Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
EMBO J ; 39(1): e102406, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31782549

ABSTRACT

The Hippo pathway, which plays a critical role in organ size control and cancer, features numerous WW domain-based protein-protein interactions. However, ~100 WW domains and 2,000 PY motif-containing peptide ligands are found in the human proteome, raising a "WW-PY" binding specificity issue in the Hippo pathway. In this study, we have established the WW domain binding specificity for Hippo pathway components and uncovered a unique amino acid sequence required for it. By using this criterion, we have identified a WW domain-containing protein, STXBP4, as a negative regulator of YAP. Mechanistically, STXBP4 assembles a protein complex comprising α-catenin and a group of Hippo PY motif-containing components/regulators to inhibit YAP, a process that is regulated by actin cytoskeleton tension. Interestingly, STXBP4 is a potential tumor suppressor for human kidney cancer, whose downregulation is correlated with YAP activation in clear cell renal cell carcinoma. Taken together, our study not only elucidates the WW domain binding specificity for the Hippo pathway, but also reveals STXBP4 as a player in actin cytoskeleton tension-mediated Hippo pathway regulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , Vesicular Transport Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Proliferation , Female , Hippo Signaling Pathway , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Protein Binding , Protein Serine-Threonine Kinases/genetics , Survival Rate , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription, Genetic , Tumor Cells, Cultured , Vesicular Transport Proteins/genetics , WW Domains , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
2.
Am J Pathol ; 190(2): 484-502, 2020 02.
Article in English | MEDLINE | ID: mdl-31843498

ABSTRACT

Targeted therapeutics are needed for triple-negative breast cancer (TNBC). In this study, we investigated the activation of Src family of cytoplasmic tyrosine kinases (SFKs) and two SFK substrates-CUB-domain containing protein 1 (CDCP1) and protein kinase C δ (PKCδ)-in 56 formalin-fixed, paraffin-embedded (FFPE) TNBCs. Expression of SFK phosphorylated at Y416 (SFK_pY416+) in tumor cells was strongly associated with phosphorylation of CDCP1 and PKCδ (CDCP1_ pY743+ and PKCδ_pY311+), as assessed by immunohistochemistry, indicating increased SFK activity in situ. To enable biochemical analysis, protein extraction from FFPE tissue was optimized. Cleaved CDCP1 isoform (70 kDa) was expressed to a varying degree in all samples but only phosphorylated in TNBC tumor cells that expressed SFK_pY416. Interestingly, active SFK was found to be biphosphorylated (SFK_pY416+/pY527+). Biphosphorylated active SFK was observed more frequently in forkhead box protein A1 (FOXA1)- TNBCs. In addition, in SFK_pY416- samples, FOXA1+ TNBC tended to be SFK_pY527+ (classic inactive SFK), and FOXA1- TNBC tended to be SFK_pY527- (SFK poised for activation). Strong SFK_pY416 staining was also observed in tumor-infiltrating lymphocytes in a subset of TNBCs with high tumor-infiltrating lymphocyte content. This report will facilitate protein biochemical analysis of FFPE tumor samples and justifies the development of therapies targeting the SFK/CDCP1/PKCδ pathway for TNBC treatment.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Cell Adhesion Molecules/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Protein Kinase C-delta/metabolism , Triple Negative Breast Neoplasms/pathology , Tyrosine/metabolism , src-Family Kinases/metabolism , Aged , Cell Adhesion , Cell Movement , Female , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Middle Aged , Phosphorylation , Prognosis , Signal Transduction , Triple Negative Breast Neoplasms/classification , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Vimentin/metabolism
3.
Proc Natl Acad Sci U S A ; 114(32): E6556-E6565, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28739932

ABSTRACT

Triple-negative breast cancer (TNBC) is notoriously aggressive with high metastatic potential, which has recently been linked to high rates of fatty acid oxidation (FAO). Here we report the mechanism of lipid metabolism dysregulation in TNBC through the prometastatic protein, CUB-domain containing protein 1 (CDCP1). We show that a "low-lipid" phenotype is characteristic of breast cancer cells compared with normal breast epithelial cells and negatively correlates with invasiveness in 3D culture. Using coherent anti-Stokes Raman scattering and two-photon excited fluorescence microscopy, we show that CDCP1 depletes lipids from cytoplasmic lipid droplets (LDs) through reduced acyl-CoA production and increased lipid utilization in the mitochondria through FAO, fueling oxidative phosphorylation. These findings are supported by CDCP1's interaction with and inhibition of acyl CoA-synthetase ligase (ACSL) activity. Importantly, CDCP1 knockdown increases LD abundance and reduces TNBC 2D migration in vitro, which can be partially rescued by the ACSL inhibitor, Triacsin C. Furthermore, CDCP1 knockdown reduced 3D invasion, which can be rescued by ACSL3 co-knockdown. In vivo, inhibiting CDCP1 activity with an engineered blocking fragment (extracellular portion of cleaved CDCP1) lead to increased LD abundance in primary tumors, decreased metastasis, and increased ACSL activity in two animal models of TNBC. Finally, TNBC lung metastases have lower LD abundance than their corresponding primary tumors, indicating that LD abundance in primary tumor might serve as a prognostic marker for metastatic potential. Our studies have important implications for the development of TNBC therapeutics to specifically block CDCP1-driven FAO and oxidative phosphorylation, which contribute to TNBC migration and metastasis.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Fatty Acids/metabolism , Lipid Droplets/metabolism , Neoplasm Proteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Antigens, CD/genetics , Antigens, Neoplasm , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Fatty Acids/genetics , HEK293 Cells , Heterografts , Humans , Lipid Droplets/pathology , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Oxidation-Reduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
4.
Nucleic Acids Res ; 45(7): 3674-3692, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28073943

ABSTRACT

The p53 tumor suppressor protein plays a critical role in orchestrating the genomic response to various stress signals by acting as a master transcriptional regulator. Differential gene activity is controlled by transcription factors but also dependent on the underlying chromatin structure, especially on covalent histone modifications. After screening different histone lysine methyltransferases and demethylases, we identified JMJD2B/KDM4B as a p53-inducible gene in response to DNA damage. p53 directly regulates JMJD2B gene expression by binding to a canonical p53-consensus motif in the JMJD2B promoter. JMJD2B induction attenuates the transcription of key p53 transcriptional targets including p21, PIG3 and PUMA, and this modulation is dependent on the catalytic capacity of JMJD2B. Conversely, JMJD2B silencing led to an enhancement of the DNA-damage driven induction of p21 and PIG3. These findings indicate that JMJD2B acts in an auto-regulatory loop by which p53, through JMJD2B activation, is able to influence its own transcriptional program. Functionally, exogenous expression of JMJD2B enhanced subcutaneous tumor growth of colon cancer cells in a p53-dependent manner, and genetic inhibition of JMJD2B impaired tumor growth in vivo. These studies provide new insights into the regulatory effect exerted by JMJD2B on tumor growth through the modulation of p53 target genes.


Subject(s)
DNA Damage , Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Cells, Cultured , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/biosynthesis , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mutagens/toxicity , Neoplasms/pathology , Promoter Regions, Genetic , Transcriptional Activation
5.
Nature ; 466(7302): 133-7, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20596026

ABSTRACT

The question of whether tumorigenic cancer stem cells exist in human melanomas has arisen in the last few years. Here we show that in melanomas, tumour stem cells (MTSCs, for melanoma tumour stem cells) can be isolated prospectively as a highly enriched CD271(+) MTSC population using a process that maximizes viable cell transplantation. The tumours sampled in this study were taken from a broad spectrum of sites and stages. High-viability cells isolated by fluorescence-activated cell sorting and re-suspended in a matrigel vehicle were implanted into T-, B- and natural-killer-deficient Rag2(-/-)gammac(-/-) mice. The CD271(+) subset of cells was the tumour-initiating population in 90% (nine out of ten) of melanomas tested. Transplantation of isolated CD271(+) melanoma cells into engrafted human skin or bone in Rag2(-/-)gammac(-/-) mice resulted in melanoma; however, melanoma did not develop after transplantation of isolated CD271(-) cells. We also show that in mice, tumours derived from transplanted human CD271(+) melanoma cells were capable of metastatsis in vivo. CD271(+) melanoma cells lacked expression of TYR, MART1 and MAGE in 86%, 69% and 68% of melanoma patients, respectively, which helps to explain why T-cell therapies directed at these antigens usually result in only temporary tumour shrinkage.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/metabolism , Neural Crest/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Antigens, Neoplasm/analysis , Antigens, Neoplasm/metabolism , Bone Transplantation , Bone and Bones/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Humans , Lung Neoplasms/secondary , Melanoma-Specific Antigens , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/transplantation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neural Crest/cytology , Neural Crest/pathology , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Skin/pathology , Skin Transplantation , Transplantation, Heterologous/pathology
6.
Yale J Biol Med ; 88(2): 145-55, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26029013

ABSTRACT

Targeting synthetic lethal interactions is a promising new therapeutic approach to exploit specific changes that occur within cancer cells. Multiple approaches to investigate these interactions have been developed and successfully implemented, including chemical, siRNA, shRNA, and CRISPR library screens. Genome-wide computational approaches, such as DAISY, also have been successful in predicting synthetic lethal interactions from both cancer cell lines and patient samples. Each approach has its advantages and disadvantages that need to be considered depending on the cancer type and its molecular alterations. This review discusses these approaches and examines case studies that highlight their use.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Genetic Therapy/methods , Molecular Targeted Therapy/methods , Neoplasms/physiopathology , Neoplasms/therapy , Animals , Drug Discovery , Humans , Neoplasms/pathology
7.
Proc Natl Acad Sci U S A ; 108(5): 1931-6, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21233420

ABSTRACT

A common genetic mutation found in clear cell renal cell carcinoma (CC-RCC) is the loss of the von Hippel-Lindau (VHL) gene, which results in stabilization of hypoxia-inducible factors (HIFs), and contributes to cancer progression and metastasis. CUB-domain-containing protein 1 (CDCP1) was shown to promote metastasis in scirrhous and lung adenocarcinomas as well as in prostate cancer. In this study, we established a molecular mechanism linking VHL loss to induction of the CDCP1 gene through the HIF-1/2 pathway in renal cancer. Also, we report that Fyn, which forms a complex with CDCP1 and mediates its signaling to PKCδ, is a HIF-1 target gene. Mechanistically, we found that CDCP1 specifically regulates phosphorylation of PKCδ, but not of focal adhesion kinase or Crk-associated substrate. Signal transduction from CDCP1 to PKCδ leads to its activation, increasing migration of CC-RCC. Furthermore, patient survival can be stratified by CDCP1 expression at the cell surface of the tumor. Taken together, our data indicates that CDCP1 protein might serve as a therapeutic target for CC-RCC.


Subject(s)
Antigens, CD/physiology , Carcinoma, Renal Cell/genetics , Cell Adhesion Molecules/physiology , Kidney Neoplasms/genetics , Neoplasm Proteins/physiology , Protein Kinase C-delta/metabolism , Up-Regulation , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Antigens, CD/genetics , Antigens, Neoplasm , Cell Adhesion Molecules/genetics , Humans , Mutation , Neoplasm Proteins/genetics , Phosphorylation , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein/genetics
8.
bioRxiv ; 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38948754

ABSTRACT

Severe invagination of the nuclear envelope is a hallmark of cancers, aging, neurodegeneration, and infections. However, the outcomes of nuclear invagination remain unclear. This work identified a new function of nuclear invagination: regulating ribosome biogenesis. With expansion microscopy, we observed frequent physical contact between nuclear invaginations and nucleoli. Surprisingly, the higher the invagination curvature, the more ribosomal RNA and pre-ribosomes are made in the contacted nucleolus. By growing cells on nanopillars that generate nuclear invaginations with desired curvatures, we can increase and decrease ribosome biogenesis. Based on this causation, we repressed the ribosome levels in breast cancer and progeria cells by growing cells on low-curvature nanopillars, indicating that overactivated ribosome biogenesis can be rescued by reshaping nuclei. Mechanistically, high-curvature nuclear invaginations reduce heterochromatin and enrich nuclear pore complexes, which promote ribosome biogenesis. We anticipate that our findings will serve as a foundation for further studies on nuclear deformation.

9.
Cell Cycle ; 21(10): 1103-1119, 2022 05.
Article in English | MEDLINE | ID: mdl-35240916

ABSTRACT

Clear cell renal cell carcinoma (CC-RCC) remains one of the most deadly forms of kidney cancer despite recent advancements in targeted therapeutics, including tyrosine kinase and immune checkpoint inhibitors. Unfortunately, these therapies have not been able to show better than a 16% complete response rate. In this study we evaluated a cyclin-dependent kinase inhibitor, Dinaciclib, as a potential new targeted therapeutic for CC-RCC. In vitro, Dinaciclib showed anti-proliferative and pro-apoptotic effects on CC-RCC cell lines in Cell Titer Glo, Crystal Violet, FACS-based cell cycle analysis, and TUNEL assays. Additionally, these responses were accompanied by a reduction in phospho-Rb and pro-survival MCL-1 cell signaling responses, as well as the induction of caspase 3 and PARP cleavage. In vivo, Dinaciclib efficiently inhibited primary tumor growth in an orthotopic, patient-derived xenograft-based CC-RCC mouse model. Importantly, Dinaciclib targeted both CD105+ cancer stem cells (CSCs) and CD105- non-CSCs in vivo. Moreover, normal cell lines, as well as a CC-RCC cell line with re-expressed von-Hippel Lindau (VHL) tumor suppressor gene, were protected from Dinaciclib-induced cytotoxicity when not actively dividing, indicating an effective therapeutic window due to synthetic lethality of Dinaciclib treatment with VHL loss. Thus, Dinaciclib represents a novel potential therapeutic for CC-RCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cyclic N-Oxides , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinases/genetics , Female , Humans , Indolizines , Kidney Neoplasms/pathology , Male , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridinium Compounds , Synthetic Lethal Mutations , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
10.
Small GTPases ; 11(1): 32-38, 2020 01.
Article in English | MEDLINE | ID: mdl-28632992

ABSTRACT

The loss of the von Hippel-Lindau (VHL) tumor-suppressor is a major driver of Clear Cell Renal Cell Carcinoma (CC-RCC) resulting in the stabilization and overactivation of hypoxia inducible factors (HIFs). ROCK1 is a well-known protein serine/threonine kinase which is recognized as having a role in cancer including alterations in cell motility, metastasis and angiogenesis. We recently investigated and identified a synthetic lethal interaction between VHL loss and ROCK1 inhibition in CC-RCC that is dependent on HIF overactivation. Increased expression and activity of both HIFs and ROCK1 occurs in many types of cancer supporting the potential therapeutic role of ROCK inhibitors beyond CC-RCC. We also discuss future research required to establish prognostic markers to predict tumor response to ROCK inhibitors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Humans , Neoplasms/metabolism , Neoplasms/pathology
11.
Mol Cancer Ther ; 17(8): 1781-1792, 2018 08.
Article in English | MEDLINE | ID: mdl-29720560

ABSTRACT

Clear cell renal cell carcinoma (CC-RCC) is a devastating disease with limited therapeutic options available for advanced stages. The objective of this study was to investigate HMG-CoA reductase inhibitors, also known as statins, as potential therapeutics for CC-RCC. Importantly, treatment with statins was found to be synthetically lethal with the loss of the von Hippel-Lindau (VHL) tumor suppressor gene, which occurs in 90% of CC-RCC driving the disease. This effect has been confirmed in three different CC-RCC cell lines with three different lipophilic statins. Inhibition of mevalonate synthesis by statins causes a profound cytostatic effect at nanomolar concentrations and becomes cytotoxic at low micromolar concentrations in VHL-deficient CC-RCC. The synthetic lethal effect can be fully rescued by both mevalonate and geranylgeranylpyrophosphate, but not by squalene, indicating that the effect is due to disruption of small GTPase isoprenylation and not the inhibition of cholesterol synthesis. Inhibition of Rho and Rho kinase (ROCK) signaling contributes to the synthetic lethality effect, and overactivation of hypoxia-inducible factor signaling resulting from VHL loss is required. Finally, statin treatment is able to inhibit both tumor initiation and progression of subcutaneous 786-OT1-based CC-RCC tumors in mice. Thus, statins represent potential therapeutics for the treatment of VHL-deficient CC-RCC. Mol Cancer Ther; 17(8); 1781-92. ©2018 AACR.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , Mevalonic Acid/therapeutic use , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kidney Neoplasms/pathology , Mevalonic Acid/pharmacology
12.
FASEB J ; 19(9): 1158-60, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15833768

ABSTRACT

Expression of cell adhesion molecules (CAM) responsible for leukocyte-endothelium interactions plays a crucial role in inflammation and atherogenesis. Up-regulation of vascular CAM-1 (VCAM-1), intracellular CAM-1 (ICAM-1), and E-selectin expression promotes monocyte recruitment to sites of injury and is considered to be a critical step in atherosclerotic plaque development. Factors that trigger this initial response are not well understood. As platelet activation not only promotes thrombosis but also early stages of atherogenesis, we considered the role of thrombospondin-1 (TSP-1), a matricellular protein released in abundance from activated platelets and accumulated in sites of vascular injury, as a regulator of CAM expression. TSP-1 induced expression of VCAM-1 and ICAM-1 on endothelium of various origins, which in turn, resulted in a significant increase of monocyte attachment. This effect could be mimicked by a peptide derived from the C-terminal domain of TSP-1 and known to interact with CD47 on the cell surface. The essential role of CD47 in the cellular responses to TSP-1 was demonstrated further using inhibitory antibodies and knockdown of CD47 with small interfering RNA. Furthermore, we demonstrated that secretion of endogenous TSP-1 and its interaction with CD47 on the cell surface mediates endothelial response to the major proinflammatory agent, tumor necrosis factor alpha (TNF-alpha). Taken together, this study identifies a novel mechanism regulating CAM expression and subsequent monocyte binding to endothelium, which might influence the development of anti-atherosclerosis therapeutic strategies.


Subject(s)
Cell Adhesion Molecules/genetics , Endothelial Cells/metabolism , Gene Expression Regulation , Monocytes/physiology , Thrombospondin 1/physiology , Atherosclerosis/etiology , CD47 Antigen/physiology , Cell Adhesion , Cells, Cultured , E-Selectin/genetics , Endothelial Cells/cytology , Gene Expression Regulation/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , NF-kappa B/metabolism , RNA, Small Interfering/pharmacology , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , Vascular Cell Adhesion Molecule-1/genetics
13.
Cancer Res ; 64(6): 1951-8, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15026329

ABSTRACT

Renal cell carcinoma (RCC) rarely acquires mutations in p53 tumor suppressor gene, suggesting that p53 signaling in this tumor type might be repressed by some other mechanism. In fact, all four RCC-derived cell lines we tested maintained wild-type p53 but were not capable of transactivating p53-responsive reporters and endogenous p53-responsive genes. p53 protein in RCC showed normal response to genotoxic stress, including accumulation, nuclear translocation, and activation of specific DNA binding. Functional and expression analysis of Mdm2, MdmX, and Arf showed lack of involvement of these p53 regulators in the observed defect of p53 function in RCC. However, activation of p53-mediated transactivation could be achieved by extremely high levels of p53 attained by lentivirus vector-driven transduction, suggesting the involvement of a dominant inhibitor in repression of p53-dependent transactivation in RCC. Consistently, p53 inactivation prevailed in the hybrids of RCC cells with the cells possessing fully functional p53. Remarkably, cells of normal kidney epithelium also caused partial p53 repression in cell fusion experiments, suggesting that RCC-specific p53 repression may be based on an unknown dominant mechanism also acting in normal kidney tissue.


Subject(s)
Carcinoma, Renal Cell/metabolism , Genes, Dominant , Kidney Neoplasms/metabolism , Nuclear Proteins , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Carcinoma, Renal Cell/genetics , Cell Fusion , Cell Nucleus/metabolism , DNA Damage , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Neoplastic , Humans , Hybrid Cells , Kidney/metabolism , Kidney Neoplasms/genetics , Lentivirus/genetics , Luciferases , Promoter Regions, Genetic , Protein Transport , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Retroviridae/genetics , Transcriptional Activation , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
14.
J Biomed Opt ; 21(6): 60503, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27300321

ABSTRACT

Two-photon excited fluorescence (TPEF) imaging of the cellular cofactors nicotinamide adenine dinucleotide and oxidized flavin adenine dinucleotide is widely used to measure cellular metabolism, both in normal and pathological cells and tissues. When dual-wavelength excitation is used, ratiometric TPEF imaging of the intrinsic cofactor fluorescence provides a metabolic index of cells­the "optical redox ratio" (ORR). With increased interest in understanding and controlling cellular metabolism in cancer, there is a need to evaluate the performance of ORR in malignant cells. We compare TPEF metabolic imaging with seahorse flux analysis of cellular oxygen consumption in two different breast cancer cell lines (MCF-7 and MDA-MB-231). We monitor metabolic index in living cells under both normal culture conditions and, for MCF-7, in response to cell respiration inhibitors and uncouplers. We observe a significant correlation between the TPEF-derived ORR and the flux analyzer measurements (R=0.7901, p<0.001). Our results confirm that the ORR is a valid dynamic index of cell metabolism under a range of oxygen consumption conditions relevant for cancer imaging.


Subject(s)
Breast Neoplasms/diagnostic imaging , Optical Imaging , Oxygen Consumption , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Oxidation-Reduction
15.
Acta Biomater ; 43: 122-138, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27475528

ABSTRACT

UNLABELLED: Human neural stem/progenitor cells (hNSPCs) are good candidates for treating central nervous system (CNS) trauma since they secrete beneficial trophic factors and differentiate into mature CNS cells; however, many cells die after transplantation. This cell death can be ameliorated by inclusion of a biomaterial scaffold, making identification of optimal scaffolds for hNSPCs a critical research focus. We investigated the properties of fibrin-based scaffolds and their effects on hNSPCs and found that fibrin generated from salmon fibrinogen and thrombin stimulates greater hNSPC proliferation than mammalian fibrin. Fibrin scaffolds degrade over the course of a few days in vivo, so we sought to develop a novel scaffold that would retain the beneficial properties of fibrin but degrade more slowly to provide longer support for hNSPCs. We found combination scaffolds of salmon fibrin with interpenetrating networks (IPNs) of hyaluronic acid (HA) with and without laminin polymerize more effectively than fibrin alone and generate compliant hydrogels matching the physical properties of brain tissue. Furthermore, combination scaffolds support hNSPC proliferation and differentiation while significantly attenuating the cell-mediated degradation seen with fibrin alone. HNSPCs express two fibrinogen-binding integrins, αVß1 and α5ß1, and several laminin binding integrins (α7ß1, α6ß1, α3ß1) that can mediate interaction with the scaffold. Lastly, to test the ability of scaffolds to support vascularization, we analyzed human cord blood-derived endothelial cells alone and in co-culture with hNSPCs and found enhanced vessel formation and complexity in co-cultures within combination scaffolds. Overall, combination scaffolds of fibrin, HA, and laminin are excellent biomaterials for hNSPCs. STATEMENT OF SIGNIFICANCE: Interest has increased recently in the development of biomaterials as neural stem cell transplantation scaffolds to treat central nervous system (CNS) injury since scaffolds improve survival and integration of transplanted cells. We report here on a novel combination scaffold composed of fibrin, hyaluronic acid, and laminin to support human neural stem/progenitor cell (hNSPC) function. This combined biomaterial scaffold has appropriate physical properties for hNSPCs and the CNS, supports hNSPC proliferation and differentiation, and attenuates rapid cell-mediated scaffold degradation. The hNSPCs and scaffold components synergistically encourage new vessel formation from human endothelial cells. This work marks the first report of a combination scaffold supporting human neural and vascular cells to encourage vasculogenesis, and sets a benchmark for biomaterials to treat CNS injury.


Subject(s)
Blood Vessels/physiology , Fibrin/pharmacology , Hyaluronic Acid/pharmacology , Laminin/pharmacology , Neural Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Blood Vessels/drug effects , Cattle , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Integrins/metabolism , Neovascularization, Physiologic/drug effects , Neural Stem Cells/drug effects , Polymerization/drug effects , Salmon
16.
Mol Cell Biol ; 34(4): 739-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344197

ABSTRACT

The induction of hypoxia-inducible factors (HIFs) is essential for the adaptation of tumor cells to a low-oxygen environment. We found that the expression of the apoptosis inhibitor ARC (apoptosis repressor with a CARD domain) was induced by hypoxia in a variety of cancer cell types, and its induction is primarily HIF1 dependent. Chromatin immunoprecipitation (ChIP) and reporter assays also indicate that the ARC gene is regulated by direct binding of HIF1 to a hypoxia response element (HRE) located at bp -190 upstream of the transcription start site. HIFs play an essential role in the pathogenesis of renal cell carcinoma (RCC) under normoxic conditions, through the loss of the Von Hippel-Lindau gene (VHL). Accordingly, our results show that ARC is not expressed in normal renal tissue but is highly expressed in 65% of RCC tumors, which also express high levels of carbonic anhydrase IX (CAIX), a HIF1-dependent protein. Compared to controls, ARC-deficient RCCs exhibited decreased colony formation and increased apoptosis in vitro. In addition, loss of ARC resulted in a dramatic reduction of RCC tumor growth in SCID mice in vivo. Thus, HIF-mediated increased expression of ARC in RCC can explain how loss of VHL can promote survival early in tumor formation.


Subject(s)
Apoptosis/physiology , Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , AIDS-Related Complex/genetics , Animals , Apoptosis/genetics , Carcinoma, Renal Cell/genetics , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Proliferation , Cell Survival , Cells, Cultured , Gene Expression Regulation, Neoplastic/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
17.
Cancer Res ; 72(8): 2111-9, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22354749

ABSTRACT

Mammalian Bre1 complexes (BRE1A/B (RNF20/40) in humans and Bre1a/b (Rnf20/40) in mice) function similarly to their yeast homolog Bre1 as ubiquitin ligases in monoubiquitination of histone H2B. This ubiquitination facilitates methylation of histone H3 at K4 and K79, and accounts for the roles of Bre1 and its homologs in transcriptional regulation. Recent studies by others suggested that Bre1 acts as a tumor suppressor, augmenting expression of select tumor suppressor genes and suppressing select oncogenes. In this study, we present an additional mechanism of tumor suppression by Bre1 through maintenance of genomic stability. We track the evolution of genomic instability in Bre1-deficient cells from replication-associated double-strand breaks (DSB) to specific genomic rearrangements that explain a rapid increase in DNA content and trigger breakage-fusion-bridge cycles. We show that aberrant RNA-DNA structures (R-loops) constitute a significant source of DSBs in Bre1-deficient cells. Combined with a previously reported defect in homologous recombination, generation of R-loops is a likely initiator of replication stress and genomic instability in Bre1-deficient cells. We propose that genomic instability triggered by Bre1 deficiency may be an important early step that precedes acquisition of an invasive phenotype, as we find decreased levels of BRE1A/B and dimethylated H3K79 in testicular seminoma and in the premalignant lesion in situ carcinoma.


Subject(s)
Chromosomal Instability/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis/genetics , Blotting, Western , Cell Proliferation , DNA Breaks, Double-Stranded , Gene Knockdown Techniques , Humans , Male , Mice , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA3 , Seminoma/genetics , Seminoma/metabolism , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Tissue Array Analysis , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
19.
Radiat Res ; 174(5): 558-65, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20738173

ABSTRACT

The pathway involving Bre1-dependent monoubiquitination of histone H2B lysine 123, which leads to Dot1-dependent methylation of histone H3 lysine 79 (H3K79me2), has been implicated in survival after exposure to ionizing radiation in Saccharomyces cerevisiae. We found that depletion of mammalian homologs of Bre1 compromises the response to ionizing radiation, leading to increased radiosensitivity and a G(2)/M checkpoint defect. The deficiency in Bre1a/b function was also associated with increased sensitivity to crosslinking drugs and defective formation of Rad51 foci in mouse cells, suggesting a defect in homologous recombinational repair analogous to that seen in Saccharomyces. In budding yeast, H3K79me2 is important for the recruitment of the checkpoint signaling protein Rad9 to sites of double-strand breaks (DSBs). However, in mammalian cells, 53BP1 (the Rad9 ortholog) in addition to H3K79me2 recognizes a different residue, H4K20me2, and some studies argue that it is H4K20me2 and not H3K79me2 that is the preferred target for 53BP1. We show here that depletion of Bre1b specifically reduced dimethylation of H3K79 without affecting dimethylation of H4K20. Thus our data suggest that the observed defects in the radiation response of Bre1a/b-deficient cells are associated with reduced H3K79me2 and not with H4K20me2.


Subject(s)
Cell Cycle/radiation effects , Recombination, Genetic/radiation effects , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Animals , Base Sequence , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Division/radiation effects , Cell Line, Tumor , Chlorambucil/pharmacology , G2 Phase/drug effects , G2 Phase/genetics , G2 Phase/radiation effects , Gene Knockdown Techniques , Histones/metabolism , Humans , Methylation/drug effects , Methylation/radiation effects , Mice , RNA, Small Interfering/genetics , Rad51 Recombinase/metabolism , Radiation Tolerance/genetics , Recombination, Genetic/drug effects , Recombination, Genetic/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Ubiquitination/genetics , Ubiquitination/radiation effects
20.
Cell Cycle ; 7(3): 335-47, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18287811

ABSTRACT

Integrins, transmembrane glycoprotein receptors, play vital roles in pathological angiogenesis, but their precise regulatory functions are not completely understood and remain controversial. This study aims to assess the regulatory functions of individual beta subunits of endothelial integrins in angiogenic responses induced by vascular endothelial growth factor (VEGF). Inhibition of expression of beta(1), beta(3), or beta(5) integrins in endothelial cells resulted in down regulation of EC adhesion and migration on the primary ligand for the corresponding integrin receptor, while no effects on the recognition of other ligands were detected. Although inhibition of expression of each subunit substantially affected capillary growth stimulated by VEGF, the loss of beta(3) integrin was the most inhibitory.


Subject(s)
Integrins/metabolism , Neovascularization, Pathologic/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Integrins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL