Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Hepatol ; 66(1): 75-85, 2017 01.
Article in English | MEDLINE | ID: mdl-27520877

ABSTRACT

BACKGROUND & AIMS: We have established a clinically relevant animal model of hepatocellular cancer (HCC) in immune competent mice to elucidate the complex dialog between host immunity and tumors during HCC initiation and progression. Mechanistic findings have been leveraged to develop a clinically feasible anti-tumor chemoimmunotherapeutic strategy. METHODS: Intraperitoneal injection of carbon tetrachloride and intrasplenic inoculation of oncogenic hepatocytes were combined to induce progressive HCCs in fibrotic livers of immunocompetent mice. Immunization and adoptive cell transfer (ACT) were used to dissect the tumor antigen-specific immune response. The ability of the tyrosine kinase inhibitor sunitinib to enhance immunotherapy in the setting of HCC was evaluated. RESULTS: This new mouse model mimics human HCC and reflects its typical features. Tumor-antigen-specific CD8+ T cells maintained a naïve phenotype and remained responsive during early-stage tumor progression. Late tumor progression produced circulating tumor cells, tumor migration into draining lymph nodes, and profound exhaustion of tumor-antigen-specific CD8+ T cells associated with accumulation of programmed cell death protein 1 (PD-1)hi CD8+ T cells and regulatory T cells (Tregs). Sunitinib-mediated tumoricidal effect and Treg suppression synergized with antibody-mediated blockade of PD-1 to powerfully suppress tumor growth and activate anti-tumor immunity. CONCLUSION: Treg accumulation and upregulation of PD-1 provide two independent mechanisms to induce profound immune tolerance in HCC. Chemoimmunotherapy using Food and Drug Administration-approved sunitinib with anti-PD-1 antibodies achieved significant tumor control, supporting translation of this approach for the treatment of HCC patients. LAY SUMMARY: In the current study, we have established a clinically relevant mouse model which mimics human liver cancer. Using this unique model, we studied the response of the immune system to this aggressive cancer. Findings from this trial have led to the development of an innovative and clinically feasible chemoimmunotherapeutic strategy.


Subject(s)
Carcinoma, Hepatocellular , Immunotherapy/methods , Indoles/pharmacology , Liver Neoplasms , Pyrroles/pharmacology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cytotoxicity, Immunologic/physiology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Histocompatibility Antigens Class II/immunology , Immune Tolerance , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Neoplasm Staging , Programmed Cell Death 1 Receptor/metabolism , Sunitinib , T-Lymphocytes, Regulatory/immunology
2.
Sci Rep ; 6: 33098, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27608605

ABSTRACT

The understanding of domain structures, specifically domain walls, currently attracts a significant attention in the field of (multi)-ferroic materials. In this article, we analyze contrast formation in full field electron microscopy applied to domains and domain walls in the uniaxial ferroelectric lithium niobate, which presents a large 3.8 eV band gap and for which conductive domain walls have been reported. We show that the transition from Mirror Electron Microscopy (MEM - electrons reflected) to Low Energy Electron Microscopy (LEEM - electrons backscattered) gives rise to a robust contrast between domains with upwards (Pup) and downwards (Pdown) polarization, and provides a measure of the difference in surface potential between the domains. We demonstrate that out-of-focus conditions of imaging produce contrast inversion, due to image distortion induced by charged surfaces, and also carry information on the polarization direction in the domains. Finally, we show that the intensity profile at domain walls provides experimental evidence for a local stray, lateral electric field.

3.
Pharmaceuticals (Basel) ; 9(2)2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27023566

ABSTRACT

Hepatocellular carcinoma (HCC) is the third most deadly cancer in the world. New treatment strategies are desperately needed due to limited standard therapies. Activation of the Erk, Akt, and STAT3pathways is implicated in the prognosis of HCC. The Se,Se'-1,4-phenylenebis(1,2-ethanediyl) bisisoselenourea (PBISe), is a selenium-containing MAPK and PI3 kinase inhibitor, effectively inhibit tumorigenesis in a variety of experimental models. The aim of our study is to demonstrate the potential role of PBISe in the treatment of HCC. The anti-proliferative and pro-apoptotic ability of PBISe is studied in vitro in four human HCC cell lines and in vivo in a spontaneous murine HCC model. Inhibition of cancer growth was performed by cell viability assay and apoptosis by caspase 3/7, PARP cleavage, annexin-V, and TUNEL assays. Role of PBISe on PI3 kinase, MAPK and STAT3 signaling is determined by Western blotting. In vivo effects of PBISe on tumor sizes were monitored using MRI in a spontaneous murine HCC. Liver tissues from the PBISe-treated mice are analyzed for angiogenesis, proliferation, and signaling pathway markers. Overall, PBISe activated caspase-3/7 and increased DNA fragmentation, which is positively correlated with the increased PARP cleavage. PBISe promoted apoptosis by inhibiting PI3K, MAPK, and STAT3 signaling with significant reduction in the tumor sizes (p < 0.007). PBISe-treated tumors reduced survival marker PCNA, and angiogenesis markers Vegf-A, Vegf-R3 and CD34. These results demonstrate the chemotherapeutic effects of PBISe, by inhibiting tumor growth and facilitating tumor apoptosis for HCC treatment.

4.
PLoS One ; 10(5): e0128159, 2015.
Article in English | MEDLINE | ID: mdl-26000702

ABSTRACT

BACKGROUND: c-Met, a high-affinity receptor for Hepatocyte Growth Factor (HGF), plays a critical role in tumor growth, invasion, and metastasis. Hepatocellular carcinoma (HCC) patients with activated HGF/c-Met signaling have a significantly worse prognosis. Targeted therapies using c-Met tyrosine kinase inhibitors are currently in clinical trials for HCC, although receptor tyrosine kinase inhibition in other cancers has demonstrated early success. Unfortunately, therapeutic effect is frequently not durable due to acquired resistance. METHODS: We utilized the human MHCC97-H c-Met positive (c-Met+) HCC cell line to explore the compensatory survival mechanisms that are acquired after c-Met inhibition. MHCC97-H cells with stable c-Met knockdown (MHCC97-H c-Met KD cells) were generated using a c-Met shRNA vector with puromycin selection and stably transfected scrambled shRNA as a control. Gene expression profiling was conducted, and protein expression was analyzed to characterize MHCC97-H cells after blockade of the c-Met oncogene. A high-throughput siRNA screen was performed to find putative compensatory survival proteins, which could drive HCC growth in the absence of c-Met. Findings from this screen were validated through subsequent analyses. RESULTS: We have previously demonstrated that treatment of MHCC97-H cells with a c-Met inhibitor, PHA665752, results in stasis of tumor growth in vivo. MHCC97-H c-Met KD cells demonstrate slower growth kinetics, similar to c-Met inhibitor treated tumors. Using gene expression profiling and siRNA screening against 873 kinases and phosphatases, we identified ErbB3 and TGF-α as compensatory survival factors that are upregulated after c-Met inhibition. Suppressing these factors in c-Met KD MHCC97-H cells suppresses tumor growth in vitro. In addition, we found that the PI3K/Akt signaling pathway serves as a negative feedback signal responsible for the ErbB3 upregulation after c-Met inhibition. Furthermore, in vitro studies demonstrate that combination therapy with PHA665752 and Gefitinib (an EGFR inhibitor) significantly reduced cell viability and increased apoptosis compared with either PHA665752 or Gefitinib treatment alone. CONCLUSION: c-Met inhibition monotherapy is not sufficient to eliminate c-Met+ HCC tumor growth. Inhibition of both c-Met and EGFR oncogenic pathways provides superior suppression of HCC tumor growth. Thus, combination of c-Met and EGFR inhibition may represent a superior therapeutic regimen for c-Met+ HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Survival/physiology , ErbB Receptors/metabolism , Liver Neoplasms/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction/physiology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , ErbB Receptors/genetics , Female , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Indoles/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-3/genetics , Signal Transduction/drug effects , Sulfones/pharmacology
5.
PLoS One ; 7(11): e50401, 2012.
Article in English | MEDLINE | ID: mdl-23308088

ABSTRACT

UNLABELLED: Fibroblast Growth Factor (FGF)-10 promotes the proliferation and survival of murine hepatoblasts during early stages of hepatogenesis through a Wnt-ß-catenin dependent pathway. To determine the mechanism by which this occurs, we expanded primary culture of hepatoblasts enriched for progenitor markers CD133 and CD49f from embryonic day (E) 12.5 fetal liver and an established tumor initiating stem cell line from Mat1a(-/-) livers in media conditioned with recombinant (r) FGF10 or rFGF7. FGF Receptor (R) activation resulted in the downstream activation of MAPK, PI3K-AKT, and ß-catenin pathways, as well as cellular proliferation. Additionally, increased levels of nuclear ß-catenin phosphorylated at Serine-552 in cultured primary hepatoblasts, Mat1a(-/-) cells, and also in ex vivo embryonic liver explants indicate AKT-dependent activation of ß-catenin downstream of FGFR activation; conversely, the addition of AKT inhibitor Ly294002 completely abrogated ß-catenin activation. FGFR activation-induced cell proliferation and survival were also inhibited by the compound ICG-001, a small molecule inhibitor of ß-catenin-CREB Binding Protein (CBP) in hepatoblasts, further indicating a CBP-dependent regulatory mechanism of ß-catenin activity. CONCLUSION: FGF signaling regulates the proliferation and survival of embryonic and transformed progenitor cells in part through AKT-mediated activation of ß-catenin and downstream interaction with the transcriptional co-activator CBP.


Subject(s)
CREB-Binding Protein/metabolism , Liver Neoplasms/pathology , Liver/cytology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , beta Catenin/metabolism , Animals , Cell Cycle , Cell Proliferation , Cell Survival , Embryonic Stem Cells/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver/pathology , Methionine Adenosyltransferase/deficiency , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
6.
Hepatology ; 55(1): 141-52, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21898502

ABSTRACT

UNLABELLED: The high rate of mortality and frequent incidence of recurrence associated with hepatocellular carcinoma (HCC) reveal the need for new therapeutic approaches. In this study we evaluated the efficacy of a novel chemoimmunotherapeutic strategy to control HCC and investigated the underlying mechanism that increased the antitumor immune response. We developed a novel orthotopic mouse model of HCC through seeding of tumorigenic hepatocytes from SV40 T antigen (Tag) transgenic MTD2 mice into the livers of syngeneic C57BL/6 mice. These MTD2-derived hepatocytes form Tag-expressing HCC tumors specifically within the liver. This approach provides a platform to test therapeutic strategies and antigen-specific immune-directed therapy in an immunocompetent murine model. Using this model we tested the efficacy of a combination of oral sunitinib, a small molecule multitargeted receptor tyrosine kinase (RTK) inhibitor, and adoptive transfer of tumor antigen-specific CD8(+) T cells to eliminate HCC. Sunitinib treatment alone promoted a transient reduction in tumor size. Sunitinib treatment combined with adoptive transfer of tumor antigen-specific CD8(+) T cells led to elimination of established tumors without recurrence. In vitro studies revealed that HCC growth was inhibited through suppression of STAT3 signaling. In addition, sunitinib treatment of tumor-bearing mice was associated with suppression of STAT3 and a block in T-cell tolerance. CONCLUSION: These findings indicate that sunitinib inhibits HCC tumor growth directly through the STAT3 pathway and prevents tumor antigen-specific CD8(+) T-cell tolerance, thus defining a synergistic chemoimmunotherapeutic approach for HCC.


Subject(s)
Adoptive Transfer/methods , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Indoles/pharmacology , Liver Neoplasms/drug therapy , Pyrroles/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Combined Modality Therapy , Disease Models, Animal , Hep G2 Cells , Hepatocytes/immunology , Hepatocytes/transplantation , Humans , Immune Tolerance/immunology , Immunocompetence/immunology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Sunitinib
7.
Hepatology ; 55(1): 298-306, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22030746

ABSTRACT

Stem cells have potential for therapy of liver diseases, but may also be involved in the formation of liver cancer. Recently, the American Association for the Study of Liver Diseases Henry M. and Lillian Stratton Basic Research Single Topic Conference "Stem Cells in Liver Diseases and Cancer: Discovery and Promise" brought together a diverse group of investigators to define the status of research on stem cells and cancer stem cells in the liver and identify problems and solutions on the path to clinical translation. This report summarizes the outcomes of the conference and provides an update on recent research advances. Progress in liver stem cell research includes isolation of primary liver progenitor cells (LPCs), directed hepatocyte differentiation of primary LPCs and pluripotent stem cells, findings of transdifferentiation, disease-specific considerations for establishing a therapeutically effective cell mass, and disease modeling in cell culture. Tumor-initiating stem-like cells (TISCs) that emerge during chronic liver injury share the expression of signaling pathways, including those organized around transforming growth factor beta and ß-catenin, and surface markers with normal LPCs. Recent investigations of the role of TISCs in hepatocellular carcinoma have provided insight into the transcriptional and post-transcriptional regulation of hepatocarcinogenesis. Targeted chemotherapies for TISC are in development as a means to overcome cellular resistance and mechanisms driving disease progression in liver cancer.


Subject(s)
Liver Neoplasms/pathology , Liver Neoplasms/therapy , Neoplastic Stem Cells/pathology , Pluripotent Stem Cells/cytology , Stem Cell Transplantation/methods , Animals , Humans , Liver Diseases/pathology , Liver Diseases/therapy , Stem Cell Transplantation/trends , Translational Research, Biomedical/trends
8.
Oncogenesis ; 1: e15, 2012 Jun 11.
Article in English | MEDLINE | ID: mdl-23552699

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is associated with poor prognosis and metastasis in hepatocellular carcinoma. We have previously demonstrated an in vivo model of liver cancer in which mesenchymal cells post-EMT demonstrate a high rate of invasive growth and metastasis. Here, we investigate the role of microRNA 200 (miR-200) family members and epigenetic modifications on the maintenance of mesenchymal/metastatic phenotype after EMT. Mesenchymal cells post-EMT demonstrates high levels of E-box repressors Zeb1 and Zeb2 and downregulation of four miR-200 family members (miR-200a, miR-200b, miR-200c and miR-429). In addition, DNA sequencing after bisulfite modification demonstrates that several CpG sites within the E-cadherin promoter are methylated in mesenchymal cells. In mesenchymal cells, forced expression of miR-200b results in a significant increase in E-cadherin and a reduction in cell migration/invasion. Despite these mesenchymal-to-epithelial transition (MET) changes in vitro, there is no significant change in metastatic potential after miR-200b upregulation in vivo. After the mesenchymal cells were treated with combination of DNA methyltransferase (DNMT) inhibitor and upregulation of miR-200b, invasive phenotype was significantly reduced and metastatic potential was eliminated. Direct targeting of E-cadherin with short hairpin RNA does not restore metastatic potential after DNMT inhibition and miR-200b re-expression. In addition, restoration of E-cadherin alone was unable to block metastatic potential in primary mesenchymal cells. In conclusion, targeting mesenchymal liver cancer cells with miR-200b and DNMT inhibitor reduces metastatic potential irrespective of E-cadherin expression. Thus, the broader differentiation and MET effects of DNMT inhibition and miR-200b must be considered in terms of rescuing metastatic potential.

9.
J Vis Exp ; (56)2011 Oct 10.
Article in English | MEDLINE | ID: mdl-22006186

ABSTRACT

Liver stem cell, or oval cells, proliferate during chronic liver injury, and are proposed to differentiate into both hepatocytes and cholangiocytes. In addition, liver stem cells are hypothesized to be the precursors for a subset of liver cancer, Hepatocellular carcinoma. One of the primary challenges to stem cell work in any solid organ like the liver is the isolation of a rare population of cells for detailed analysis. For example, the vast majority of cells in the liver are hepatocytes (parenchymal fraction), which are significantly larger than non-parenchymal cells. By enriching the specific cellular compartments of the liver (i.e. parenchymal and non-parenchymal fractions), and selecting for CD45 negative cells, we are able to enrich the starting population of stem cells by over 600-fold.The proceduresdetailed in this report allow for a relatively rare population of cells from a solid organ to be sorted efficiently. This process can be utilized to isolateliver stem cells from normal murine liver as well as chronic liver injury models, which demonstrate increased liver stem cell proliferation. This method has clear advantages over standard immunohistochemistry of frozen or formalin fixed liver as functional studies using live cells can be performed after initial co-localization experiments. To accomplish the procedure outlined in this report, a working relationship with a research based flow-cytometry core is strongly encouraged as the details of FACS isolation are highly dependent on specialized instrumentation and a strong working knowledge of basic flow-cytometry procedures. The specific goal of this process is to isolate a population of liver stem cells that can be clonally expanded in vitro.


Subject(s)
Antigens, CD/biosynthesis , Cell Separation/methods , Flow Cytometry/methods , Glycoproteins/biosynthesis , Liver/cytology , Liver/immunology , Stem Cells/cytology , Stem Cells/immunology , AC133 Antigen , Animals , Clone Cells , Mice , Peptides
10.
BMC Cancer ; 11: 396, 2011 Sep 19.
Article in English | MEDLINE | ID: mdl-21929801

ABSTRACT

BACKGROUND: Tumor initiating stem-like cells (TISCs) are a subset of neoplastic cells that possess distinct survival mechanisms and self-renewal characteristics crucial for tumor maintenance and propagation. The induction of epithelial-mesenchymal-transition (EMT) by TGFß has been recently linked to the acquisition of TISC characteristics in breast cancer. In HCC, a TISC and EMT phenotype correlates with a worse prognosis. In this work, our aim is to elucidate the underlying mechanism by which cells acquire tumor initiating characteristics after EMT. METHODS: Gene and protein expression assays and Nanog-promoter luciferase reporter were utilized in epithelial and mesenchymal phenotype liver cancer cell lines. EMT was analyzed with migration/invasion assays. TISC characteristics were analyzed with tumor-sphere self-renewal and chemotherapy resistance assays. In vivo tumor assay was performed to investigate the role of Snail1 in tumor initiation. CONCLUSION: TGFß induced EMT in epithelial cells through the up-regulation of Snail1 in Smad-dependent signaling. Mesenchymal liver cancer post-EMT demonstrates TISC characteristics such as tumor-sphere formation but are not resistant to cytotoxic therapy. The inhibition of Snail1 in mesenchymal cells results in decreased Nanog promoter luciferase activity and loss of self-renewal characteristics in vitro. These changes confirm the direct role of Snail1 in some TISC traits. In vivo, the down-regulation of Snail1 reduced tumor growth but was not sufficient to eliminate tumor initiation. In summary, TGFß induces EMT and TISC characteristics through Snail1 and Nanog up-regulation. In mesenchymal cells post-EMT, Snail1 directly regulates Nanog expression, and loss of Snail1 regulates tumor growth without affecting tumor initiation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition/genetics , Neoplastic Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Homeodomain Proteins/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Nanog Homeobox Protein , Neoplastic Stem Cells/drug effects , Promoter Regions, Genetic , Signal Transduction , Smad Proteins , Snail Family Transcription Factors , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism
11.
Hepatology ; 54(3): 879-89, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21618573

ABSTRACT

UNLABELLED: c-Met, a high-affinity receptor for hepatocyte growth factor (HGF), plays a critical role in cancer growth, invasion, and metastasis. Hepatocellular carcinoma (HCC) patients with an active HGF/c-Met signaling pathway have a significantly worse prognosis. Although targeting the HGF/c-Met pathway has been proposed for the treatment of multiple cancers, the effect of c-Met inhibition in HCC remains unclear. The human HCC cell lines Huh7, Hep3B, MHCC97-L, and MHCC97-H were used in this study to investigate the effect of c-Met inhibition using the small molecule selective c-Met tyrosine kinase inhibitor PHA665752. MHCC97-L and MHCC97-H cells demonstrate a mesenchymal phenotype with decreased expression of E-cadherin and increased expression of c-Met, fibronectin, and Zeb2 compared with Huh7 and Hep3B cells, which have an epithelial phenotype. PHA665752 treatment blocked phosphorylation of c-Met and downstream phosphoinositide 3-kinase/Akt and mitogen-activated protein kinase/Erk pathways, inhibited cell proliferation, and induced apoptosis in c-Met-positive MHCC97-L and MHCC97-H cells. In xenograft models, administration of PHA665752 significantly inhibited c-Met-positive MHCC97-L and MHCC97-H tumor growth, and PHA665752-treated tumors demonstrated marked reduction of both c-Met phosphorylation and cell proliferation. c-Met-negative Huh7 and Hep3B cells were not affected by c-Met inhibitor treatment in vitro or in vivo. In addition, c-Met-positive MHCC97-L and MHCC97-H cells demonstrated cancer stem cell-like characteristics, such as resistance to chemotherapy, tumor sphere formation, and increased expression of CD44 and ABCG2, and PHA665752 treatment suppressed tumor sphere formation and inhibited CD44 expression. CONCLUSION: c-Met represents a potential target of personalized treatment for HCC with an active HGF/c-Met pathway.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Indoles/therapeutic use , Liver Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Sulfones/therapeutic use , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Liver Neoplasms/pathology , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Phosphorylation , Precision Medicine , Proto-Oncogene Proteins c-met/metabolism
12.
Phys Rev Lett ; 106(16): 165504, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21599383

ABSTRACT

We measure the thickness of the heavy water layer trapped under the stress corrosion fracture surface of silica using neutron reflectivity experiments. We show that the penetration depth is 65-85   Å, suggesting the presence of a damaged zone of ∼100   Šextending ahead of the crack tip during its propagation. This estimate of the size of the damaged zone is compatible with other recent results.

13.
Genome Med ; 3(3): 15, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21418554

ABSTRACT

A report on the Stem Cells World Congress held in San Diego, USA, 24-25 January 2011.

14.
Proteome Sci ; 8: 61, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-21092202

ABSTRACT

BACKGROUND: Protein biomarkers will play a pivotal role in the future of personalized medicine for both diagnosis and treatment decision-making. While the results of several pre-clinical and small-scale clinical studies have demonstrated the value of protein biomarkers, there have been significant challenges to translating these findings into routine clinical care. Challenges to the use of protein biomarkers include inter-sample variability introduced by differences in post-collection handling and ex vivo degradation of proteins and protein modifications. RESULTS: In this report, we re-create laboratory and clinical scenarios for sample collection and test the utility of a new tissue stabilization technique in preserving proteins and protein modifications. In the laboratory setting, tissue stabilization with the Denator Stabilizor T1 resulted in a significantly higher yield of phospho-protein when compared to standard snap freeze preservation. Furthermore, in a clinical scenario, tissue stabilization at collection resulted in a higher yield of total phospho-protein, total phospho-tyrosine, pErkT202/Y204 and pAktS473 when compared to standard methods. Tissue stabilization did not have a significant effect on other post-translational modifications such as acetylation and glycosylation, which are more stable ex-vivo. Tissue stabilization did decrease total RNA quantity and quality. CONCLUSION: Stabilization at the time of collection offers the potential to better preserve tissue protein and protein modification levels, as well as reduce the variability related to tissue processing delays that are often associated with clinical samples.

15.
Gastroenterology ; 139(6): 2170-82, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20837017

ABSTRACT

BACKGROUND & AIMS: The tumor suppressor PTEN inhibits AKT2 signaling; both are aberrantly expressed in liver tumors. We investigated how PTEN and AKT2 regulate liver carcinogenesis. Loss of PTEN leads to spontaneous development of liver tumors from progenitor cells. We investigated how the loss of PTEN activates liver progenitor cells and induces tumorigenesis. METHODS: We studied mice with liver-specific disruptions in Pten and the combination of Pten and Akt2 to investigate mechanisms of liver carcinogenesis. RESULTS: PTEN loss leads to hepatic injury and establishes selective pressure for tumor-initiating cells (TICs), which proliferate to form mixed-lineage tumors. The Pten-null mice had increasing levels of hepatic injury before proliferation of hepatic progenitors. Attenuation of hepatic injury by deletion of Akt2 reduced progenitor cell proliferation and delayed tumor development. In Pten/Akt2-null mice given 3,5-diethoxycarbonyl-1,4 dihydrocollidine (DDC), we found that the primary effect of AKT2 loss was attenuation of hepatic injury and not inhibition of progenitor-cell proliferation in response to injury. CONCLUSIONS: Liver carcinogenesis in Pten-null mice requires not only the transformation of TICs but selection pressure from hepatic injury and cell death, which activates TICs. Further research is required to elucidate the mechanism for hepatic injury and its relationship with TIC activation.


Subject(s)
Chemical and Drug Induced Liver Injury , Liver Neoplasms , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Cell Survival/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Gene Expression Regulation, Neoplastic/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pyridines/toxicity , Signal Transduction/physiology , Stem Cells/pathology
16.
Hepatology ; 52(3): 945-53, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20564331

ABSTRACT

UNLABELLED: Epithelial-to-mesenchymal transition (EMT) is predicted to play a critical role in metastatic disease in hepatocellular carcinoma. In this study, we used a novel murine model of EMT to elucidate a mechanism of tumor progression and metastasis. A total of 2 x 10(6) liver cells isolated from Pten(loxp/loxp)/Alb-Cre(+) mice, expanded from a single CD133(+)CD45(-) cell clone, passage 0 (P0), were sequentially transplanted to obtain two passages of tumor cells, P1 and P2. Cells were analyzed for gene expression using microarray and real-time polymerase chain reaction. Functional analysis included cell proliferation, migration, and invasion in vitro and orthotopic tumor metastasis assays in vivo. Although P0, P1, and P2 each formed tumors consistent with mixed liver epithelium, within the P2 cells, two distinct cell types were clearly visible: cells with epithelial morphology similar to P0 cells and cells with fibroblastoid morphology. These P2 mesenchymal cells demonstrated increased locomotion on wound healing; increased cell invasion on Matrigel basement membrane; increased EMT-associated gene expression of Snail1, Zeb1, and Zeb2; and down-regulated E-cadherin. P2 mesenchymal cells demonstrated significantly faster tumor growth in vivo compared with P2 epithelial counterparts, with invasion of intestine, pancreas, spleen, and lymph nodes. Furthermore, P2 mesenchymal cells secreted high levels of hepatocyte growth factor (HGF), which we propose acts in a paracrine fashion to drive epithelial cells to undergo EMT. In addition, a second murine liver cancer stem cell line with methionine adenosyltransferase 1a deficiency acquired EMT after sequential transplantations, indicating that EMT was not restricted to Pten-deleted tumors. CONCLUSION: EMT is associated with a high rate of liver tumor proliferation, invasion, and metastasis in vivo, which is driven by HGF secreted from mesenchymal tumor cells in a feed-forward mechanism.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Liver Neoplasms/pathology , Mesoderm/pathology , AC133 Antigen , Animals , Antigens, CD/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Movement/physiology , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , Glycoproteins/metabolism , Hepatocyte Growth Factor/metabolism , Liver Neoplasms/metabolism , Mesoderm/metabolism , Methionine Adenosyltransferase/genetics , Methionine Adenosyltransferase/metabolism , Mice , Mice, Knockout , Mice, Nude , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Peptides/metabolism , Signal Transduction/physiology
17.
Phys Rev Lett ; 104(2): 025502, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20366607

ABSTRACT

The roughness of fracture surfaces exhibits self-affinity for a wide variety of materials and loading conditions. The universality and the range of scales over which this regime extends are still debated. The topography of these surfaces is however often investigated with a finite contact probe. In this case, we show that the correlation function of the roughness can only be measured down to a length scale Deltax{c} which depends on the probe size R, the Hurst exponent zeta of the surface and its topothesy l, and exhibits spurious behavior at smaller scales. First, we derive the dependence of Deltax{c} on these parameters from a simple scaling argument. Then, we verify this dependence numerically. Finally, we establish the relevance of this analysis from AFM measurements on an experimental glass fracture surface and provide a metrological procedure for roughness measurements.

18.
Hepatology ; 51(5): 1635-44, 2010 May.
Article in English | MEDLINE | ID: mdl-20196115

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality worldwide. CD133, a transmembrane glycoprotein, is an important cell surface marker for both stem cells and cancer stem cells in various tissues including liver. CD133 expression has been recently linked to poor prognosis in HCC patients. CD133+ liver cancer cells are characterized by resistance to chemotherapy, self-renewal, multilineage potential, increased colony formation, and in vivo cancer initiation at limited dilution. Recent studies demonstrate that CD133 expression is regulated by DNA methylation. In this study, we explored the role of transforming growth factor beta (TGFbeta), a multifunctional cytokine that plays a critical role in chronic liver injury, in the regulation of CD133 expression. TGFbeta1 is capable of up-regulating CD133 expression specifically within the Huh7 HCC cell line in a time- and dose-dependent manner. Most important, TGFbeta1-induced CD133+ Huh7 cells demonstrate increased tumor initiation in vivo. Forced expression of inhibitory Smads, including Smad6 and Smad7, attenuated TGFbeta1-induced CD133 expression. Within CD133- Huh7 cells, TGFbeta1 stimulation inhibited the expression of DNA methyltransferases (DNMT) 1 and DNMT3beta, which are critical in the maintenance of regional DNA methylation, and global DNMT activity in CD133- Huh7 cells was inhibited by TGFbeta1. DNMT3beta inhibition by TGFbeta1 was partially rescued with overexpression of inhibitory Smads. Lastly, TGFbeta1 treatment led to significant demethylation in CD133 promoter-1 in CD133- Huh7 cells. CONCLUSION: TGFbeta1 is able to regulate CD133 expression through inhibition of DNMT1 and DNMT3beta expression and subsequent demethylation of promoter-1. TGFbeta1-induced CD133+ Huh7 cells are tumorigenic. The mechanism by which TGFbeta induces CD133 expression is partially dependent on the Smads pathway.


Subject(s)
Antigens, CD/genetics , Carcinoma, Hepatocellular/metabolism , Epigenesis, Genetic/physiology , Glycoproteins/genetics , Liver Neoplasms/metabolism , Peptides/genetics , Transforming Growth Factor beta/pharmacology , AC133 Antigen , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , Humans , Neoplastic Stem Cells/metabolism , Smad Proteins/physiology , Up-Regulation , DNA Methyltransferase 3B
19.
Phys Rev Lett ; 102(19): 195501, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19518968

ABSTRACT

Amorphous silica density at ambient pressure is known to depend on thermal history (through the quenching rate) but also, at room temperature, on the maximum pressure applied in the past. Here we show that beyond density, a mechanical loading can endow the structure with an orientational order. Molecular dynamics simulations show evidence that amorphous silica develops a permanent anisotropic structure after extended shear plastic flow. This anisotropy which survives for an unstressed specimen is revealed markedly by the fabric tensor computed over the Si-O-Si orientations, albeit the SiO4 tetrahedra microstructure remains mostly unaltered.

20.
Stem Cells ; 27(2): 290-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19008348

ABSTRACT

PTEN (phosphatase and tensin homolog deleted on chromosome 10) is a lipid phosphatase that regulates mitogenic signaling pathways, and deficiency of PTEN results in cell proliferation, survival, and malignancy. Murine liver-specific Pten deletion models develop liver malignancy by 12 months of age. Using this model, we describe a population of CD133+ liver cancer stem cells isolated during the chronic injury phase of disease progression and before primary carcinoma formation. We performed immunohistochemistry and flow cytometry isolation using livers from 3- and 6-month-old Pten(loxP/loxP); Alb-Cre+ mice (mutants) and controls. CD133+CD45- nonparenchymal (NP) cells were analyzed for gene expression profile and protein levels. Single CD133+CD45- oval cells were isolated for clonal expansion and tumor analysis. Cultured and freshly isolated liver CD133+CD45- and CD133-CD45- NP cells were injected into immune-deficient and immune-competent mice. In mutant mice, the NP fraction increased in CD133+CD45- cells in 3- and 6-month-old Pten-deleted animals compared with controls. Clone lines expanded from single CD133+CD45- cells demonstrated consistent liver progenitor cell phenotype, with bilineage gene expression of hepatocyte and cholangiocyte markers. CD133+ cells from expanded clone lines formed robust tumors in immune-deficient and immune-competent mice. Furthermore, freshly isolated CD133+CD45- NP liver cells from 6-month-old mutants formed tumors in vivo, and CD133-CD45- NP cells did not. Consistent with a cancer stem cell phenotype, CD133+ cells demonstrate resistance to chemotherapy agents compared with CD133- cells. CD133+CD45- nonparenchymal cells from chronic injury Pten(loxP/loxP); Alb-Cre+ mice represent a bipotent liver progenitor cell population with cancer stem cell phenotype.


Subject(s)
Antigens, CD/metabolism , Chromosomes, Mammalian/genetics , Glycoproteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplastic Stem Cells/metabolism , PTEN Phosphohydrolase/genetics , Peptides/metabolism , AC133 Antigen , Animals , Blotting, Western , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Flow Cytometry , Immunohistochemistry , Leukocyte Common Antigens/metabolism , Mice , Mice, Mutant Strains , Mice, Nude , Neoplastic Stem Cells/pathology , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...