Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
2.
Elife ; 112022 10 10.
Article in English | MEDLINE | ID: mdl-36214451

ABSTRACT

The differentiation of oligodendroglia from oligodendrocyte precursor cells (OPCs) to complex and extensive myelinating oligodendrocytes (OLs) is a multistep process that involves large-scale morphological changes with significant strain on the cytoskeleton. While key chromatin and transcriptional regulators of differentiation have been identified, their target genes responsible for the morphological changes occurring during OL myelination are still largely unknown. Here, we show that the regulator of focal adhesion, Tensin3 (Tns3), is a direct target gene of Olig2, Chd7, and Chd8, transcriptional regulators of OL differentiation. Tns3 is transiently upregulated and localized to cell processes of immature OLs, together with integrin-ß1, a key mediator of survival at this transient stage. Constitutive <i>Tns3</i> loss of function leads to reduced viability in mouse and humans, with surviving knockout mice still expressing Tns3 in oligodendroglia. Acute deletion of <i>Tns3</i> in vivo, either in postnatal neural stem cells (NSCs) or in OPCs, leads to a twofold reduction in OL numbers. We find that the transient upregulation of Tns3 is required to protect differentiating OPCs and immature OLs from cell death by preventing the upregulation of p53, a key regulator of apoptosis. Altogether, our findings reveal a specific time window during which transcriptional upregulation of Tns3 in immature OLs is required for OL differentiation likely by mediating integrin-ß1 survival signaling to the actin cytoskeleton as OL undergo the large morphological changes required for their terminal differentiation.


Subject(s)
Focal Adhesions , Tumor Suppressor Protein p53 , Humans , Animals , Mice , Focal Adhesions/metabolism , Tumor Suppressor Protein p53/genetics , Oligodendroglia/metabolism , Cell Differentiation/genetics , Mice, Knockout , Transcription Factors/metabolism , Chromatin/metabolism , Integrins/metabolism
3.
N Engl J Med ; 386(22): 2112-2119, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35648703

ABSTRACT

A patient with progressive metastatic pancreatic cancer was treated with a single infusion of 16.2×109 autologous T cells that had been genetically engineered to clonally express two allogeneic HLA-C*08:02-restricted T-cell receptors (TCRs) targeting mutant KRAS G12D expressed by the tumors. The patient had regression of visceral metastases (overall partial response of 72% according to the Response Evaluation Criteria in Solid Tumors, version 1.1); the response was ongoing at 6 months. The engineered T cells constituted more than 2% of all the circulating peripheral-blood T cells 6 months after the cell transfer. In this patient, TCR gene therapy targeting the KRAS G12D driver mutation mediated the objective regression of metastatic pancreatic cancer. (Funded by the Providence Portland Medical Foundation.).


Subject(s)
Genetic Therapy , Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Receptors, Antigen, T-Cell , Genes, T-Cell Receptor/genetics , Genetic Therapy/methods , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/therapeutic use , Pancreatic Neoplasms
4.
Thyroid ; 32(7): 789-798, 2022 07.
Article in English | MEDLINE | ID: mdl-35587601

ABSTRACT

Background: Medullary thyroid cancer (MTC) is a rare malignancy originating from the calcitonin-producing C cells of the thyroid. Despite recent therapeutic advances, metastatic MTC remains incurable. Adoptive cell therapy (ACT) using genetically engineered T cells targeting either tissue-restricted tumor-associated antigens or mutated neoantigens has led to durable remissions in other metastatic solid tumors. The majority of MTC express the tumor-associated antigens calcitonin and carcinoembryonic antigen (CEA), and ∼40% of MTC harbor the RET M918T oncogenic driver mutation. Methods: We developed and characterized three immunoreceptors that recognize extracellular CEA, a calcitonin epitope presented by HLA-A*24:02, or an RET M918T neoepitope restricted by HLA-DPB1*04:01/02. The chimeric antigen receptor (CAR) targeting CEA was synthetically designed, while the T cell receptors (TCRs) targeting calcitonin and RET M918T were isolated from a transgenic mouse and patient with MTC, respectively. These immunoreceptors were genetically engineered into peripheral blood T cells and tested for antigen specificity and antitumor activity. Results: T cells expressing the anti-CEA CAR or the calcitonin-reactive TCR produced effector cytokines and displayed cytotoxicity against cell lines expressing their cognate antigen in vitro. In immunodeficient mice harboring a human MTC cell line, the adoptive transfer of T cells engineered to express the anti-CEA CAR or calcitonin-reactive TCR led to complete tumor regression. T cells expressing the HLA-DPB1*04:01/02-restricted TCR targeting RET M918T, which was cloned from peripheral blood CD4+ T cells of a patient with MTC, demonstrated specific reactivity against cells pulsed with the mutated peptide and MTC tumor cells that expressed HLA-DPB1*04:01 and RET M918T. Conclusion: The preclinical data presented herein demonstrate the potential of using genetically engineered T cells targeting CEA, calcitonin, and/or RET M918T to treat metastatic MTC.


Subject(s)
Calcitonin , Carcinoembryonic Antigen , Cell Engineering , Proto-Oncogene Proteins c-ret , Receptors, Antigen, T-Cell , T-Lymphocytes , Animals , Calcitonin/genetics , Calcitonin/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/therapy , Cell Line, Tumor , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Mice , Mutation , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/therapy
5.
Cancer Cell ; 40(4): 410-423.e7, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35413272

ABSTRACT

Tumor-infiltrating neoantigen-reactive T cells can mediate regression of metastatic gastrointestinal cancers yet remain poorly characterized. We performed immunological screening against personalized neoantigens in combination with single-cell RNA sequencing on tumor-infiltrating lymphocytes from bile duct and pancreatic cancer patients to characterize the transcriptomic landscape of neoantigen-reactive T cells. We found that most neoantigen-reactive CD8+ T cells displayed an exhausted state with significant CXCL13 and GZMA co-expression compared with non-neoantigen-reactive bystander cells. Most neoantigen-reactive CD4+ T cells from a patient with bile duct cancer also exhibited an exhausted phenotype but with overexpression of HOPX or ADGRG1 while lacking IL7R expression. Thus, neoantigen-reactive T cells infiltrating gastrointestinal cancers harbor distinct transcriptomic signatures, which may provide new opportunities for harnessing these cells for therapy.


Subject(s)
CD8-Positive T-Lymphocytes , Gastrointestinal Neoplasms , Antigens, Neoplasm , Gastrointestinal Neoplasms/genetics , Humans , Lymphocytes, Tumor-Infiltrating , Transcriptome
6.
Biochim Biophys Acta Mol Cell Res ; 1866(3): 450-458, 2019 03.
Article in English | MEDLINE | ID: mdl-30321615

ABSTRACT

Spatial and temporal subcellular localization plays critical roles in regulating protein function. Cten (C-terminal tensin like) is a member of the tensin family. Cten recruits signaling molecules, such as DLC1, to focal adhesions, modulates homeostasis of receptor tyrosine kinases, including EGFR and c-Met, and promotes cell migration. These functions are likely controlled by Cten localization at focal adhesions and/or in the cytoplasm. In addition, Cten has been detected in the nucleus by which mechanism is unknown. To this end, we have examined the distribution of Cten in various cell lines, determined primary sequence requirements for its nuclear and focal adhesion localizations, and analyzed potential roles of nuclear Cten. Our results show that a proportion of Cten translocates to nuclei in cancer cell lines and that nuclear exporting of Cten is a CRM1-dependent process. A nuclear localization sequence and a nuclear export sequence are identified within Cten. In addition, like other tensins, Cten contains two independent focal adhesion binding sites. Although further expression of recombinant Cten showed no effect on cancer cell proliferation, silencing of Cten significantly reduced cell growth. Furthermore, expression of Cten mutants either with defective nuclear export sequence or tagged with SV40 nuclear localization sequence promoted cell growth. These results suggest that nuclear Cten contributes to cancer cell proliferation. Our findings identify a molecular mechanism for regulating Cten protein trafficking in mammalian cells and provide new insights into the dynamics of focal adhesion complexes in health and disease.


Subject(s)
Tensins/genetics , Tensins/metabolism , Amino Acid Sequence , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement , Cell Proliferation/physiology , Focal Adhesions/genetics , Focal Adhesions/physiology , GTPase-Activating Proteins/metabolism , Humans , Intracellular Space/metabolism , Karyopherins/physiology , Nuclear Export Signals , Nuclear Localization Signals , Protein Binding , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction , Tumor Suppressor Proteins/metabolism , Exportin 1 Protein
8.
Cancer Lett ; 398: 46-51, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28408355

ABSTRACT

DLC1 is a RhoGAP-containing tumor suppressor that inhibits angiogenesis by repressing VEGF production in epithelial cells. Here we report the roles of DLC1 in endothelial cells. Silencing of DLC1 (siDLC1) enhances cell migration but reduces tube formation activities of human umbilical vein endothelial cells (HUVECs). Biochemically, RhoA activity and paxillin protein level are markedly increased in siDLC1 HUVECs. Although further silencing of RhoA restores the cell migration phenotype, the tube formation defect and up-regulated paxillin level remain unchanged. On the other hand, paxillin knockdown rescues tube formation and migration phenotypes but not the up-regulated RhoA activity. These results indicate that DLC1 regulates endothelial cell migration through RhoA and paxillin independently and controls tube formation mainly via paxillin. To further determine endothelial DLC1's function, we have generated endothelial specific knockout mice (DLC1-Tek). DLC1-Tek mice appear to be normal and healthy but their angiogenesis processes are compromised as shown in gel plug and aortic ring sprouting assays. Analysis of endothelial cells isolated from DLC1-Tek mice has further affirmed the cellular and biochemical phenotypes established in siDLC1 HUVECs. Our studies have demonstrated a positive regulatory role of endothelial DLC1 in angiogenesis.


Subject(s)
Endothelial Cells/metabolism , GTPase-Activating Proteins/metabolism , Neovascularization, Physiologic , Tumor Suppressor Proteins/metabolism , Animals , Cell Movement , Cells, Cultured , Down-Regulation , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , Genotype , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Knockout , Paxillin/metabolism , Phenotype , RNA Interference , Signal Transduction , Transfection , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
9.
Oncotarget ; 7(25): 38143-38153, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27203214

ABSTRACT

Tensin family members, including tensin2 (TNS2), are present as major components of the focal adhesions. The N-terminal end of TNS2 contains a C1 region (protein kinase C conserved region 1) that is not found in other tensin members. Three isoforms of TNS2 have been identified with previous reports describing the shortest V3 isoform as lacking the C1 region. Although TNS2 is known to regulate cell proliferation and migration, its role in tumorigenicity is controversial. By gain-of-function overexpression approaches, results supporting either promotion or reduction of cancer cell tumorigenicity were reported. Here we report that the complete V3 isoform also contains the C1 region and describe the expression patterns of the three human TNS2 isoforms. By loss-of-function approaches, we show that silencing of TNS2 up-regulates the activities of Akt, Mek, and IRS1, and increases tumorigenicities in A549 and Hela cells. Using public database analyses we found that TNS2 is down-regulated in head and neck, esophageal, breast, lung, liver, and colon cancer. In addition, patients with low TNS2 expression showed poor relapse-free survival rates for breast and lung cancers. These results strongly suggest a role of tensin2 in suppressing cell transformation and reduction of tumorigenicity.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Tensins/metabolism , A549 Cells , Animals , Carcinogenesis , Cell Line, Tumor , Cell Proliferation/physiology , Down-Regulation , Focal Adhesions , HeLa Cells , Heterografts , Humans , Mice , Neoplasms/genetics , Transfection
10.
Biochim Biophys Acta ; 1853(12): 3258-65, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26427649

ABSTRACT

DLC1 is a RhoGAP-containing tumor suppressor and many of DLC1's functions are absolutely dependent on its RhoGAP activity. Through its RhoGAP domain, DLC1 inhibits the activity of RhoA GTPase, which regulates actin cytoskeleton networks and dis/assembly of focal adhesions. Tensin1 (TNS1) is a focal adhesion molecule that links the actin cytoskeleton to integrins and forms signaling complexes through its multiple binding domains. Here, we report that TNS1 enhances RhoA activity in a DLC1-dependent manner. This is accomplished by binding to DLC1 through TNS1's C2, SH2, and PTB domains. Point mutations at these three sites disrupt TNS1's interaction with DLC1 as well as its effect on RhoA activity. The biological relevance of this TNS1-DLC1-RhoA signaling axis is investigated in TNS1 knockout (KO) cells and mice. Endothelial cells isolated from TNS1 KO mice or those silenced with TNS1 siRNA show significant reduction in proliferation, migration, and tube formation activities. Concomitantly, the RhoA activity is down-regulated in TNS1 KO cells and this reduction is restored by further silencing of DLC1. Furthermore, the angiogenic process is compromised in TNS1 KO mice. These studies demonstrate that TNS1 binds to DLC1 and fine-tunes its RhoGAP activity toward RhoA and that the TNS1-DLC1-RhoA signaling axis is critical in regulating cellular functions that lead to angiogenesis.


Subject(s)
GTPase-Activating Proteins/metabolism , Microfilament Proteins/physiology , Tumor Suppressor Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Amino Acid Sequence , Animals , Binding Sites , Human Umbilical Vein Endothelial Cells , Mice , Mice, Knockout , Microfilament Proteins/chemistry , Molecular Sequence Data , Protein Binding , Sequence Homology, Amino Acid , Tensins
11.
Mol Carcinog ; 53(10): 787-92, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23625726

ABSTRACT

Cten is a focal adhesion molecule that is expressed at very low levels in most normal tissues. Nonetheless, its expression has been found to increase dramatically in many types of cancer including colorectal, breast, gastric, and pancreatic cancer, suggesting that cten may play a critical role during tumorigenesis. To study the mechanisms that induce cten expression and the function of up-regulated cten, we examined the effects of several cancer-associated growth factors and cytokines on cten expression. We found that EGF, FGF2, NGF, PDGF, TGF-ß, IGF-1, IL-6, and IL-13 were able to induce cten expression in a dose- and time-dependent manner. The Mek-Erk and PI3K-Akt pathways were two main signaling cascades responsible for cten up-regulation, whereas the Jak-Stat pathway could contribute to the increase in some conditions. Since many of these factors are known to promote cell migration, we hypothesized that up-regulated cten might contribute to this process. This hypothesis was investigated in FGF2-mediated cell migration. Silencing of cten not only reduced regular cell motility but also FGF2-mediated cell migration. Overexpression of cten promoted cell migration and FGF2 treatment failed to further enhance cell migration. Our findings that (1) cten is a common downstream molecule of these cancer-associated growth factors and cytokines; and that (2) up-regulated cten modulates cell migration induced by FGF2 and likely other growth factors as well, strongly suggest that cten could be a potential downstream therapeutic target for treating cancers associated with aberrant signaling of these growth factors and cytokines.


Subject(s)
Cell Movement , Fibroblast Growth Factor 2/physiology , Microfilament Proteins/metabolism , Cell Line, Tumor , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Microfilament Proteins/genetics , Neoplasm Invasiveness , Signal Transduction , Tensins , Up-Regulation
12.
Cancer Res ; 73(16): 5266-76, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23774213

ABSTRACT

Activation of EGF receptor (EGFR) triggers signaling pathways regulating various cellular events that contribute to tissue development and function. Aberrant activation of EGFR contributes to tumor progression as well as therapeutic resistance in patients with cancer. C-terminal tensin-like (CTEN; TNS4) is a focal adhesion molecule that is a member of the tensin family. Its expression is upregulated by EGF and elevated CTEN mediates EGF-induced cell migration. In the presence of CTEN, we found that EGF treatment elevated the level of EGFR protein but not mRNA. The extended half-life of activated EGFR sustained its signaling cascades. CTEN reduced ligand-induced EGFR degradation by binding to the E3 ubiquitin ligase c-Cbl and decreasing the ubiquitination of EGFR. The Src homology 2 domain of CTEN is not only required for binding to the phosphorylated tyrosine residue at codon 774 of c-Cbl, but is also essential for the tumorigenicity observed in the presence of CTEN. Public database analyses indicated that CTEN mRNA levels are elevated in breast, colon, lung, and pancreas cancers, but not correlated with EGFR mRNA levels in these cancers. In contrast, immunohistochemistry analyses of lung cancer specimens showed that CTEN and EGFR protein levels were positively associated, in support of our finding that CTEN regulates EGFR protein levels through a posttranslational mechanism. Overall, this work defines a function for CTEN in prolonging signaling from EGFR by reducing its ligand-induced degradation.


Subject(s)
ErbB Receptors/genetics , ErbB Receptors/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Signal Transduction/genetics , Cell Line , Cell Line, Tumor , Epidermal Growth Factor/pharmacology , HCT116 Cells , HEK293 Cells , Humans , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs/drug effects , Protein Interaction Domains and Motifs/genetics , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Proteolysis/drug effects , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , RNA, Messenger/genetics , Signal Transduction/drug effects , Tensins , Tyrosine/genetics , Tyrosine/metabolism , Ubiquitination/drug effects , Ubiquitination/genetics , src Homology Domains/genetics
13.
Gene ; 520(2): 90-7, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23500447

ABSTRACT

Cten is a focal adhesion molecule and a member of the tensin family. Its expression is highly enriched in the prostate and placenta, suggesting that cten gene might be closely associated with mammalian species. Recent studies have reported that cten expression is frequently up-regulated in a variety of cancers and its levels appear to correlate with tumorigenicity. Here, we have (1) analyzed cten sequences of various species to build a phylogenetic tree, (2) examined cten mRNA levels in human and mouse tissues to establish its expression profiles, and (3) determined the promoter region of human CTEN gene in cell lines and in a mouse model to understand its transcriptional regulation. Our analyses indicate that all currently known cten genes are present in mammals. The prostate and placenta are the two most cten abundant tissues in human and mouse, meanwhile brain and lung also express low levels of cten. Results from cell culture reporter assays demonstrate that a 327-bp fragment is the shortest functional promoter. All functional promoter constructs produce 40- to 160-fold increases in luciferase reporter activities in normal prostate cells, whereas lower activities (<40-fold) are detected in non-prostatic cell lines. To evaluate CTEN promoter activity in mice and develop a new tissue specific Cre recombinase mouse model, we have established pCTEN-Cre:R26R mice by crossing R26R ß-galactosidase reporter mice with pCTEN-Cre transgenic mice, in which the 327-bp cten promoter drives the expression of Cre recombinase. X-gal analysis has shown strong ß-galactosidase activities in the prostate, brain, and few other tissues in pCTEN-Cre:R26R mice. Altogether, we have identified the promoter region of human cten gene and provided a useful tool for investigating cell lineages and generating tissue-specific knockout or knockin mice.


Subject(s)
Microfilament Proteins/genetics , Phylogeny , Animals , Base Sequence , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Microfilament Proteins/metabolism , Molecular Sequence Data , NIH 3T3 Cells , Pregnancy , Tensins , Transcription, Genetic
14.
Mol Cancer Res ; 10(1): 34-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22064653

ABSTRACT

Deleted in liver cancer 1 (DLC1) is a GTPase-activating protein (GAP) domain containing tumor suppressor that localizes to focal adhesions. In cancer cells, loss of DLC1 is known to enhance cancer cell migration. However, the role of DLC1 in normal cell migration has not been well studied. Here, we show that silencing of DLC1 (shDLC1) in normal prostate epithelial cells reduces cell migration in both Transwell and wound-healing assays. This migration defect is mainly due to upregulation of plasminogen activator inhibitor 1 (PAI-1). Silencing of PAI-1 rescues the shDLC1-reduced migration phenotype. Reexpression of DLC1 suppresses PAI-1 and restores the migration defect as well. In contrast, DLC1-K714E (GAP inactive) mutant neither decreases the PAI-1 level nor rescues the shDLC1 migration defect. Interestingly, DLC1-Y442F (tensin-binding and focal adhesion-localizing defective) mutant is able to suppress PAI-1 expression but does not restore the migration defect. Furthermore, PAI-1 upregulation in shDLC1 cells is EGFR-MEK pathway dependent and is able to promote in vitro angiogenesis. Together, our results show that at least the following two new mechanisms are involved in DLC1-mediated normal cell migration: (i) DLC1 modulates the expression of PAI-1, which is a negative regulator for cell migration, in a GAP domain and EGFR-MEK-dependent manner and (ii) Independent of PAI-1, the interaction of DLC1 with tensin members positively regulates cell migration.


Subject(s)
Cell Movement/genetics , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , Plasminogen Activator Inhibitor 1/genetics , Prostate/drug effects , RNA, Small Interfering/pharmacology , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Cell Movement/drug effects , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/physiology , ErbB Receptors/metabolism , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/physiology , Gene Expression/drug effects , Gene Expression/physiology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Plasminogen Activator Inhibitor 1/metabolism , Prostate/cytology , Prostate/metabolism , Prostate/physiology , RNA Interference/physiology , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology , Up-Regulation/genetics
15.
Cancer Res ; 70(21): 8270-5, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20861185

ABSTRACT

The Rho GTPase-activating protein DLC1 is a tumor suppressor that is often deleted in liver cancer and downregulated in other cancers. DLC1 regulates the actin cytoskeleton, cell shape, adhesion, migration, and proliferation through its Rho GTPase-activating protein activity and focal adhesion localization. In this study, we silenced DLC1 in nonmalignant prostate epithelial cells to explore its tumor suppression functions. Small hairpin RNA-mediated silencing of DLC1 was insufficient to promote more aggressive phenotypes associated with tumor cell growth. In contrast, DLC1 silencing promoted pro-angiogenic responses through vascular endothelial growth factor (VEGF) upregulation, accompanied by the accumulation of hypoxia-inducible factor 1α and its nuclear localization. Notably, modulation of VEGF expression by DLC1 was dependent on epidermal growth factor receptor-MAP/ERK kinase-hypoxia-inducible factor 1 signaling but on RhoA pathways. Clinically, VEGF upregulation is a highly significant event in prostate cancers in which DLC1 is downregulated. Thus, our results strongly suggest that loss of DLC1 may serve as a "second hit" in promoting angiogenesis in a paracrine fashion during tumorigenesis.


Subject(s)
GTPase-Activating Proteins/physiology , Neovascularization, Pathologic/metabolism , Paracrine Communication , Prostatic Neoplasms/metabolism , Tumor Suppressor Proteins/physiology , Animals , Aorta/cytology , Aorta/metabolism , Blotting, Western , Cell Adhesion , Cell Movement , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tissue Array Analysis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
16.
Cancer Res ; 69(11): 4563-6, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487278

ABSTRACT

C-terminal tensin-like (cten) is a focal adhesion molecule belonging to the tensin family. Previous studies have suggested that cten may function as a prostate-specific tumor suppressor. Here, we show that although cten is expressed at a very low level in normal colon, its expression is significantly up-regulated in colon cancer. Furthermore, a high population of cten is found in the nucleus, where it interacts with beta-catenin, a critical player in the canonical Wnt pathway. This interaction may contribute to the role of cten in enhancing the colony formation, anchorage-independent growth, and invasiveness of colon cancer cells. Our studies have identified cten as a novel nuclear partner of beta-catenin, showed an oncogenic activity of cten in colon cancers, and revealed cten as a potential biomarker and target for colon cancers.


Subject(s)
Colonic Neoplasms/pathology , Microfilament Proteins/physiology , beta Catenin/physiology , Cell Nucleus/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Disease Progression , HCT116 Cells , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neoplasm Invasiveness , Protein Binding , Tensins , Tumor Cells, Cultured , Tumor Stem Cell Assay , Up-Regulation/physiology , beta Catenin/genetics , beta Catenin/metabolism
17.
Gene ; 443(1-2): 151-7, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19439180

ABSTRACT

Glycine N-methyltransferase (GNMT) is a tumor susceptibility gene for both hepatocellular carcinoma and prostate cancer. We have previously characterized GNMT genomic structure and mapped its chromosomal localization to 6p12. For this study we identified a GNMT transcriptional start site at the 14th position upstream of the ATG codon. Electrophoretic mobility shift assay results indicate binding of the nuclear factor-Y (NF-Y) transcription factor to the CCAAT box (-71/-67) of the GNMT gene. Mutation assay results suggest that the nucleotide sequence in the -56/-47 region is a binding site for a putative transcriptional factor. The TATA-less core promoter (-133/+14) contains three major elements: an Sp1 site, CCAAT box, and a novel box within the CTGTCGGCTG sequence. One functional xenobiotic response element (XRE) located at the -104/-82 region is inducable by benzo[a]pyrene treatment. We believe our results have value for the study of GNMT transcriptional regulation.


Subject(s)
Glycine N-Methyltransferase/genetics , Regulatory Sequences, Nucleic Acid , Base Sequence , CCAAT-Binding Factor/metabolism , Electrophoretic Mobility Shift Assay , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Transcription Initiation Site
18.
J Phys Chem B ; 113(18): 6511-9, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19366241

ABSTRACT

Cell tracing dyes are very frequently utilized in cellular biology research because they provide highly sensitive fluorescent tags that do not compromise cellular functions such as growth and proliferation. In many investigations concerning cellular adhesion and mechanics, fluorescent dyes have been employed with the assumption of little impact on the results. Using the single cell compression technique developed by our team, the single cell mechanics of MDA-MB-468 and MLC-SV40 cells were investigated as a function of dye uptake. Cell tracing dyes increase living cell stiffness 3-6 times and cell-to-probe adhesion up to 7 times. These results suggest a more significant effect than toxins, such as thrombin. A simple analytical model was derived to enable the extraction of the Young's moduli of the cell membrane and cytoskeleton from the force-deformation profiles measured for individual cells. The increase in Young's modulus of the membrane is 3-7 times, which is more significant than that of the cytoskeleton (1.1-3.4 times). We propose that changes in cell mechanics upon the addition of fluorescent tracing dye are primarily due to the incorporation of amphiphilic dye molecules into the cellular plasma membrane, which increases the lateral interaction among phospholipid chains and thus enhances their rigidity and adhesion.


Subject(s)
Fluoresceins/chemistry , Fluorescent Dyes/metabolism , Succinimides/chemistry , Cell Adhesion , Cell Line, Tumor , Humans
19.
Cancer Res ; 68(19): 7718-22, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829524

ABSTRACT

Deleted in liver cancer-1 (DLC-1) is a RhoGTPase-activating protein (RhoGAP) domain containing tumor suppressor that is often down-regulated in various cancer types. Previously, we have shown that DLC-1 is recruited to focal adhesions by binding to the Src homology 2 domains of tensins and the focal adhesion localization is critical for the tumor suppression activity of DLC-1. To investigate whether mutations in the focal adhesion targeting (FAT) region might occur and attenuate the expression, localization, and function of DLC-1, we have first mapped the FAT region to the amino acid residues from 201 to 500, and then sequenced cDNAs and genomic DNAs encoding the FAT region from cancer patients. Several missense and nonsense mutations were detected. All missense mutations were further examined for the potential effect on the function of DLC-1. Although these mutations did not seem to affect the focal adhesion localization of DLC-1, the activities of suppressing tumor cell growth were impaired in two mutants: T301K and S308I. Consistent with the fact that the RhoGAP activity of DLC-1 is essential for inhibiting tumor cell growth, the RhoGAP activities were significantly reduced in these mutants, suggesting that the FAT region also contains a regulatory element for its COOH-terminal RhoGAP domain. Our studies have shown that mutations in DLC-1 may lead to loss of function and contribute to the tumorigenesis, and have revealed an allosteric regulation site for its RhoGAP activity.


Subject(s)
Focal Adhesions/metabolism , Mutation/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Mutational Analysis , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Protein Transport/genetics , Tissue Distribution , Tumor Suppressor Proteins/metabolism
20.
Biochem Biophys Res Commun ; 358(3): 716-21, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17506989

ABSTRACT

Three peptides, D1 (amino acid residues 175-201), D2 (a.a. 434-467), and TM (a.a. 1128-1159), corresponding to the spike protein (S) of severe acute respiratory syndrome corona virus (SARS CoV) were synthesized and their immunological functions were investigated in three different animals models (mice, guinea pigs, and rabbits). The peptides mixture formulated either with Freund's adjuvant or synthetic adjuvant Montanide ISA-51/oligodeoxy nucleotide CpG (ISA/CpG) could elicit antisera in immunized animals which were capable of inhibiting SARS/HIV pseudovirus entry into HepG2 cells. The neutralizing epitopes were identified using peptides to block the neutralizing effect of guinea pig antisera. The major neutralizing epitope was located on the D2 peptide, and the amino acid residue was fine mapped to 434-453. In BALB/c mice T-cell proliferation assay revealed that only D2 peptide contained T-cell epitope, the sequence of which corresponded to amino acid residue 434-448. The ISA/CpG formulation generated anti-D2 IgG titer comparable to those obtained from Freund's adjuvant formulation, but generated fewer antibodies against D1 or TM peptides. The highly immunogenic D2 peptide contains both neutralizing and Th cell epitopes. These results suggest that synthetic peptide D2 would be useful as a component of SARS vaccine candidates.


Subject(s)
Severe Acute Respiratory Syndrome/prevention & control , Vaccines, Synthetic/chemistry , Animals , Drug Design , Epitopes/chemistry , Freund's Adjuvant , Guinea Pigs , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Neutralization Tests , Peptides/chemistry , Rabbits , Severe Acute Respiratory Syndrome/metabolism , T-Lymphocytes/metabolism , Th1 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...