Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 1352, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409164

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) poses therapeutic challenges due to the limited treatment options. Building upon our previous research that demonstrates the efficacy of histone deacetylase 6 (HDAC6) inhibition in a genetic cardiomyopathy model, we investigate HDAC6's role in HFpEF due to their shared mechanisms of inflammation and metabolism. Here, we show that inhibiting HDAC6 with TYA-018 effectively reverses established heart failure and its associated symptoms in male HFpEF mouse models. Additionally, in male mice lacking Hdac6 gene, HFpEF progression is delayed and they are resistant to TYA-018's effects. The efficacy of TYA-018 is comparable to a sodium-glucose cotransporter 2 (SGLT2) inhibitor, and the combination shows enhanced effects. Mechanistically, TYA-018 restores gene expression related to hypertrophy, fibrosis, and mitochondrial energy production in HFpEF heart tissues. Furthermore, TYA-018 also inhibits activation of human cardiac fibroblasts and enhances mitochondrial respiratory capacity in cardiomyocytes. In this work, our findings show that HDAC6 impacts on heart pathophysiology and is a promising target for HFpEF treatment.


Subject(s)
Cardiomyopathies , Heart Failure , Animals , Humans , Male , Mice , Heart Failure/drug therapy , Heart Failure/genetics , Heart Failure/diagnosis , Histone Deacetylase 6/genetics , Myocytes, Cardiac/metabolism , Stroke Volume/physiology
2.
Anticancer Res ; 32(2): 537-52, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22287743

ABSTRACT

E-Cadherin and vimentin protein expression was assessed in late stage non-small cell lung cancer tumors from the placebo controlled clinical trial, NCIC-CTG BR.21, to determine if these markers had the potential to predict outcome of erlotinib therapy. E-Cadherin and vimentin protein expression levels were assessed in tumors from 95 patients, who were representative of the overall population, using semi-quantitative immunohistochemistry. The percentage of tumor cells with grades 0, 1, 2, or 3 membrane staining of E-cadherin and cytoplasmic staining of vimentin was measured. Three scoring methods and multiple cut-offs were explored to determine if these markers were able to divide patients into groups with different overall survival (OS). A cut-off point for E-cadherin of ≥40% tumor cells with staining of +2 and +3 and a cut-off for vimentin of ≥10% of tumors cell with any staining provided the optimal stratification. The OS hazard ratio (HR) for E-cadherin(+) versus E-cadherin(-) in the erlotinib-treated patients was 0.68 (0.35-1.33) compared with 1.48 (0.69-3.15) in the placebo patients and the OS (HR) for erlotinib versus placebo was 0.47 (0.26-0.88) in E-cadherin(+) patients compared with 1.12 (0.52-2.44) in the E-cadherin(-) patients. The OS (HR) for vimentin(+) versus vimentin(-) in the erlotinib-treated patients was 0.65 (0.31-1.38) compared to 2.32 (1.09-4.94) in the placebo-treated patients and the OS (HR) for erlotinib versus placebo was 0.26 (0.11-0.63) in vimentin(+) compared to 0.99 (0.55-1.76) in the vimentin(-) patients. Similar trends were observed for progression-free survival and response rate. E-Cadherin and vimentin are biomarkers worthy of additional study as predictive markers of outcome of erlotinib therapy.


Subject(s)
Biomarkers, Tumor/biosynthesis , Cadherins/biosynthesis , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Vimentin/biosynthesis , Adolescent , Adult , Aged , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease-Free Survival , Erlotinib Hydrochloride , Female , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Young Adult
3.
Clin Exp Metastasis ; 28(7): 593-614, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21643654

ABSTRACT

The progression of cancer from non-metastatic to metastatic is the critical transition in the course of the disease. The epithelial to mesenchymal transition (EMT) is a mechanism by which tumor cells acquire characteristics that improve metastatic efficiency. Targeting EMT processes in patients is therefore a potential strategy to block the transition to metastatic cancer and improve patient outcome. To develop models of EMT applicable to in vitro and in vivo settings, we engineered NCI-H358 non-small cell lung carcinoma cells to inducibly express three well-established drivers of EMT: activated transforming growth factor ß (aTGFß), Snail or Zeb1. We characterized the morphological, molecular and phenotypic changes induced by each of the drivers and compared the different end-states of EMT between the models. Both in vitro and in vivo, induction of the transgenes Snail and Zeb1 resulted in downregulation of epithelial markers and upregulation of mesenchymal markers, and reduced the ability of the cells to proliferate. Induced autocrine expression of aTGFß caused marker and phenotypic changes consistent with EMT, a modest effect on growth rate, and a shift to a more invasive phenotype. In vivo, this manifested as tumor cell infiltration of the surrounding mouse stromal tissue. Overall, Snail and Zeb1 were sufficient to induce EMT in the cells, but aTGFß induced a more complex EMT, in which changes in extracellular matrix remodeling components were pronounced.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Female , Homeodomain Proteins/genetics , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Phenotype , Snail Family Transcription Factors , Transcription Factors/genetics , Transgenes , Transplantation, Heterologous , Zinc Finger E-box-Binding Homeobox 1
4.
Clin Exp Metastasis ; 28(2): 137-55, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21194007

ABSTRACT

Epithelial-mesenchymal transition (EMT) is an important contributor to the invasion and metastasis of epithelial-derived cancers. While considerable effort has focused in the regulators involved in the transition process, we have focused on consequences of EMT to prosurvival signaling. Changes in distinct metastable and 'epigentically-fixed' EMT states were measured by correlation of protein, phosphoprotein, phosphopeptide and RNA transcript abundance. The assembly of 1167 modulated components into functional systems or machines simplified biological understanding and increased prediction confidence highlighting four functional groups: cell adhesion and migration, metabolism, transcription nodes and proliferation/survival networks. A coordinate metabolic reduction in a cluster of 17 free-radical stress pathway components was observed and correlated with reduced glycolytic and increased oxidative phosphorylation enzyme capacity, consistent with reduced cell cycling and reduced need for macromolecular biosynthesis in the mesenchymal state. An attenuation of EGFR autophosphorylation and a switch from autocrine to paracrine-competent EGFR signaling was implicated in the enablement of tumor cell chemotaxis. A similar attenuation of IGF1R, MET and RON signaling with EMT was observed. In contrast, EMT increased prosurvival autocrine IL11/IL6-JAK2-STAT signaling, autocrine fibronectin-integrin α5ß1 activation, autocrine Axl/Tyro3/PDGFR/FGFR RTK signaling and autocrine TGFßR signaling. A relatively uniform loss of polarity and cell-cell junction linkages to actin cytoskeleton and intermediate filaments was measured at a systems level. A more heterogeneous gain of ECM remodeling and associated with invasion and migration was observed. Correlation to stem cell, EMT, invasion and metastasis datasets revealed the greatest similarity with normal and cancerous breast stem cell populations, CD49f(hi)/EpCAM(-/lo) and CD44(hi)/CD24(lo), respectively.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Signal Transduction , Carcinoma, Non-Small-Cell Lung/pathology , Epithelial Cells/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Homeodomain Proteins/metabolism , Humans , Phosphorylation , Receptors, Fibroblast Growth Factor/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured , Zinc Finger E-box-Binding Homeobox 1
5.
Int J Biochem Cell Biol ; 43(2): 180-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20708092

ABSTRACT

There is increasing evidence that p38 MAPK, which is classified as a stress-activated kinase, also participates in cell cycle regulation, functioning as a suppressor of cell proliferation and tumorigenesis. We conducted a study of p38 MAPK phosphorylation during liver regeneration in mice to determine whether p38 MAPK activation or inactivation may correlate with events that lead to DNA replication after partial hepatectomy (PH), and whether p38 MAPK activation may be required for hepatocyte DNA replication in vivo and in culture. We report that active p38 (Pi-p38 MAPK) is present in normal liver, is rapidly inactivated starting 30 min after PH, and is re-activated by 12h. Although levels of Pi-MKK 3/6, the upstream kinases that activate p38 MAPK increase after PH, the expression of the dual protein phosphatase 1 is also elevated, and may be responsible for Pi-p38 MAPK dephosphorylation after PH. Inactivation and re-activation of p38 MAPK inversely correlates with the stimulation of protein synthesis and translation pathways, as indicated by activation of p70S6 kinase, increases in the phosphorylation of initiation factor elF-4E and translational repressor, 4E-BP. The activity of a p38 MAPK downstream substrate, MAPKAPK2 (MK2), did not reflect the changing levels of Pi-p38 MAPK during liver regeneration. Pi-p38 MAPK may be involved in TNF-stimulated DNA replication of murine hepatocytes in culture, but is not necessary for hepatocyte DNA replication after PH. Our results suggest that p38 MAPK inactivation plays a permissible role in DNA replication during liver regeneration and is consistent with a role for p38 MAPK in the maintenance of hepatocyte cell cycle arrest in adult liver.


Subject(s)
Liver Regeneration , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Cycle Proteins/biosynthesis , Cell Line , Cell Proliferation/drug effects , DNA Replication/drug effects , Dual Specificity Phosphatase 1/biosynthesis , Dual-Specificity Phosphatases/biosynthesis , Enzyme Activation , Enzyme Assays , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Liver/surgery , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NIH 3T3 Cells , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/pharmacology , Signal Transduction , Tumor Necrosis Factors/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Cells Tissues Organs ; 193(1-2): 114-32, 2011.
Article in English | MEDLINE | ID: mdl-21041998

ABSTRACT

Epithelial to mesenchymal transition (EMT) plays a dual role in tumor progression. It enhances metastasis of tumor cells by increasing invasive capacity and promoting survival, and it decreases tumor cell sensitivity to epithelial cell-targeting agents such as epithelial growth factor receptor kinase inhibitors. In order to study EMT in tumor cells, we have characterized 3 new models of ligand-driven EMT: the CFPAC1 pancreatic tumor model and the H358 and H1650 lung tumor models. We identified a diverse set of ligands that drives EMT in these models. Hepatocyte growth factor and oncostatin M induced EMT in all models, while transforming growth factor-ß induced EMT in both lung models. We observed morphologic, marker and phenotypic changes in response to chronic ligand treatment. Interestingly, stimulation with 2 ligands resulted in more pronounced EMT compared with single-ligand treatment, demonstrating a spectrum of EMT states induced by parallel signaling, such as the JAK and PI3K pathways. The EMT changes observed in response to the ligand were reversed upon ligand withdrawal, demonstrating the 'metastable' nature of these models. To study the impact of EMT on cell morphology and invasion in a 3D setting, we cultured cells in a semisolid basement membrane extract. Upon stimulation with EMT ligands, the colonies exhibited changes to EMT markers and showed phenotypes ranging from modest differences in colony architecture (CFPAC1) to complex branching structures (H358, H1650). Collectively, these 3 models offer robust cell systems with which to study the roles that EMT plays in cancer progression.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Hepatocyte Growth Factor/metabolism , Lung Neoplasms/metabolism , Oncostatin M/metabolism , Pancreatic Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Blotting, Western , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Fluorescent Antibody Technique , Hepatocyte Growth Factor/genetics , Humans , Lung Neoplasms/genetics , Microscopy, Confocal , Oncostatin M/genetics , Pancreatic Neoplasms/genetics , Polymerase Chain Reaction , Transforming Growth Factor beta/genetics
7.
Mol Cell Proteomics ; 8(10): 2308-20, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19620624

ABSTRACT

RhoA controls changes in cell morphology and invasion associated with cancer phenotypes. Cell lines derived from melanoma tumors at varying stages revealed that RhoA is selectively activated in cells of metastatic origin. We describe a functional proteomics strategy to identify proteins regulated by RhoA and report a previously uncharacterized human protein, named "mediator of RhoA-dependent invasion (MRDI)," that is induced in metastatic cells by constitutive RhoA activation and promotes cell invasion. In human melanomas, MRDI localization correlated with stage, showing nuclear localization in nevi and early stage tumors and cytoplasmic localization with plasma membrane accentuation in late stage tumors. Consistent with its role in promoting cell invasion, MRDI localized to cell protrusions and leading edge membranes in cultured cells and was required for cell motility, tyrosine phosphorylation of focal adhesion kinase, and modulation of actin stress fibers. Unexpectedly MRDI had enzymatic function as an isomerase that converts the S-adenosylmethionine catabolite 5-methylribose 1-phosphate into 5-methylribulose 1-phosphate. The enzymatic function of MRDI was required for methionine salvage from S-adenosylmethionine but distinct from its function in cell invasion. Thus, mechanisms used by signal transduction pathways to control cell movement have evolved from proteins with ancient function in amino acid metabolism.


Subject(s)
Aldose-Ketose Isomerases/metabolism , Melanoma , Methionine/metabolism , rhoA GTP-Binding Protein/metabolism , Aldose-Ketose Isomerases/genetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Enzyme Activation , Female , Humans , Melanoma/enzymology , Melanoma/pathology , Methionine/chemistry , Mice , Mice, Nude , Molecular Sequence Data , Molecular Structure , Neoplasm Invasiveness , Neoplasm Metastasis , Proteomics/methods , RNA Interference , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Signal Transduction/physiology , Transplantation, Heterologous , rhoA GTP-Binding Protein/genetics
8.
Science ; 320(5874): 365-9, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18420933

ABSTRACT

Mechanisms by which Wnt pathways integrate the organization of receptors, organelles, and cytoskeletal proteins to confer cell polarity and directional cell movement are incompletely understood. We show that acute responses to Wnt5a involve recruitment of actin, myosin IIB, Frizzled 3, and melanoma cell adhesion molecule into an intracellular structure in a melanoma cell line. In the presence of a chemokine gradient, this Wnt-mediated receptor-actin-myosin polarity (W-RAMP) structure accumulates asymmetrically at the cell periphery, where it triggers membrane contractility and nuclear movement in the direction of membrane retraction. The process requires endosome trafficking, is associated with multivesicular bodies, and is regulated by Wnt5a through the small guanosine triphosphatases Rab4 and RhoB. Thus, cell-autonomous mechanisms allow Wnt5a to control cell orientation, polarity, and directional movement in response to positional cues from chemokine gradients.


Subject(s)
Cell Polarity , Melanoma/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Wnt Proteins/metabolism , Actins/metabolism , Animals , CD146 Antigen/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Chemokine CXCL12/metabolism , Chemotaxis , Endosomes/metabolism , Golgi Apparatus/metabolism , Humans , Melanoma/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Nonmuscle Myosin Type IIB/metabolism , Transplantation, Heterologous , Wnt-5a Protein , rab4 GTP-Binding Proteins/metabolism , rhoB GTP-Binding Protein/metabolism
9.
Mol Cell Biochem ; 268(1-2): 129-40, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15724446

ABSTRACT

Pyridinyl imidazole inhibitors of p38 mitogen-activated protein kinase (MAPK) have been used extensively in vitro and in vivo to investigate the role of p38 in physiological processes. As with other pharmacological inhibitors, non-specific targets of the p38 inhibitors have been reported. We have found that the protein kinase receptor interacting protein-2 (RIP2) is another target for the family of p38 inhibitors. The autophosphorylation of RIP2 was inhibited in vitro by the p38 inhibitors SB220025, SB203580 and PD169316 at concentrations comparable to those used to inhibit p38. We also identified two new in vitro substrates for RIP2, myelin basic protein and histone H3 with apparent Km values of 2.1 microM and 0.65 microM, respectively. The ability of RIP2 to phosphorylate these two substrates was sensitive to the p38 inhibitors as well. As was shown for p38alpha, a conserved threonine in the kinase domain of RIP2 is required for sensitivity to the inhibitors, indicating that the mechanism of inhibition of RIP2 is similar to that of p38. These results demonstrate that the pyridinyl imidazole inhibitors block RIP2 as well as p38 kinase activity.


Subject(s)
Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Cell Line , Humans , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary/physiology , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Substrate Specificity , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Biol Chem ; 279(33): 34530-6, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15199050

ABSTRACT

Tumor necrosis factor (TNF) has multiple biological effects such as participating in inflammation, apoptosis, and cell proliferation, but the mechanisms of its effects on epithelial cell proliferation have not been examined in detail. At the early stages of liver regeneration, TNF functions as a priming agent for hepatocyte replication and increases the sensitivity of hepatocytes to growth factors such as transforming growth factor alpha (TGFalpha); however, the mechanisms by which TNF interacts with growth factors and enhances hepatocyte replication are not known. Using the AML-12 hepatocyte cell line, we show that TNF stimulates proliferation of these cells through transactivation of the epidermal growth factor receptor (EGFR). The transactivation mechanism involves the release of TGFalpha into the medium through activation of the metalloproteinase TNFalpha-converting enzyme (also known as ADAM 17). Binding of the ligand to EGFR initiates a mitogenic cascade through extracellular signal-regulated kinases 1 and 2 and the partial involvement of protein kinase B. TNF-induced release of TGFalpha and activation of EGFR signaling were inhibited by TNFalpha protease inhibitor-1, an agent that interferes with TNFalpha-converting enzyme activity. We suggest that TNF-induced transactivation of EGFR may provide an early signal for the entry of hepatocytes into the cell cycle and may integrate proliferative and survival pathways at the start of liver regeneration.


Subject(s)
ErbB Receptors/metabolism , Hepatocytes/metabolism , Transcriptional Activation , ADAM Proteins , ADAM17 Protein , Animals , Blotting, Western , Bromodeoxyuridine/pharmacology , Buffers , Cell Division , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Coloring Agents/pharmacology , Culture Media, Serum-Free/metabolism , DNA/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Humans , Ligands , Liver/physiology , Metalloendopeptidases/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Precipitin Tests , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Regeneration , Signal Transduction , Thymidine/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
11.
Cancer Res ; 63(20): 6716-25, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14583466

ABSTRACT

Functional proteomics provides a powerful approach to screen for alterations in protein expression and posttranslational modifications under conditions of human disease. In this study, we use protein screening to examine markers of melanoma progression, by profiling melanocyte versus melanoma cell lines using two-dimensional electrophoresis and mass spectrometry. Eight candidate markers were identified as differentially regulated in transformed cells. In particular, hepatoma-derived growth factor (HDGF) and nucleophosmin B23 were strongly correlated with melanoma. Nucleophosmin B23 is a nucleolar and centrosome-associated protein, which has been implicated as a target for cyclin E/cyclin-dependent kinase 2 (CDK2) in modulating centrosome duplication and cell cycle control. Western blotting of one-dimensional and two-dimensional gels showed that the form of nucleophosmin B23 that is up-regulated in melanoma represents a posttranslationally modified form, most likely reflecting enhanced phosphorylation in the tumor-derived cells. In contrast, Western analysis of HDGF demonstrated increased expression of all forms in melanoma cell lines compared with melanocytes. Immunohistochemical analysis of human tissue biopsies showed strong expression of HDGF in early and late stage melanomas and low expression in melanocytes and nontumorigenic nevi. Interestingly, biopsies of nevi showed a graded effect in which HDGF immunoreactivity was reduced in nevoid nests penetrating deep into the dermis compared with nests at the epidermal-dermal junction, suggesting that HDGF expression in nevi is dependent on epidermal cell interactions. In contrast, biopsies of melanoma showed strong expression of HDGF throughout the tumor, including cells located deeply within dermis. Thus, expression of this antigen likely reports a reduced dependence of protein expression on epidermal interactions.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Proteomics/methods , Biomarkers, Tumor/metabolism , Cathepsin D/biosynthesis , Cell Line, Tumor , Disease Progression , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Mass Spectrometry , Melanocytes/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL
...