Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Int J Cancer ; 154(10): 1814-1827, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38282121

ABSTRACT

Adenoid cystic carcinoma (ACC) and basal cell adenoma (BCA) share many histological characteristics and often need a differential diagnosis in clinical pathology. Recently, we found homeobox protein engrailed-1 (EN1) was a potential diagnostic marker for ACC in an organoids library of salivary gland tumors (SGTs). Here we aim to confirm EN1 as a differential diagnostic marker for ACC, and further investigate the regulatory mechanism and biological function of EN1 in tumor progression. The transcriptional analysis, quantitative polymerase chain reaction, Western blot and immunohistochemistry staining were performed and revealed that EN1 was specifically and highly expressed in ACC, and accurately differentiated ACC from BCA. Furthermore, TGFß signaling pathway was found associated with ACC, and the regulation of EN1 through TGFß was detected in the human ACC cell lines and patient-derived organoids (PDOs). TGFß-induced EN1 was important in promoting tumor budding in the PDOs model. Interestingly, a high level of EN1 and TGFß1 in the budding tips was observed in ACC clinical samples, and the expression of EN1 and TGFß1 in ACC was significantly associated with the clinical stage. In summary, our study verified EN1 is a good diagnostic marker to differentiate ACC from BCA. TGFß-induced EN1 facilitates the tumor budding of ACC, which might be an important mechanism related to the malignant phenotype of ACC.


Subject(s)
Adenoma , Carcinoma, Adenoid Cystic , Salivary Gland Neoplasms , Humans , Adenoma/pathology , Biomarkers, Tumor/genetics , Carcinoma, Adenoid Cystic/pathology , Homeodomain Proteins , Salivary Gland Neoplasms/pathology , Transforming Growth Factor beta
2.
J Exp Clin Cancer Res ; 41(1): 350, 2022 Dec 17.
Article in English | MEDLINE | ID: mdl-36527158

ABSTRACT

BACKGROUND: Salivary gland tumors (SGTs) include a large group of rare neoplasms in the head and neck region, and the heterogeneous and overlapping features among the subtypes frequently make diagnostic difficulties. There is an urgent need to understand the cellular mechanisms underlying the heterogeneity and overlap among the subtypes, and explore the subtype-specific diagnostic biomarkers. METHODS: The tumor tissue and the adjacent normal tissue from the 6 most common types of SGTs were processed for organoid culture which only maintained tumor epithelial cells. Organoids were histologically evaluated based on phenotype markers, followed by transcriptional profiling using RNA-sequencing. The transcriptomic similarities and differences among the subtypes were analyzed by subtype consensus clustering and hierarchical clustering. Furthermore, by comparative transcriptional analysis for these 6 types of SGTs and the matched organoids, the potential diagnostic biomarkers from tumor epithelium were identified, in which two selected biomarkers were evaluated by qPCR and confirmed by immunohistochemistry staining using a tissue microarray. RESULTS: We generated a biobank of patient-derived organoids (PDOs) with 6 subtypes of SGTs, including 21 benign and 24 malignant SGTs. The PDOs recapitulated the morphological and transcriptional characteristics of the parental tumors. The overlap in the cell types and the heterogenous growth patterns were observed in the different subtypes of organoids. Comparing the bulk tissues, the cluster analysis of the PDOs remarkably revealed the epithelial characteristics, and visualized the intrinsic relationship among these subtypes. Finally, the exclusive biomarkers for the 6 most common types of SGTs were uncovered by comparative analysis, and PTP4A1 was demonstrated as a useful diagnostic biomarker for mucoepidermoid carcinoma. CONCLUSIONS: We established the first organoid biobank with multiple subtypes of SGTs. PDOs of SGTs recapitulate the morphological and transcriptional characteristics of the original tumors, which uncovers subtype-specific biomarkers and reveals the molecular distance among the subtype of SGTs.


Subject(s)
Salivary Gland Neoplasms , Humans , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/diagnosis , Salivary Gland Neoplasms/pathology , Immunohistochemistry , Biomarkers , Organoids/metabolism
3.
EBioMedicine ; 45: 192-207, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31262711

ABSTRACT

BACKGROUND: Tumour necrosis factor receptor associated factor 6 (TRAF6) promotes inflammation in response to various cytokines. Aberrant Wnt3a signals promotes cancer progression through accumulation of ß-Catenin. Here we investigated a potential role for TRAF6 in Wnt signaling. METHODS: TRAF6 expression was silenced by siRNA in human prostate cancer (PC3U) and human colorectal SW480 cells and by CRISPR/Cas9 in zebrafish. Several biochemical methods and analyses of mutant phenotype in zebrafish were used to analyse the function of TRAF6 in Wnt signaling. FINDINGS: Wnt3a-treatment promoted binding of TRAF6 to the Wnt co-receptors LRP5/LRP6 in PC3U and LNCaP cells in vitro. TRAF6 positively regulated mRNA expression of ß-Catenin and subsequent activation of Wnt target genes in PC3U cells. Wnt3a-induced invasion of PC3U and SW480 cells were significantly reduced when TRAF6 was silenced by siRNA. Database analysis revealed a correlation between TRAF6 mRNA and Wnt target genes in patients with prostate cancer, and high expression of LRP5, TRAF6 and c-Myc correlated with poor prognosis. By using CRISPR/Cas9 to silence TRAF6 in zebrafish, we confirm TRAF6 as a key molecule in Wnt3a signaling for expression of Wnt target genes. INTERPRETATION: We identify TRAF6 as an important component in Wnt3a signaling to promote activation of Wnt target genes, a finding important for understanding mechanisms driving prostate cancer progression. FUND: KAW 2012.0090, CAN 2017/544, Swedish Medical Research Council (2016-02513), Prostatacancerförbundet, Konung Gustaf V:s Frimurarestiftelse and Cancerforskningsfonden Norrland. The funders did not play a role in manuscript design, data collection, data analysis, interpretation nor writing of the manuscript.


Subject(s)
Inflammation/genetics , Prostatic Neoplasms/genetics , TNF Receptor-Associated Factor 6/genetics , Wnt3A Protein/genetics , Zebrafish Proteins/genetics , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Inflammation/pathology , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Male , Prostatic Neoplasms/pathology , Wnt Signaling Pathway/genetics , Zebrafish , beta Catenin/genetics
4.
Oncogene ; 38(22): 4215-4231, 2019 05.
Article in English | MEDLINE | ID: mdl-30705401

ABSTRACT

Prostate cancer disseminates primarily into the adjacent lymph nodes, which is related to a poor outcome. Atypical protein kinase C ζ (PKCζ) is highly expressed in aggressive prostate cancer and correlates with Gleason score, clinical stage, and poor prognosis. Here, we report the molecular mechanisms of PKCζ in lymphatic metastasis during prostate cancer progression. Using zinc-finger nuclease technology or PKCζ shRNA lentiviral particles, and orthotopic mouse xenografts, we show that PKCζ-knockout or knockdown from aggressive prostate cancer (PC3 and PC3U) cells, decreasesd tumor growth and lymphatic metastasis in vivo. Intriguingly, PKCζ-knockout or knockdown impaired the activation of AKT, ERK, and NF-κB signaling in prostate cancer cells, thereby impairing the expression of lymphangiogenic factors and macrophage recruitment, resulting in aberrant lymphangiogenesis. Moreover, PKCζ regulated the expression of hyaluronan synthase enzymes, which is important for hyaluronan-mediated lymphatic drainage and tumor dissemination. Thus, PKCζ plays a crucial oncogenic role in the lymphatic metastasis of prostate cancer and is predicted to be a novel therapeutic target for prostate cancer.


Subject(s)
Lymphatic Metastasis/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase C/metabolism , A549 Cells , Animals , Cell Line, Tumor , Humans , Hyaluronan Synthases/metabolism , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphangiogenesis/physiology , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , MAP Kinase Signaling System/physiology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Nude , NF-kappa B/metabolism , PC-3 Cells , Prostate/metabolism , Prostate/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
5.
J Mol Endocrinol ; 56(1): R21-31, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26489764

ABSTRACT

This review will describe the SH2-domain signaling protein Src Homology-2 domain containing protein B (SHB) and its role in various physiological processes relating in particular to glucose homeostasis and ß cell function. SHB operates downstream of several tyrosine kinase receptors and assembles signaling complexes in response to receptor activation by interacting with other signaling proteins via its other domains (proline-rich, phosphotyrosine-binding and tyrosine-phosphorylation sites). The subsequent responses are context-dependent. Absence of Shb in mice has been found to exert effects on hematopoiesis, angiogenesis and glucose metabolism. Specifically, first-phase insulin secretion in response to glucose was impaired and this effect was related to altered characteristics of focal adhesion kinase activation modulating signaling through Akt, ERK, ß catenin and cAMP. It is believed that SHB plays a role in integrating adaptive responses to various stimuli by simultaneously modulating cellular responses in different cell-types, thus playing a role in maintaining physiological homeostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Insulin-Secreting Cells/metabolism , Proto-Oncogene Proteins/physiology , Amino Acid Sequence , Animals , Conserved Sequence , Diabetes Mellitus, Type 2/metabolism , Focal Adhesion Kinase 1/metabolism , Humans , Molecular Sequence Data
6.
J Cell Biol ; 210(2): 319-32, 2015 Jul 20.
Article in English | MEDLINE | ID: mdl-26169354

ABSTRACT

Members of the transforming growth factor ß (TGFß) family initiate cellular responses by binding to TGFß receptor type II (TßRII) and type I (TßRI) serine/threonine kinases, whereby Smad2 and Smad3 are phosphorylated and activated, promoting their association with Smad4. We report here that TßRI interacts with the SH3 domains of the adaptor protein CIN85 in response to TGFß stimulation in a TRAF6-dependent manner. Small interfering RNA-mediated knockdown of CIN85 resulted in accumulation of TßRI in intracellular compartments and diminished TGFß-stimulated Smad2 phosphorylation. Overexpression of CIN85 instead increased the amount of TßRI at the cell surface. This effect was inhibited by a dominant-negative mutant of Rab11, suggesting that CIN85 promoted recycling of TGFß receptors. CIN85 enhanced TGFß-stimulated Smad2 phosphorylation, transcriptional responses, and cell migration. CIN85 expression correlated with the degree of malignancy of prostate cancers. Collectively, our results reveal that CIN85 promotes recycling of TGFß receptors and thereby positively regulates TGFß signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , Animals , COS Cells , Cell Line, Tumor , Cell Membrane/metabolism , Chlorocebus aethiops , HEK293 Cells , Humans , Male , Molecular Sequence Data , Prostatic Neoplasms/metabolism , Protein Binding , Protein Interaction Mapping , Protein Transport , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Transcription, Genetic , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
7.
BMC Cancer ; 15: 234, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25885274

ABSTRACT

BACKGROUND: Shb is a signaling protein downstream of vascular endothelial growth factor receptor-2 and Shb deficiency has been found to restrict tumor angiogenesis. The present study was performed in order to assess metastasis in Shb deficiency using B16F10 melanoma cells. METHODS: B16F10 melanoma cells were inoculated subcutaneously on wild type or Shb +/- mice. Primary tumors were resected and lung metastasis determined after tumor relapse. Lung metastasis was also assessed after bone marrow transplantation of wild type bone marrow to Shb +/- recipients and Shb +/- bone marrow to wild type recipients. Primary tumors were subject to immunofluorescence staining for CD31, VE-cadherin, desmin and CD8, RNA isolation and isolation of vascular fragments for further RNA isolation. RNA was used for real-time RT-PCR and microarray analysis. RESULTS: Numbers of lung metastases were increased in Shb +/- or -/- mice and this coincided with reduced pericyte coverage and increased vascular permeability. Gene expression profiling of vascular fragments isolated from primary tumors and total tumor RNA revealed decreased expression of different markers for cytotoxic T cells in tumors grown on Shb +/- mice, suggesting that vascular aberrations caused altered immune responses. CONCLUSIONS: It is concluded that a unique combinatorial response of increased vascular permeability and reduced recruitment of cytotoxic CD8+ cells occurs as a consequence of Shb deficiency in B16F10 melanomas. These changes may promote tumor cell intravasation and metastasis.


Subject(s)
Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins/genetics , Animals , Bone Marrow Transplantation , Capillary Permeability/genetics , Disease Models, Animal , Gene Expression , Genotype , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Mice , Mice, Knockout , Neoplasm Metastasis , Neovascularization, Pathologic/metabolism , Pericytes/metabolism , Proto-Oncogene Proteins/deficiency , Tumor Burden
8.
Ups J Med Sci ; 120(3): 169-80, 2015.
Article in English | MEDLINE | ID: mdl-25854824

ABSTRACT

BACKGROUND: Pancreatic or islet fibrosis is often associated with activated pancreatic stellate cells (PSCs). PSCs are considered not only to promote fibrosis, but also to be associated with glucose intolerance in some diseases. We therefore evaluated morphological and functional relationships between islets and PSCs in the normal mouse pancreas and transplanted islets. METHODS: Immunohistochemistry was used to map the presence of PSCs in the normal mouse pancreas and islets implanted under the renal capsule. We isolated and cultured mouse PSCs and characterized them morphologically by immunofluorescence staining. Furthermore, we measured their cytokine production and determined their effects on insulin release from simultaneously cultured islets. RESULTS: PSCs were scattered throughout the pancreas, with occasional cells within the islets, particularly in the islet capsule. In islet transplants they were found mainly in the graft periphery. Cultured PSCs became functionally activated and produced several cytokines. Throughout the culture period they linearly increased their production of interleukin-6 and mammalian keratinocyte-derived chemokine. PSC cytokine production was not affected by acute hyperglycemia. Syngeneic islets co-cultured with PSCs for 24-48 h increased their insulin release and lowered their insulin content. However, short-term insulin release in batch-type incubations was unaffected after 48 h of co-culture. Increased islet cell caspase-3 activation and a decreased islet cell replication were consistently observed after co-culture for 2 or 7 days. CONCLUSION: Activated PSCs may contribute to impaired islet endocrine function seen in exocrine pancreatitis and in islet fibrosis associated with some cases of type 2 diabetes.


Subject(s)
Islets of Langerhans/physiopathology , Pancreatic Stellate Cells/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Hyperglycemia/metabolism , Immunohistochemistry , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
9.
Sci Signal ; 7(307): ra2, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24399296

ABSTRACT

Transforming growth factor-ß (TGFß) can be both a tumor promoter and suppressor, although the mechanisms behind the protumorigenic switch remain to be fully elucidated. The TGFß type I receptor (TßRI) is proteolytically cleaved in the ectodomain region. Cleavage requires the combined activities of tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) and TNF-α-converting enzyme (TACE). The cleavage event occurs selectively in cancer cells and generates an intracellular domain (ICD) of TßRI, which enters the nucleus to mediate gene transcription. Presenilin 1 (PS1), a γ-secretase catalytic core component, mediates intramembrane proteolysis of transmembrane receptors, such as Notch. We showed that TGFß increased both the abundance and activity of PS1. TRAF6 recruited PS1 to the TßRI complex and promoted lysine-63-linked polyubiquitination of PS1, which activated PS1. Furthermore, PS1 cleaved TßRI in the transmembrane domain between valine-129 and isoleucine-130, and ICD generation was inhibited when these residues were mutated to alanine. We also showed that, after entering the nucleus, TßRI-ICD bound to the promoter and increased the transcription of the gene encoding TßRI. The TRAF6- and PS1-induced intramembrane proteolysis of TßRI promoted TGFß-induced invasion of various cancer cells in vitro. Furthermore, when a mouse xenograft model of prostate cancer was treated with the γ-secretase inhibitor DBZ {(2S)-2-[2-(3,5-difluorophenyl)-acetylamino]-N-(5-methyl-6-oxo-6,7-dihydro-5H-dibenzo[b,d]azepin-7-yl)-propionamide}, generation of TßRI-ICD was prevented, transcription of the gene encoding the proinvasive transcription factor Snail1 was reduced, and tumor growth was inhibited. These results suggest that γ-secretase inhibitors may be useful for treating aggressive prostate cancer.


Subject(s)
Presenilin-1/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , TNF Receptor-Associated Factor 6/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cells, Cultured , Dibenzazepines/pharmacology , Embryo, Mammalian/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , HEK293 Cells , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Polyubiquitin/metabolism , Presenilin-1/genetics , Protein Binding/drug effects , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , TNF Receptor-Associated Factor 6/genetics , Transforming Growth Factor beta1/pharmacology , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
10.
Cell Signal ; 25(1): 85-92, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23000345

ABSTRACT

Vascular permeability is a hallmark response to the main angiogenic factor VEGF-A and we have previously described a reduction of this response in Shb knockout mice. To characterize the molecular mechanisms responsible for this effect, endothelial cells were isolated from lungs and analyzed in vitro. Shb deficient endothelial cells exhibited less migration in a scratch wound-healing assay both under basal conditions and after vascular endothelial growth factor-A (VEGF-A) stimulation, suggesting a functional impairment of these cells in vitro. Staining for VE-cadherin and vascular endothelial growth factor receptor-2 (VEGFR-2) showed co-localization in adherens junctions and in intracellular sites such as the perinuclear region in wild-type and Shb knockout cells. VEGF-A decreased the VE-cadherin/VEGFR-2 co-localization in membrane structures resembling adherens junctions in wild-type cells whereas no such response was noted in the Shb knockout cells. VE-cadherin/VEGFR-2 co-localization was also recorded using spinning-disk confocal microscopy and VEGF-A caused a reduced association in the wild-type cells whereas the opposite pattern was observed in the Shb knockout cells. The latter expressed slightly more of cell surface VEGFR-2. VEGF-A stimulated extracellular-signal regulated kinase, Akt and Rac1 activities in the wild-type cells whereas no such responses were noted in the knockout cells. We conclude that aberrant signaling characteristics with respect to ERK, Akt and Rac1 are likely explanations for the observed altered pattern of VE-cadherin/VEGFR-2 association. The latter is important for understanding the reduced in vivo vascular permeability response in Shb knockout mice, a phenomenon that has patho-physiological relevance.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cell Movement/drug effects , Endothelial Cells/metabolism , Proto-Oncogene Proteins/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Endothelial Cells/cytology , Lung/cytology , Lung/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Signal Transduction
11.
J Exp Med ; 209(7): 1363-77, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22689825

ABSTRACT

Regulation of vascular endothelial (VE) growth factor (VEGF)-induced permeability is critical in physiological and pathological processes. We show that tyrosine phosphorylation of VEGF receptor 2 (VEGFR2) at Y951 facilitates binding of VEGFR2 to the Rous sarcoma (Src) homology 2-domain of T cell-specific adaptor (TSAd), which in turn regulates VEGF-induced activation of the c-Src tyrosine kinase and vascular permeability. c-Src was activated in vivo and in vitro in a VEGF/TSAd-dependent manner, and was regulated via increased phosphorylation at pY418 and reduced phosphorylation at pY527. Tsad silencing blocked VEGF-induced c-Src activation, but did not affect pathways involving phospholipase Cγ, extracellular regulated kinase, and endothelial nitric oxide. VEGF-induced rearrangement of VE-cadherin-positive junctions in endothelial cells isolated from mouse lungs, or in mouse cremaster vessels, was dependent on TSAd expression, and TSAd formed a complex with VE-cadherin, VEGFR2, and c-Src at endothelial junctions. Vessels in tsad(-/-) mice showed undisturbed flow and pressure, but impaired VEGF-induced permeability, as measured by extravasation of Evans blue, dextran, and microspheres in the skin and the trachea. Histamine-induced extravasation was not affected by TSAd deficiency. We conclude that TSAd is required for VEGF-induced, c-Src-mediated regulation of endothelial cell junctions and for vascular permeability.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Capillary Permeability/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD/metabolism , Blotting, Western , Cadherins/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extravasation of Diagnostic and Therapeutic Materials/etiology , Female , Fluorescein-5-isothiocyanate/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microspheres , Phosphorylation/drug effects , Protein Binding , RNA Interference , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology
12.
Angiogenesis ; 15(3): 469-80, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22562363

ABSTRACT

Vascular endothelial growth factor (VEGF)-A regulates angiogenesis, vascular morphology and permeability by signaling through its receptor VEGFR-2. The Shb adapter protein has previously been found to relay certain VEGFR-2 dependent signals and consequently vascular physiology and structure was assessed in Shb knockout mice. X-ray computed tomography of vessels larger than 24 µm diameter (micro-CT) after contrast injection revealed an increased frequency of 48-96 µm arterioles in the hindlimb calf muscle in Shb knockout mice. Intravital microscopy of the cremaster muscle demonstrated a less regular vasculature with fewer branch points and increased vessel tortuosity, changes that led to an increased blood flow velocity. Reduced in vivo angiogenesis was observed in Shb knockout Matrigel™ plugs. Unlike the wild-type situation, VEGF-A did not provoke a dissociation of VE-cadherin from adherens junctions in Shb knockout venules. The reduced angiogenesis and altered properties of junctions had consequences for two patho-physiological responses to arterial occlusion: vascular permeability was reduced in the Shb knockout cremaster muscle after ligation of one supplying artery and heat-induced blood flow determined by Laser-Doppler measurements was decreased in the hindlimb after ligation of the femoral artery. Consequently, the Shb knockout mouse exhibited structural and functional (angiogenesis and vascular permeability) vascular abnormalities that have implications for understanding the function of VEGF-A under physiological conditions.


Subject(s)
Adaptation, Physiological , Endothelium, Vascular/physiology , Proto-Oncogene Proteins/physiology , Animals , Capillary Permeability , Hindlimb/blood supply , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Proto-Oncogene Proteins/genetics , Tomography, X-Ray Computed , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
Mol Oncol ; 6(3): 333-46, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22336752

ABSTRACT

The Shb adapter protein is a signaling intermediate that operates downstream of vascular endothelial growth factor receptor-2 (VEGFR-2) in endothelial cells. The Shb knockout mouse displays a dysfunctional microvasculature and impaired growth of subcutaneously implanted tumor cells. We decided to investigate tumor growth and angiogenesis in the absence of Shb in an inheritable tumor model, the RIP-Tag2 mouse, which produces insulinomas in a manner highly dependent on de novo angiogenesis. We observed a reduced tumor incidence and burden in both RIP-Tag2 Shb-/- and RIP-Tag2 Shb+/- mice. This correlated with a reduced microvascular density, measured as a percentage of insulinoma area positive for CD31 staining, and altered vascular morphology. However, treatment with a VEGF-A blocking antibody was without effect on the Shb mutant tumor volume whereas it significantly inhibited tumor volume in the wild-type mice, suggesting that in mice with reduced Shb expression tumor angiogenesis was primarily sustained by VEGF-A independent pathway(s). This notion was further substantiated by gene expression analysis of angiogenic markers showing reduced VEGF-A expression in Shb-deficient tumors. Considerable heterogeneity with respect to the gene expression profiles of other angiogenic markers and the signal-transduction characteristics was observed between different tumors, suggesting that multiple "rescue" pathways could be operating. The numbers of invasive tumors or metastases were unchanged in the Shb mutant. It is concluded that the Shb mutant background reduces tumor frequency by chronically suppressing VEGF-A dependent angiogenesis. However, VEGF-A independent angiogenesis supports a significant degree of tumor expansion in Shb-deficient mice, indicating heterogeneity in the mechanisms by which tumor expansion is promoted. Interference with Shb signaling may provide novel means for future cancer therapy.


Subject(s)
Insulinoma/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Insulinoma/genetics , Mice , Mice, Mutant Strains , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , X-Ray Microtomography
14.
Diabetes ; 61(3): 665-73, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22315321

ABSTRACT

Experimental studies indicate low revascularization of intraportally transplanted islets. This study aimed to quantify, for the first time, the blood perfusion of intrahepatically transplanted islets and elucidate necessary factors for proper islet graft revascularization at this site. Yellow chameleon protein 3.0 islets expressing fluorescent protein in all cells were transplanted. Graft blood perfusion was determined by microspheres. The vascular density and relative contribution of donor blood vessels in revascularization was evaluated using islets expressing green fluorescent protein under the Tie-2 promoter. Blood perfusion of intrahepatic islets was as a mean only 5% of that of native islets at 1-month posttransplantation. However, there was a marked heterogeneity where blood perfusion was less decreased in islets transplanted without prior culture and in many cases restored in islets with disrupted integrity. Analysis of vascular density showed that distorted islets were well revascularized, whereas islets still intact at 1-month posttransplantation were almost avascular. Few donor endothelial cells were observed in the new islet vasculature. The very low blood perfusion of intraportally transplanted islets is likely to predispose for ischemia and hamper islet function. Since donor endothelial cells do not expand posttransplantation, disruption of islet integrity is necessary for revascularization to occur by recipient blood vessels.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans/blood supply , Liver/surgery , Animals , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Regional Blood Flow , Vascular Endothelial Growth Factor A/physiology
15.
Cancer Biol Ther ; 8(10): 966-72, 2009 May.
Article in English | MEDLINE | ID: mdl-19587531

ABSTRACT

Adenoid cystic carcinoma (ACC) is a common malignant tumor in salivary glands. Unfortunately, current treatment modalities which include surgery, radiation and chemotherapy have limited success rates. To develop new treatment strategies we hypothesized that a cancer-specific apoptotic ligand driven by a tumor specific promoter would specifically induce apoptosis in ACC. To test this concept, we selected tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and the human telomerase reverse transcriptase (hTERT) promoter. The latter is highly active in 85% of human cancer cells while it is mostly inactive in somatic cells. Using immunohistochemistry we confirmed that ACC samples but not normal salivary cells were positive for hTERT. Similar results were also seen in an ACC cell line, SACC-83. We then constructed first generation Ad5 vectors which used the hTERT promoter to drive TRAIL (AdTERT-TRAIL). Transduction of SACC-83, but not of control human embryo-fibrocyte lung (HEL) cells, led to apoptosis as measured by MTT assay and flow cytomerty. We used the SACC-83 cells for a subcutaneous tumor model in vivo. Intratumoral injections of AdTERT-TRAIL (5 x 109 particles/tumor) but not of AdTERT-EGFP or PBS resulted in significant (p < 0.01) reduction in tumor sizes, which demonstrated that the human TRAIL driven by tumor-specific promoter could efficiently induce apoptosis in SACC-83 cells in vitro and in vivo. These results suggest that a novel gene therapy strategy can also be used in the treatment of ACC in the near future.


Subject(s)
Apoptosis/genetics , Carcinoma, Adenoid Cystic/therapy , Salivary Gland Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/genetics , Telomerase/genetics , Adenoviridae/genetics , Carcinoma, Adenoid Cystic/genetics , Carcinoma, Adenoid Cystic/pathology , Case-Control Studies , Cell Line, Tumor , Formazans/metabolism , Genetic Therapy/methods , Genetic Vectors , Humans , Immunohistochemistry , Promoter Regions, Genetic , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology , Telomerase/metabolism , Tetrazolium Salts/metabolism , Transduction, Genetic
16.
Cancer Res ; 69(5): 2141-8, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19223532

ABSTRACT

Shb (Src homology 2 protein B) is an adapter protein downstream of the vascular endothelial growth factor receptor receptor-2 (VEGFR-2). Previous experiments have suggested a role for Shb in endothelial cell function. Recently, the Shb gene was inactivated and Shb null mice were obtained on a mixed genetic background, but not on C57Bl6 mice. The present study was performed to address endothelial function in the Shb knockout mouse and its relevance for tumor angiogenesis. Tumor growth was retarded in Shb mutant mice, and this correlated with decreased angiogenesis both in tumors and in Matrigel plugs. Shb null mice display an abnormal endothelial ultrastructure in liver sinusoids and heart capillaries with cytoplasmic extensions projecting toward the lumen. Shb null heart VE-cadherin staining was less distinct than that of control heart, exhibiting in the former case a wavy and punctuate pattern. Experiments on isolated endothelial cells suggest that these changes could partly reflect cytoskeletal abnormalities. Vascular permeability was increased in Shb null mice in heart, kidney, and skin, whereas VEGF-stimulated vascular permeability was reduced in Shb null mice. It is concluded that Shb plays an important role in maintaining a functional vasculature in adult mice, and that interference with Shb signaling may provide novel means to regulate tumor angiogenesis.


Subject(s)
Neoplasms, Experimental/blood supply , Proto-Oncogene Proteins/physiology , Animals , Antigens, CD/analysis , Cadherins/analysis , Capillary Permeability , Endothelial Cells/physiology , Endothelial Cells/ultrastructure , Liver/blood supply , Liver/ultrastructure , Mice , Mice, Inbred C57BL , Myocardium/ultrastructure , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Proto-Oncogene Proteins/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
17.
Biomed Eng Online ; 6: 41, 2007 Nov 04.
Article in English | MEDLINE | ID: mdl-17980048

ABSTRACT

It is well established that vascularization is critical for osteogenesis. However, adequate vascularization also remains one of the major challenges in tissue engineering of bone. This problem is further accentuated in regeneration of large volume of tissue. Although a complex process, vascularization involves reciprocal regulation and functional interaction between endothelial and osteoblast-like cells during osteogenesis. This prompted us to investigate the possibility of producing bone tissue both in vitro and ectopically in vivo using vascular endothelial cells because we hypothesized that the direct contact or interaction between vascular endothelial cells and bone marrow mesenchymal stem cells are of benefit to osteogenesis in vitro and in vivo. For that purpose we co-cultured rat bone marrow mesenchymal stem cells (MSC) and kidney vascular endothelial cells (VEC) with polylactide-glycolic acid scaffolds. In vitro experiments using alkaline phosphatase and osteocalcin assays demonstrated the proliferation and differentiation of MSC into osteoblast-like cells, especially the direct contact between VEC and MSC. In addition, histochemical analysis with CD31 and von-Willebrand factor staining showed that VEC retained their endothelial characteristics. In vivo implantation of MSC and VEC co-cultures into rat's muscle resulted in pre-vascular network-like structure established by the VEC in the PLGA. These structures developed into vascularized tissue, and increased the amount and size of the new bone compared to the control group (p < 0.05). These results suggest that the vascular endothelial cells could efficiently stimulate the in vitro proliferation and differentiation of osteoblast-like cells and promote osteogenesis in vivo by the direct contact or interaction with the MSC. This technique for optimal regeneration of bone should be further investigated.


Subject(s)
Bone Marrow Cells/metabolism , Endothelium, Vascular/cytology , Kidney/metabolism , Lactic Acid/chemistry , Mesenchymal Stem Cells/cytology , Polyglycolic Acid/chemistry , Polymers/chemistry , Alkaline Phosphatase/metabolism , Animals , Cell Culture Techniques/instrumentation , Cell Differentiation , Cell Proliferation , Coculture Techniques , Endothelial Cells/cytology , Endothelium, Vascular/metabolism , Models, Biological , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteogenesis , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Tissue Engineering/instrumentation , Tissue Engineering/methods
18.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 42(3): 184-5, 2007 Mar.
Article in Chinese | MEDLINE | ID: mdl-17565833

ABSTRACT

OBJECTIVE: To investigate the expression of gene TRAIL driven by human telomerase reverse transcriptase (hTERT) promoter in SACC-83 cell tumor necrosis factor related apoptosis in dncing ligand. METHODS: After pACTERT-TRAIL plasmid transfected was into SACC-83 and HEL cells through liposome, the expression of TRAIL was examined using RT-PCR technique, the cells' survival rate by methyl thiazolyl tetrazolium (MTT) method and apoptosis rate by flow cytometry. RESULTS: Expression of extrinsic TRAIL gene driven by hTERT promoter was detected in SACC-83 cells, and not detected in human embryonic lung fibroblast (HEL) cells. After transfection of pACTERT-TRAIL, the proliferation of SACC-83 cells was significantly inhibited, and its apoptotic rate was promoted, whereas no inhibited effect was observed on HEL cells and its apoptotic rate showed little change. CONCLUSIONS: hTERT promoter can be used to induce tumor-specific expression of TRAIL gene and apoptosis in SACC-83 cells.


Subject(s)
Carcinoma, Adenoid Cystic/pathology , Salivary Gland Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/genetics , Telomerase/genetics , Apoptosis , Carcinoma, Adenoid Cystic/genetics , Cell Line, Tumor , Cell Proliferation , Gene Targeting , Genetic Vectors , Humans , Salivary Gland Neoplasms/genetics , Transfection
19.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 25(6): 544-7, 2007 Dec.
Article in Chinese | MEDLINE | ID: mdl-18306621

ABSTRACT

OBJECTIVE: To investigate the mechanical character, microleakage and mineralizing potential of nano-hydroxyapatite (nano-HAP)-added glass ionomer cement(GIC). METHODS: 8% nano-HAP were incorporated into GIC as composite, and pure GIC as control. Both types of material were used to make 20 cylinders respectively in order to detect three-point flexural strength and compressive strength. Class V cavities were prepared in 120 molars extracted for orthodontic treatment, then were filled by two kinds of material. The microleakage at the composite-dentine interface was observed with confocal laser scanning microscope (CLSM) after stained with 1% rhodamin-B-isothiocyanate for 24 hours. Class V cavities were prepared in the molars of 4 healthy dogs, filled with composite, and the same molars in the other side were filled with GIC as control. The teeth were extracted to observe the mineralizing property with polarimetric microscope in 8 weeks after filling. RESULTS: Three-point flexural strength and compressive of nano-HAP-added GIC were increased compared with pure GIC (P < 0.001, P < 0.05). The nanoleakages and microleakages appeared at the material-dentine interface in the two groups, but there were more microleakages in control group than in experiment group (P = 0.004). New crystals of hydroxyapatite were formed into a new mineralizing zone at the interface of tooth and nano-HAP-added GIC, while there was no hydroxyapatite crystals formed at the interface of tooth and pure GIC. CONCLUSION: 8% nano-HAP-added GIC can tightly fill tooth and have mineralizing potential, and can be used as liner or filling material for prevention.


Subject(s)
Durapatite , Glass Ionomer Cements , Animals , Dentin , Dogs
20.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 25(6): 584-7, 2007 Dec.
Article in Chinese | MEDLINE | ID: mdl-18306632

ABSTRACT

OBJECTIVE: To study the apoptotic effect on the squamous cell carcinoma cell line TCa83 induced by recombined adenovirus vector containing TRAIL gene and CMV promoter. METHODS: The TCa83 cell line was firstly infected with different titre of AdCMV-EGFP containing enhanced green fluorescence protein gene (EGFP) as control, and investigated the transducing rate through fluorescence to obtain the definite titre. Then TCa83 cell line was infected with AdCMV-TRAIL in proper titre, and TRAIL gene was detected by means of RT-PCR. After TCa83 cell line was infected with AdCMV-TRAIL and AdCMV-EGFP at day 1, 3, 5, 7, the activity of TCa83 cell line were evaluated by MIT and the apoptosis were detected by flow cytometer. RESULTS: Proper titre was of 1,000 particles/cell, and TCa83 cell line could be infected 100% in this titre. TRAIL gene was detected by RT-PCR after infected with AdCMV-TRAIL. The activity of TCa83 decreased in both groups, but the AdCMV-TRAIL group decreased more sharply than AdCMV-EGFP group (P < 0.001). Both AdCMV-TRAIL and AdCMV-EGFP could lead to apoptosis of TCa83 cells, but the AdCMV-TRAIL, function stronger than AdCMV-EGFP. Especially there was remarkable statistic difference between two groups (P < 0.0001). CONCLUSION: AdCMV-TRAIL could effectively decrease the activity of TCa83 cell line and induce apoptosis.


Subject(s)
Adenoviridae , Genetic Vectors , Apoptosis , Carcinoma, Squamous Cell , Cell Line , Green Fluorescent Proteins , Humans , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...