparable cellular binding affinity to PMN-HPMEC pair because the ICAM-1 expressions on MDA-MB-231 cells and HPMECs are similar. Furthermore, differences were observed in the intrinsic forward and reverse rates of the ß(2)-integrin-ICAM-1 bond between PMN-TC and PMN-EC pairs. This GDMAT assay enables us to quantify the binding kinetics of cell adhesion molecules physiologically expressed on nucleated cells. The findings also further the understanding of leukocyte-facilitated tumor cell adhesion from the viewpoint of molecular binding kinetics.


Subject(s)
CD18 Antigens/metabolism , Endothelial Cells/cytology , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/metabolism , Neoplasms/pathology , Animals , Cell Adhesion , Cell Line, Tumor , Cell Nucleus/metabolism , Gases , Humans , Kinetics , Mice , Neoplasm Metastasis , Protein Binding
4.
J Med Entomol ; 47(5): 907-12, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20939389

ABSTRACT

Although there is some detailed research on anaphylactic reactions to Hymenoptera venom, there continues to be little epidemiological data about the distribution, trend, and factors associated with the occurrence of Hymenoptera envenomations in humans. We describe characteristics of persons suffering Hymenoptera stings from bees, wasps, and hornets as reported to the Illinois Poison Center, and assess seasonal, climatologic, and time trends of calls for envenomations between 2002 and 2007. Mean daily temperature and mean daily atmospheric pressure were positively associated with envenomations, whereas wind speed was negatively associated with envenomations. We also observed a significant increase in calls for envenomations on summer holidays (P < 0.001). In addition, our findings showed that the number of calls for envenomations declined by nearly half after 2005 (P < 0.001) compared with previous years. Our findings indicate that the decline in bees, wasps, and hornets may be widespread, affecting both wild and commercial populations, and that the decline appears to have been rapid and sustained in recent years. Poison center data are a valuable resource for the surveillance of poisoning in humans, but our findings show that the data can be used to monitor changes in nonhuman species.


Subject(s)
Hymenoptera/physiology , Insect Bites and Stings/epidemiology , Adolescent , Adult , Aged , Animals , Child , Child, Preschool , Female , Humans , Illinois/epidemiology , Infant , Male , Middle Aged , Poison Control Centers , Time Factors , Young Adult
5.
Cancer Res ; 70(14): 6071-82, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20610626

ABSTRACT

It is unknown why only a minority of circulating tumor cells trapped in lung capillaries form metastases and involvement of immune cells remains uncertain. A novel model has been developed in this study showing that neutrophils regulate lung metastasis development through physical interaction and anchoring of circulating tumor cells to endothelium. Human melanoma cells were i.v. injected into nude mice leading to the entrapment of many cancer cells; however, 24 hours later, very few remained in the lungs. In contrast, injection of human neutrophils an hour after tumor cell injection increased cancer cell retention by approximately 3-fold. Entrapped melanoma cells produced and secreted high levels of a cytokine called interleukin-8 (IL-8), attracting neutrophils and increasing tethering beta(2) integrin expression by 75% to 100%. Intercellular adhesion molecule-1 on melanoma cells and beta(2) integrin on neutrophils interacted, promoting anchoring to vascular endothelium. Decreasing IL-8 secretion from melanoma cells lowered extracellular levels by 20% to 50%, decreased beta(2) integrin on neutrophils by approximately 50%, and reduced neutrophil-mediated extravasation by 25% to 60%, resulting in approximately 50% fewer melanoma cells being tethered to endothelium and retained in lungs. Thus, transendothelial migration and lung metastasis development decreased by approximately 50%, showing that targeting IL-8 in melanoma cells has the potential to decrease metastasis development by disrupting interaction with neutrophils.


Subject(s)
Lung Neoplasms/secondary , Melanoma/secondary , Neoplastic Cells, Circulating/pathology , Neutrophils/pathology , Skin Neoplasms/pathology , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Coculture Techniques , Female , Humans , Interleukin-8/biosynthesis , Interleukin-8/genetics , Interleukin-8/metabolism , Lung Neoplasms/blood , Lung Neoplasms/immunology , Melanoma/blood , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Nude , Neoplastic Cells, Circulating/immunology , Neutrophils/immunology , RNA, Small Interfering/genetics , Skin Neoplasms/blood , Skin Neoplasms/immunology
6.
Mol Cell Biomech ; 7(2): 77-91, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20379392

ABSTRACT

To complete the metastatic journey, cancer cells have to disseminate through the circulation and extravasate to distal organs. However, the extravasation process, by which tumor cells leave a blood vessel and invade the surrounding tissue from the microcirculation, remains poorly understood at the molecular level. In this study, tumor cell adhesion to the endothelium (EC) and subsequent extravasation were investigated under various flow conditions. Results have shown polymorphonuclear neutrophils (PMNs) facilitate melanoma cell adhesion to the EC and subsequent extravasation by a shear-rate dependent mechanism. Melanoma cell-PMN interactions are mediated by the binding between intercellular adhesion molecule-1 (ICAM-1) on melanoma cells and beta2 integrins on PMNs. In addition, the fluid convection affects the extent of activation of beta2 integrins on PMNs by endogenously secreted interleukin 8 (IL-8) within the tumor microenvironment. Results also indicate that shear rate affects the binding kinetics between PMNs and melanoma cells, which may contribute to the shear-rate dependence of melanoma extravasation in a shear flow when mediated by PMNs.


Subject(s)
Interleukin-8/physiology , Leukocytes/physiology , Neoplasm Metastasis/physiopathology , Animals , Biomechanical Phenomena , CD18 Antigens/physiology , Cell Adhesion/physiology , Cell Line, Tumor , Coculture Techniques , Endothelial Cells/pathology , Endothelial Cells/physiology , Humans , L Cells , Leukocyte Rolling/physiology , Leukocytes/pathology , Lymphocyte Function-Associated Antigen-1/physiology , Macrophage-1 Antigen/physiology , Melanoma/pathology , Melanoma/physiopathology , Melanoma/secondary , Mice , Neoplasm Metastasis/pathology , Neutrophils/pathology , Neutrophils/physiology , Rheology , Signal Transduction/physiology
7.
Cell Mol Bioeng ; 3(1): 3-19, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20428326

ABSTRACT

Our research focused on the polymorphonuclear neutrophils (PMNs) tethering to the vascular endothelial cells (EC) and the subsequent melanoma cell emboli formation in a shear flow, an important process of tumor cell extravasation from the circulation during metastasis. We applied population balance model based on Smoluchowski coagulation equation to study the heterotypic aggregation between PMNs and melanoma cells in the near-wall region of an in vitro parallel-plate flow chamber, which simulates in vivo cell-substrate adhesion from the vasculatures by combining mathematical modeling and numerical simulations with experimental observations. To the best of our knowledge, a multiscale near-wall aggregation model was developed, for the first time, which incorporated the effects of both cell deformation and general ratios of heterotypic cells on the cell aggregation process. Quantitative agreement was found between numerical predictions and in vitro experiments. The effects of factors, including: intrinsic binding molecule properties, near-wall heterotypic cell concentrations, and cell deformations on the coagulation process, are discussed. Several parameter identification approaches are proposed and validated which, in turn, demonstrate the importance of the reaction coefficient and the critical bond number on the aggregation process.

8.
Am J Physiol Cell Physiol ; 295(3): C701-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18632734

ABSTRACT

During their passage through the circulatory system, tumor cells undergo extensive interactions with various host cells including endothelial cells. The capacity of tumor cells to form metastasis is related to their ability to interact with and extravasate through endothelial cell layers, which involves multiple adhesive interactions between tumor cells and endothelium (EC). Thus it is essential to identify the adhesive receptors on the endothelial and melanoma surface that mediate those specific adhesive interactions. P-selectin and E-selectin have been reported as adhesion molecules that mediate the cell-cell interaction of endothelial cells and melanoma cells. However, not all melanoma cells express ligands for selectins. In this study, we elucidated the molecular constituents involved in the endothelial adhesion and extravasation of sialyl-Lewis(x/a)-negative melanoma cell lines under flow in the presence and absence of polymorphonuclear neutrophils (PMNs). Results show the interactions of alpha(4)beta(1) (VLA-4) on sialyl-Lewis(x/a)-negative melanoma cells and vascular adhesion molecule (VCAM-1) on inflamed EC supported melanoma adhesion to and subsequent extravasation through the EC in low shear flow. These findings provide clear evidence for a direct role of the VLA-4/VCAM-1 pathway in melanoma cell adhesion to and extravasation through the vascular endothelium in a shear flow. PMNs facilitated melanoma cell extravasation under both low and high shear conditions via the involvement of distinct molecular mechanisms. In the low shear regime, beta(2)-integrins were sufficient to enhance melanoma cell extravasation, whereas in the high shear regime, selectin ligands and beta(2)-integrins on PMNs were necessary for facilitating the melanoma extravasation process.


Subject(s)
Cell Adhesion , Cell Movement , Endothelial Cells/metabolism , Integrin alpha4beta1/metabolism , Melanoma/metabolism , CA-19-9 Antigen , CD18 Antigens/metabolism , Cell Line, Tumor , Endothelial Cells/pathology , Humans , Melanoma/pathology , Neoplasm Metastasis , Neutrophils/metabolism , Oligosaccharides/metabolism , Selectins/metabolism , Sialyl Lewis X Antigen , Stress, Mechanical , Vascular Cell Adhesion Molecule-1/metabolism
9.
Ann Biomed Eng ; 36(4): 661-71, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18253835

ABSTRACT

Adhesion to and subsequent extravasation through the endothelial lining of blood vessels is critical for tumor cells to establish metastases. Recent studies have indicated that polymorphonuclear neutrophils (PMNs) may enhance melanoma adhesion to the endothelium (EC) and subsequent extravasation under dynamic flow conditions. However, little is known about hydrodynamics involved in the tumor microenvironment within the microcirculation. In this study, effects of hydrodynamic flow on regulating melanoma cell adhesion to the EC have been investigated. Results indicate that under flow conditions, interactions between melanoma cells and the EC are distinctly different from PMN-EC interactions. Without expressions of surface integrins or sialylated molecules, most melanoma cells that express a high-level of intercellular adhesion molecule (ICAM-1) are not able to effectively adhere to the inflamed EC by themselves. Binding of melanoma cells and PMNs through ICAM-1 on melanoma cells and beta(2) integrins on PMNs has been shown to enhance melanoma cell arrest on the EC. Although PMN tethering on the EC is regulated by both the shear rate and shear stress, melanoma cell adhesion to the EC and subsequent extravasation via tethering PMN on the EC is predominantly regulated by shear rate, which partly is due to the shear-rate-dependent PMN-melanoma aggregation in shear flow. These findings provide a rationale and mechanistic basis for understanding of leukocyte-tumor cell interactions under flow conditions during tumor cell extravasation and metastasis.


Subject(s)
Blood Flow Velocity , Blood Pressure , Endothelium, Vascular/physiopathology , Leukocytes/metabolism , Melanoma/physiopathology , Melanoma/secondary , Neoplastic Cells, Circulating , Cell Adhesion , Cell Aggregation , Cells, Cultured , Humans , Intercellular Adhesion Molecule-1/metabolism , Mechanotransduction, Cellular , Melanoma/blood supply , Shear Strength
10.
Am J Physiol Cell Physiol ; 294(3): C743-53, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18199704

ABSTRACT

Cell adhesion, mediated by specific receptor-ligand interactions, plays an important role in biological processes such as tumor metastasis and inflammatory cascade. For example, interactions between beta 2-integrin (lymphocyte function-associated antigen-1 and/or Mac-1) on polymorphonuclear neutrophils (PMNs) and ICAM-1 on melanoma cells initiate the bindings of melanoma cells to PMNs within the tumor microenvironment in blood flow, which in turn activate PMN-melanoma cell aggregation in a near-wall region of the vascular endothelium, therefore enhancing subsequent extravasation of melanoma cells in the microcirculations. Kinetics of integrin-ligand bindings in a shear flow is the determinant of such a process, which has not been well understood. In the present study, interactions of PMNs with WM9 melanoma cells were investigated to quantify the kinetics of beta 2-integrin and ICAM-1 bindings using a cone-plate viscometer that generates a linear shear flow combined with a two-color flow cytometry technique. Aggregation fractions exhibited a transition phase where it first increased before 60 s and then decreased with shear durations. Melanoma-PMN aggregation was also found to be inversely correlated with the shear rate. A previously developed probabilistic model was modified to predict the time dependence of aggregation fractions at different shear rates and medium viscosities. Kinetic parameters of beta 2-integrin and ICAM-1 bindings were obtained by individual or global fittings, which were comparable to respectively published values. These findings provide new quantitative understanding of the biophysical basis of leukocyte-tumor cell interactions mediated by specific receptor-ligand interactions under shear flow conditions.


Subject(s)
CD18 Antigens/metabolism , Cell Adhesion , Intercellular Adhesion Molecule-1/metabolism , Melanoma/immunology , Neutrophil Activation , Neutrophils/immunology , Cell Aggregation , Cell Line, Tumor , Flow Cytometry , Hemorheology/methods , Humans , Kinetics , Melanoma/pathology , Models, Biological , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Activation/drug effects , Neutrophils/drug effects , Protein Binding , Stress, Mechanical
11.
Cell Mol Bioeng ; 1(2-3): 189-200, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-19865613

ABSTRACT

The primary cause of cancer mortality is not attributed to primary tumor formation, but rather to the growth of metastases at distant organ sites. Tumor cell adhesion to blood vessel endothelium (EC) and subsequent transendothelial migration within the circulation are critical components of the metastasis cascade. Previous studies have shown polymorphonuclear neutrophils (PMNs) may facilitate melanoma cell adhesion to the EC and subsequent extravasation under flow conditions. The melanoma cell-PMN interactions are found to be mediated by the binding between intercellular adhesion molecule-1 (ICAM-1) on melanoma cells and ß(2) integrin on PMNs and by endogenously secreted interleukin 8 (IL-8) within the tumor-leukocyte microenvironment. In this study, the effects of fluid convection on the IL-8-mediated activation of PMNs and the binding kinetics between PMNs and melanoma cells were investigated. Results indicate that the shear rate dependence of PMN-melanoma cell adhesion and melanoma cell extravasation is due, at least partly, to the convection of tumor-secreted proinflammatory cytokine IL-8.

12.
Cancer Res ; 67(12): 5814-20, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575149

ABSTRACT

Polymorphonuclear neutrophils (PMN) facilitate melanoma cell extravasation under dynamic flow conditions by the binding of intercellular adhesion molecule-1 (ICAM-1) on melanoma cells to beta2 integrins on PMNs, which is mediated by endogenously produced chemokine interleukin 8 (IL-8) from the tumor microenvironment. However, little is known about the role of B-Raf, the most mutated gene in malignant melanomas, in this process. In this study, we investigated the functional importance of B-Raf in melanoma extravasation by using short interfering RNA to reduce expression/activity of mutant (V600E)B-Raf in melanoma. Results indicated that knockdown of mutant (V600E)B-Raf inhibited melanoma cell extravasation in vitro and subsequent lung metastasis development in vivo. Mechanistic studies showed that inhibition of (V600E)B-Raf significantly reduced the constitutive secretion of IL-8 from melanoma cells as well as the capacity of endogenous IL-8 production from the melanoma-PMN microenvironment. Furthermore, a reduction in ICAM-1 expression on melanoma cells was detected following mutant (V600E)B-Raf knockdown. Together, these results suggest that targeting mutant (V600E)B-Raf reduces melanoma cell extravasation by decreasing IL-8 production and interrupting ICAM-1-beta2 integrin binding of melanoma cells to the endothelium mediated by PMNs in the microcirculation, which provides a rationale and mechanistic basis for targeting mutant (V600E)B-Raf to inhibit melanoma extravasation and subsequent metastasis development.


Subject(s)
Cell Movement/physiology , Melanoma, Experimental/pathology , Neoplasm Invasiveness/pathology , Neutrophils/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Animals , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Endothelium/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/metabolism , Lung Neoplasms/secondary , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Proto-Oncogene Proteins B-raf/genetics , RNA, Small Interfering
13.
Exp Cell Res ; 313(3): 551-9, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17141217

ABSTRACT

Inflammation facilitates tumor progression including metastasis. Interleukin-8 (IL-8) is a chemokine that regulates polymorphonuclear neutrophil (PMN) mobilization and activity and we hypothesize that this cytokine influences tumor behavior. We have demonstrated that IL-8 is crucial for PMN-mediated melanoma extravasation under flow conditions. In addition, IL-8 is up-regulated in PMNs upon co-culturing with melanoma cells. Melanoma cells induce IkappaB-alpha degradation in PMNs indicating that NF-kappaB signaling is active in PMNs. Furthermore, the production of IL-8 in PMNs is NF-kappaB dependent. We have further identified that interleukin-6 (IL-6) and interleukin-1beta (IL-1beta) from PMN-melanoma co-cultures synergistically contribute to IkappaB-alpha degradation and IL-8 synthesis in PMNs. Taken together, these findings show that melanoma cells induce PMNs to secrete IL-8 through activation of NF-kappaB and suggest a model in which this interaction promotes a microenvironment that is favorable for metastasis.


Subject(s)
Cell Movement , Inflammation/metabolism , Interleukin-8/metabolism , Melanoma/metabolism , Neutrophils/metabolism , Cell Communication , Cell Line, Tumor , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation/etiology , Melanoma/immunology , Melanoma/pathology , NF-kappa B/metabolism , Neutrophils/immunology , Transfection
14.
Cancer Res ; 66(16): 8200-9, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16912199

ABSTRACT

Malignant melanoma has a high propensity for metastatic spread, making it the most deadly form of skin cancer. B-RAF has been identified as the most mutated gene in these invasive cells and therefore an attractive therapeutic target. However, for uncertain reasons, chemotherapy inhibiting B-Raf has not been clinically effective. This has raised questions whether this pathway is important in melanoma metastasis or whether targeting a protein other than B-Raf in the signaling cascade could more effectively inhibit this pathway to reduce lung metastases. Here, we investigated the role played by (V600E)B-Raf in melanoma metastasis and showed that targeting this signaling cascade significantly reduces lung metastases. Small interfering RNA (siRNA)-mediated inhibition was used in mice to reduce expression (activity) of each member of the signaling cascade and effects on metastasis development were measured. Targeting any member of the signaling cascade reduced metastasis but inhibition of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (Mek) 1 and Mek 2 almost completely prevented lung tumor development. Mechanistically, metastatic inhibition was mediated through reduction of melanoma cell extravasation through the endothelium and decreased proliferative capacity. Targeting B-Raf with the pharmacologic inhibitor BAY 43-9006, which was found ineffective in clinical trials and seems to act primarily as an angiogenesis inhibitor, did not decrease metastasis, whereas inhibition of Mek using U0126 decreased cellular proliferative capacity, thereby effectively reducing number and size of lung metastases. In summary, this study provides a mechanistic basis for targeting Mek and not B-Raf in the mutant (V600E)B-Raf signaling cascade to inhibit melanoma metastases.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Lung Neoplasms/secondary , Melanoma/genetics , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Metastasis/prevention & control , Proto-Oncogene Proteins B-raf/genetics , Amino Acid Substitution , Animals , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Lung Neoplasms/prevention & control , Melanoma/pathology , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation , Nitriles/pharmacology , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , Signal Transduction
15.
Math Biosci Eng ; 3(4): 683-96, 2006 Oct.
Article in English | MEDLINE | ID: mdl-20361839

ABSTRACT

In this paper, we develop a population balance model for cell aggregation and adhesion process in a nonuniform shear flow. Some Monte Carlo simulation results based on the model are presented for the heterotypic cell-cell collision and adhesion to a substrate under dynamic shear forces. In particular, we focus on leukocyte (PMN)-melanoma cell emboli formation and subsequent tethering to the vascular endothelium (EC) as a result of cell-cell aggregation. The simulation results are compared with the results of experimental measurement. Discussions are made on how we could further improve the accuracy of the population balance type modelling.

16.
Exp Cell Res ; 310(2): 282-92, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16154563

ABSTRACT

Previous studies have shown that neutrophils (PMNs) facilitate melanoma cell extravasation [M.J. Slattery, C. Dong, Neutrophils influence melanoma adhesion and migration under flow conditions, Intl. J. Cancer 106 (2003) 713-722] Little is known, however, about the specific interactions between PMNs, melanoma and the endothelium (EC) or the molecular mechanism involved under flow conditions. The aim of this study is to investigate a "two-step adhesion" hypothesis that involves initial PMN tethering on the EC and subsequent melanoma cells being captured by tethered PMNs. Different effects of hydrodynamic shear stress and shear rate were analyzed using a parallel-plate flow chamber. Results indicate a novel finding that PMN-facilitated melanoma cell arrest on the EC is modulated by shear rate, which is inversely-proportional to cell-cell contact time, rather than by the shear stress, which is proportional to the force exerted on formed bonds. Beta2 integrins/ICAM-1 adhesion mechanisms were examined and the results indicate LFA-1 and Mac-1 cooperate to mediate the PMN-EC-melanoma interactions under shear conditions. In addition, endogenously produced IL-8 contributes to PMN-facilitated melanoma arrest on the EC through the CXC chemokine receptors 1 and 2 (CXCR1 and CXCR2) on PMN. These results provide new evidence for the complex role of hemodynamic forces, secreted chemokines and PMN-melanoma adhesion in the recruitment of metastatic cancer cells to the EC.


Subject(s)
Cell Adhesion Molecules/metabolism , Melanoma/metabolism , Neutrophils/metabolism , Stress, Mechanical , Cell Adhesion/drug effects , Culture Media/pharmacology , Dextrans/pharmacology , Endothelium/metabolism , Endothelium/pathology , Humans , Interleukin-8/metabolism , Melanoma/pathology , Neutrophils/drug effects , Neutrophils/pathology , Receptors, Interleukin-8A/agonists , Receptors, Interleukin-8B/agonists , Shear Strength , Tumor Cells, Cultured
17.
Mol Cell Biomech ; 2(3): 145-59, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16708476

ABSTRACT

Attachment of tumor cells to the endothelium (EC) under flow conditions is critical for the migration of tumor cells out of the vascular system to establish metastases. Innate immune system processes can potentially promote tumor progression through inflammation dependant mechanisms. White blood cells, neutrophils (PMN) in particular, are being studied to better understand how the host immune system affects cancer cell adhesion and subsequent migration and metastasis. Melanoma cell interaction with the EC is distinct from PMN-EC adhesion in the circulation. We found PMN increased melanoma cell extravasation, which involved initial PMN tethering on the EC, subsequent PMN capture of melanoma cells and maintaining close proximity to the EC. LFA-1 (CD11a/CD18 integrin) influenced the capture phase of PMN binding to both melanoma cells and the endothelium, while Mac-1 (CD11b/CD18 integrin) affected prolonged PMN-melanoma aggregation. Blocking E-selectin or ICAM-1 (intercellular adhesion molecule) on the endothelium or ICAM-1 on the melanoma surface reduced PMN-facilitated melanoma extravasation. Results indicated a novel finding that PMN-facilitated melanoma cell arrest on the EC could be modulated by endogenously produced interleukin-8 (IL-8). Functional blocking of the IL-8 receptors (CXCR1 and CXCR2) on PMN, or neutralizing soluble IL-8 in cell suspensions, significantly decreased the level of Mac-1 up-regulation on PMN while communicating with melanoma cells and reduced melanoma extravasation. These results provide new evidence for the complex role of hemodynamic forces, secreted chemokines, and PMN-melanoma adhesion in the recruitment of metastatic cancer cells to the endothelium in the microcirculation, which are significant in fostering new approaches to cancer treatment through anti-inflammatory therapeutics.


Subject(s)
Interleukin-8/biosynthesis , Melanoma/pathology , Neutrophils/metabolism , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Communication , Cell Movement , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Shear Strength , Tumor Cells, Cultured
18.
Am J Physiol Cell Physiol ; 288(4): C831-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15601752

ABSTRACT

Previously, we found polymorphonuclear neutrophils (PMNs) increased melanoma cell extravasation under flow conditions (Intl J Cancer 106: 713-722, 2003). In this study, we characterized the effect of hydrodynamic shear on PMN-facilitated melanoma extravasation using a novel flow-migration assay. The effect of shear stress and shear rate on PMN-facilitated melanoma extravasation was studied by increasing the medium viscosity with dextran to increase shear stress independently of shear rate. Under fixed shear rate conditions, melanoma cell extravasation did not change significantly. In contrast, the extravasation level increased at a fixed shear stress but with a decreasing shear rate. PMN-melanoma aggregation and adhesion to the endothelium via beta(2)-integrin/intracellular adhesion molecule-1 (ICAM-1) interactions were also studied. Lymphocyte function-associated molecule-1 (LFA-1; CD11a/CD18) influenced the capture phase of PMN binding to both melanoma cells and the endothelium, whereas Mac-1 (CD11b/CD18) affected prolonged PMN-melanoma aggregation. Blockage of E-selectin or ICAM-1 on the endothelium or ICAM-1 on the melanoma surface reduced PMN-facilitated melanoma extravasation. We have found PMN-melanoma adhesion is correlated with the inverse of shear rate, whereas the PMN-endothelial adhesion correlated with shear stress. Interleukin-8 (IL-8) also influenced PMN-melanoma cell adhesion. Functional blocking of the PMN IL-8 receptors, CXCR1 and CXCR2, decreased the level of Mac-1 upregulation on PMNs while in contact with melanoma cells and reduced melanoma extravasation. We have found PMN-facilitated melanoma adhesion to be a complex multistep process that is regulated by both microfluid mechanics and biology.


Subject(s)
Cell Movement/physiology , Endothelium, Vascular/physiology , Neoplasm Metastasis/pathology , Neutrophils/physiology , Cell Adhesion/physiology , Cell Adhesion Molecules/physiology , Flow Cytometry/instrumentation , Flow Cytometry/methods , Humans , Interleukin-8/metabolism , Melanoma/physiopathology , Stress, Mechanical , Tumor Cells, Cultured
19.
Front Biosci ; 10: 379-84, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15574376

ABSTRACT

Attachment of tumor cells to endothelial cells is critical for migration of tumor cells out of the vascular system to establish metastases. We have studied human melanoma cell (C8161) adhesion and migration in response to stimulation by soluble collagen IV (CIV) using a modified Boyden chamber. In this modified chamber, shear flow can be introduced over the cell-substrate interface affecting tumor cell chemotactic migration through a microporous filter. Our results suggest that transmigration of C8161 cells under flow conditions can be influenced by PMNs, mediated by Mac-1 and ICAM-1 adhesive interactions and enhanced by altered cytokine production.


Subject(s)
Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Humans , Intercellular Adhesion Molecule-1/metabolism , Leukocytes, Mononuclear/metabolism , Macrophage-1 Antigen/metabolism , Melanoma/metabolism , Melanoma/pathology , Neoplasm Metastasis , Neutrophils/metabolism , Neutrophils/pathology , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL
...