Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nat Commun ; 13(1): 3869, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35790753

ABSTRACT

Locally advanced cancers remain therapeutically challenging to eradicate. The most successful treatments continue to combine decades old non-targeted chemotherapies with radiotherapy that unfortunately increase normal tissue damage in the irradiated field and have systemic toxicities precluding further treatment intensification. Therefore, alternative molecularly guided systemic therapies are needed to improve patient outcomes when applied with radiotherapy. In this work, we report a trimodal precision cytotoxic chemo-radio-immunotherapy paradigm using spatially targeted auristatin warheads. Tumor-directed antibodies and peptides conjugated to radiosensitizing monomethyl auristatin E (MMAE) specifically produce CD8 T cell dependent durable tumor control of irradiated tumors and immunologic memory. In combination with ionizing radiation, MMAE sculpts the tumor immune infiltrate to potentiate immune checkpoint inhibition. Here, we report therapeutic synergies of targeted cytotoxic auristatin radiosensitization to stimulate anti-tumor immune responses providing a rationale for clinical translational of auristatin antibody drug conjugates with radio-immunotherapy combinations to improve tumor control.


Subject(s)
Immunoconjugates , Neoplasms , Aminobenzoates , Antibodies, Neoplasm , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Immunotherapy , Neoplasms/therapy , Oligopeptides , Peptides
2.
Neurooncol Adv ; 2(1): vdaa062, 2020.
Article in English | MEDLINE | ID: mdl-32642714

ABSTRACT

BACKGROUND: Identifying mechanisms of medulloblastoma recurrence is a key to improving patient survival, and targeting treatment-resistant subpopulations within tumors could reduce disease recurrence. Expression of the granulocyte colony-stimulating factor receptor (G-CSF-R, CD114) is a potential marker of cancer stem cells, and therefore we hypothesized that a subpopulation of medulloblastoma cells would also express CD114 and would demonstrate chemoresistance and responsiveness to G-CSF. METHODS: Prevalence of CD114-positive (CD114+) cells in medulloblastoma cell lines, patient-derived xenograft (PDX) tumors, and primary patient tumor samples were assessed by flow cytometry. Growth rates, chemoresistance, and responses to G-CSF of CD114+ and CD114-negative (CD114-) cells were characterized in vitro using continuous live cell imaging and flow cytometry. Gene expression profiles were compared between CD114+ and CD114- medulloblastoma cells using quantitative RT-PCR. RESULTS: CD114+ cells were identifiable in medulloblastoma cell lines, PDX tumors, and primary patient tumors and have slower growth rates than CD114- or mixed populations. G-CSF accelerates the growth of CD114+ cells, and CD114+ cells are more chemoresistant. The CD114+ population is enriched when G-CSF treatment follows chemotherapy. The CD114+ population also has higher expression of the CSF3R, NRP-1, TWIST1, and MYCN genes. CONCLUSIONS: Our data demonstrate that a subpopulation of CD114+ medulloblastoma cells exists in cell lines and tumors, which may evade traditional chemotherapy and respond to exogenous G-CSF. These properties invite further investigation into the role of G-CSF in medulloblastoma therapy and methods to specifically target these cells.

3.
Int J Mol Sci ; 21(9)2020 May 07.
Article in English | MEDLINE | ID: mdl-32392889

ABSTRACT

Neuroblastoma is the most common extracranial solid tumor in childhood. Gain of chromosome 17q material is found in >60% of neuroblastoma tumors and is associated with poor patient prognosis. The NME1 gene is located in the 17q21.3 region, and high NME1 expression is correlated with poor neuroblastoma patient outcomes. However, the functional roles and signaling activity of NME1 in neuroblastoma cells and tumors are unknown. NME1 and NME2 have been shown to possess histidine (His) kinase activity. Using anti-1- and 3-pHis specific monoclonal antibodies and polyclonal anti-pH118 NME1/2 antibodies, we demonstrated the presence of pH118-NME1/2 and multiple additional pHis-containing proteins in all tested neuroblastoma cell lines and in xenograft neuroblastoma tumors, supporting the presence of histidine kinase activity in neuroblastoma cells and demonstrating the potential significance of histidine kinase signaling in neuroblastoma pathogenesis. We have also demonstrated associations between NME1 expression and neuroblastoma cell migration and differentiation. Our demonstration of NME1 histidine phosphorylation in neuroblastoma and of the potential role of NME1 in neuroblastoma cell migration and differentiation suggest a functional role for NME1 in neuroblastoma pathogenesis and open the possibility of identifying new therapeutic targets and developing novel approaches to neuroblastoma therapy.


Subject(s)
NM23 Nucleoside Diphosphate Kinases/metabolism , Neuroblastoma/mortality , Up-Regulation , Animals , Cell Differentiation , Cell Line, Tumor , Cell Movement , Child , Gene Expression Regulation, Neoplastic , Humans , Mice , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Transplantation , Neuroblastoma/genetics , Neuroblastoma/metabolism , Phosphorylation , Prognosis , Signal Transduction , Survival Analysis
4.
Invest New Drugs ; 38(6): 1677-1686, 2020 12.
Article in English | MEDLINE | ID: mdl-32436058

ABSTRACT

Children with aggressive pediatric solid tumors have poor outcomes and novel treatments are needed. Pediatric solid tumors demonstrate aberrant expression and activity of the fibroblast growth factor receptor (FGFR) family, suggesting FGFR inhibitors may be effective therapeutic agents. AZD4547 is a multikinase inhibitor of the FGFR1-3 kinases, and we hypothesized that AZD4547 would be effective in pediatric solid tumor preclinical models. We evaluated the effects of AZD4547 on neuroblastoma, rhabdomyosarcoma, and Ewing sarcoma cells alone and in combination with STAT3 inhibition. Continuous live cell imaging was used to measure induction of apoptosis and effects on migration. Receptor inhibition and intracellular signaling were examined by western blotting. AZD4547 treatment resulted in decreased cell confluence, increased apoptosis and reduced cell migration in all tested cell lines. AZD4547 treatment led to decreased phosphorylation of signaling proteins involved in cell survival and apoptotic pathways and increased phosphorylation of STAT3, and treatment of cell lines with AZD4547 combined with STAT3 inhibition demonstrated increased efficacy. Sensitivity to AZD4547 appears to be mediated by effects on the Ras/MAPK and JAK/STAT pathways, and AZD4547 represents a potential novel therapeutic agent for children with solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Neoplasms/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Child , Cyclic S-Oxides/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Ribosomal Protein S6 Kinases/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Wound Healing/drug effects
5.
Br J Cancer ; 123(4): 568-579, 2020 08.
Article in English | MEDLINE | ID: mdl-32457362

ABSTRACT

BACKGROUND: Regorafenib is an inhibitor of multiple kinases with aberrant expression and activity in neuroblastoma tumours that have potential roles in neuroblastoma pathogenesis. METHODS: We evaluated neuroblastoma cells treated with regorafenib for cell viability and confluence, and analysed treated cells for apoptosis and cell cycle progression. We evaluated the efficacy of regorafenib in vivo using an orthotopic xenograft model. We evaluated regorafenib-mediated inhibition of kinase targets and performed reverse-phase protein array (RPPA) analysis of neuroblastoma cells treated with regorafenib. Lastly, we evaluated the efficacy and effects of the combination of regorafenib and 13-cis-retinoic acid on intracellular signalling. RESULTS: Regorafenib treatment resulted in reduced neuroblastoma cell viability and confluence, with both induction of apoptosis and of cell cycle arrest. Regorafenib treatment inhibits known receptor tyrosine kinase targets RET and PDGFRß and intracellular signalling through the RAS/MAPK, PI3K/Akt/mTOR and Fos/Jun pathways. Regorafenib is effective against neuroblastoma tumours in vivo, and the combination of regorafenib and 13-cis-retinoic acid demonstrates enhanced efficacy compared with regorafenib alone. CONCLUSIONS: The effects of regorafenib on multiple intracellular signalling pathways and the potential additional efficacy when combined with 13-cis-retinoic acid represent opportunities to develop treatment regimens incorporating regorafenib for children with neuroblastoma.


Subject(s)
Isotretinoin/administration & dosage , Neuroblastoma/drug therapy , Phenylurea Compounds/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Signal Transduction/drug effects , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isotretinoin/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phenylurea Compounds/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Pyridines/pharmacology , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays , ras Proteins/metabolism
6.
Oncotarget ; 10(59): 6323-6333, 2019 Oct 29.
Article in English | MEDLINE | ID: mdl-31695841

ABSTRACT

Neuroblastoma is the most common extracranial solid tumor of childhood and accounts for 15% of all pediatric cancer-related deaths. New therapies are needed to improve outcomes for children with high-risk and relapsed tumors. Inhibitors of the RET kinase and the RAS-MAPK pathway have previously been shown to be effective against neuroblastoma, suggesting that combined inhibition may have increased efficacy. RXDX-105 is a small molecule inhibitor of multiple kinases, including the RET and BRAF kinases. We found that treatment of neuroblastoma cells with RXDX-105 resulted in a significant decrease in cell viability and proliferation in vitro and in tumor growth and tumor vascularity in vivo. Treatment with RXDX-105 inhibited RET phosphorylation and phosphorylation of the MEK and ERK kinases in neuroblastoma cells and xenograft tumors, and RXDX-105 treatment induced both apoptosis and cell cycle arrest. RXDX-105 also showed enhanced efficacy in combination with 13-cis-retinoic acid, which is currently a component of maintenance therapy for children with high-risk neuroblastoma. Our results demonstrate that RXDX-105 shows promise as a novel therapeutic agent for children with high-risk and relapsed neuroblastoma.

7.
J Clin Invest ; 128(1): 531-544, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29227280

ABSTRACT

Breast cancer cells with stem cell properties are key contributors to metastatic disease, and there remains a need to better understand and target these cells in human cancers. Here, we identified rare stem-like cells in patients' tumors characterized by low levels of the proapoptotic molecule p53-upregulated modulator of apoptosis (PUMA) and showed that these cells play a critical role in tumor progression that is independent of clinical subtype. A signaling axis consisting of the integrin αvß3, Src kinase, and the transcription factor Slug suppresses PUMA in these cells, promoting tumor stemness. We showed that genetic or pharmacological disruption of αvß3/Src signaling drives PUMA expression, specifically depleting these stem-like tumor cells; increases their sensitivity to apoptosis; and reduces pulmonary metastasis, with no effect on primary tumor growth. Taken together, these findings point to PUMA as a key vulnerability of stem-like cells and suggest that pharmacological upregulation of PUMA via Src inhibition may represent a strategy to selectively target these cells in a wide spectrum of aggressive breast cancers.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Animals , Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Heterografts , Humans , Integrin alpha5/genetics , Integrin alpha5/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Cancer Discov ; 7(12): 1464-1479, 2017 12.
Article in English | MEDLINE | ID: mdl-28893801

ABSTRACT

Identifying the molecular basis for cancer cell dependence on oncogenes such as KRAS can provide new opportunities to target these addictions. Here, we identify a novel role for the carbohydrate-binding protein galectin-3 as a lynchpin for KRAS dependence. By directly binding to the cell surface receptor integrin αvß3, galectin-3 gives rise to KRAS addiction by enabling multiple functions of KRAS in anchorage-independent cells, including formation of macropinosomes that facilitate nutrient uptake and ability to maintain redox balance. Disrupting αvß3/galectin-3 binding with a clinically active drug prevents their association with mutant KRAS, thereby suppressing macropinocytosis while increasing reactive oxygen species to eradicate αvß3-expressing KRAS-mutant lung and pancreatic cancer patient-derived xenografts and spontaneous tumors in mice. Our work reveals galectin-3 as a druggable target for KRAS-addicted lung and pancreas cancers, and indicates integrin αvß3 as a biomarker to identify susceptible tumors.Significance: There is a significant unmet need for therapies targeting KRAS-mutant cancers. Here, we identify integrin αvß3 as a biomarker to identify mutant KRAS-addicted tumors that are highly sensitive to inhibition of galectin-3, a glycoprotein that binds to integrin αvß3 to promote KRAS-mediated activation of AKT. Cancer Discov; 7(12); 1464-79. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1355.


Subject(s)
Galectin 3/genetics , Lung Neoplasms/genetics , ras Proteins/genetics , Animals , Galectin 3/metabolism , Humans , Lung Neoplasms/pathology , Mice , Signal Transduction
9.
Dev Cell ; 30(3): 295-308, 2014 Aug 11.
Article in English | MEDLINE | ID: mdl-25117682

ABSTRACT

Although integrin αvß3 is linked to cancer progression, its role in epithelial development is unclear. Here, we show that αvß3 plays a critical role in adult mammary stem cells (MaSCs) during pregnancy. Whereas αvß3 is a luminal progenitor marker in the virgin gland, we noted increased αvß3 expression in MaSCs at midpregnancy. Accordingly, mice lacking αvß3 or expressing a signaling-deficient receptor showed defective mammary gland morphogenesis during pregnancy. This was associated with decreased MaSC expansion, clonogenicity, and expression of Slug, a master regulator of MaSCs. Surprisingly, αvß3-deficient mice displayed normal development of the virgin gland with no effect on luminal progenitors. Transforming growth factor ß2 (TGF-ß2) induced αvß3 expression, enhancing Slug nuclear accumulation and MaSC clonogenicity. In human breast cancer cells, αvß3 was necessary and sufficient for Slug activation, tumorsphere formation, and tumor initiation. Thus, pregnancy-associated MaSCs require a TGF-ß2/αvß3/Slug pathway, which may contribute to breast cancer progression and stemness.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Integrin alphaVbeta3/metabolism , Mammary Glands, Animal/cytology , Stem Cells/cytology , Transcription Factors/metabolism , Animals , Breast Neoplasms/metabolism , Cell Differentiation , Epithelial Cells/cytology , Female , Humans , Integrin alphaVbeta3/deficiency , Mice , Pregnancy , Snail Family Transcription Factors , Transforming Growth Factor beta2/metabolism
10.
Nat Cell Biol ; 16(5): 457-68, 2014 May.
Article in English | MEDLINE | ID: mdl-24747441

ABSTRACT

Tumour cells, with stem-like properties, are highly aggressive and often show drug resistance. Here, we reveal that integrin α(v)ß3 serves as a marker of breast, lung and pancreatic carcinomas with stem-like properties that are highly resistant to receptor tyrosine kinase inhibitors such as erlotinib. This was observed in vitro and in mice bearing patient-derived tumour xenografts or in clinical specimens from lung cancer patients who had progressed on erlotinib. Mechanistically, α(v)ß3, in the unliganded state, recruits KRAS and RalB to the tumour cell plasma membrane, leading to the activation of TBK1 and NF-κB. In fact, α(v)ß3 expression and the resulting KRAS-RalB-NF-κB pathway were both necessary and sufficient for tumour initiation, anchorage independence, self-renewal and erlotinib resistance. Pharmacological targeting of this pathway with bortezomib reversed both tumour stemness and erlotinib resistance. These findings not only identify α(v)ß3 as a marker/driver of carcinoma stemness but also reveal a therapeutic strategy to sensitize such tumours to RTK inhibition.


Subject(s)
Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Integrin beta3/metabolism , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/metabolism , ral GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Clinical Trials, Phase II as Topic , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Humans , Integrin alphaVbeta3/metabolism , Integrin beta3/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Molecular Targeted Therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-rel/antagonists & inhibitors , Proto-Oncogene Proteins c-rel/metabolism , Proto-Oncogene Proteins p21(ras) , Quinazolines/therapeutic use , RNA Interference , Randomized Controlled Trials as Topic , Signal Transduction/drug effects , Spheroids, Cellular , Time Factors , Transfection , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , ral GTP-Binding Proteins/genetics , ras Proteins/genetics
11.
Cancer Res ; 70(21): 8863-73, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20861186

ABSTRACT

Maternal embryonic leucine zipper kinase (MELK) is expressed in several developing tissues, in the adult germ line, and in adult neural progenitors. MELK expression is elevated in aggressive undifferentiated tumors, correlating with poor patient outcome in human breast cancer. To investigate the role of MELK in mammary tumorigenesis in vivo, we used a MELK-green fluorescent protein (GFP) reporter mouse, which allows prospective isolation of MELK-expressing cells based on GFP fluorescence. We found that in the normal mammary gland, cells expressing high levels of MELK were enriched in proliferating cells that express markers of mammary progenitors. The isolation of cells with high levels of MELK in mammary tumors from MMTV-Wnt1/MELK-GFP bitransgenic mice resulted in a significant enrichment of tumorsphere formation in culture and tumor initiation after transplantation into mammary fat pads of syngeneic mice. Furthermore, using lentiviral delivery of MELK-specific shRNA and limiting dilution cell transplantations, we showed that MELK function is required for mammary tumorigenesis in vivo. Our findings identify MELK as a potential target in breast tumor-initiating cells.


Subject(s)
Cell Transformation, Neoplastic/pathology , Mammary Glands, Animal/enzymology , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/physiology , Animals , Biomarkers/metabolism , Female , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , Humans , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Stem Cells/enzymology , Wnt1 Protein/metabolism
12.
Proc Natl Acad Sci U S A ; 107(5): 2189-94, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080647

ABSTRACT

Pancreatic cancer is one of the most lethal malignancies. To discover functionally relevant modulators of pancreatic neoplasia, we performed activity-based proteomic profiling on primary human ductal adenocarcinomas. Here, we identify retinoblastoma-binding protein 9 (RBBP9) as a tumor-associated serine hydrolase that displays elevated activity in pancreatic carcinomas. Whereas RBBP9 is expressed in normal and malignant tissues at similar levels, its elevated activity in tumor cells promotes anchorage-independent growth in vitro as well as pancreatic carcinogenesis in vivo. At the molecular level, RBBP9 activity overcomes TGF-beta-mediated antiproliferative signaling by reducing Smad2/3 phosphorylation, a previously unknown role for a serine hydrolase in cancer biology. Conversely, loss of endogenous RBBP9 or expression of mutationally inactive RBBP9 leads to elevated Smad2/3 phosphorylation, implicating this serine hydrolase as an essential suppressor of TGF-beta signaling. Finally, RBBP9-mediated suppression of TGF-beta signaling is required for E-cadherin expression as loss of the serine hydrolase activity leads to a reduction in E-cadherin levels and a concomitant decrease in the integrity of tumor cell-cell junctions. These data not only define a previously uncharacterized serine hydrolase activity associated with epithelial neoplasia, but also demonstrate the potential benefit of functional proteomics in the identification of new therapeutic targets.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/enzymology , Amino Acid Sequence , Animals , Cadherins/metabolism , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Proteins/genetics , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , Phosphorylation , Proteomics , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism
13.
Mol Reprod Dev ; 75(5): 818-27, 2008 May.
Article in English | MEDLINE | ID: mdl-18157870

ABSTRACT

Human embryonic stem cells (hESCs) hold great promise for cell-based therapies and drug screening applications. However, growing and processing large quantities of undifferentiated hESCs is a challenging task. Conventionally, hESCs are passaged as clusters, which can limit their growth efficiency and use in downstream applications. This study demonstrates that hESCs can be passaged as single cells using Accutase, a formulated mixture of digestive enzymes. In contrast to trypsin treatment, Accutase treatment does not significantly affect the viability and proliferation rate of hESC dissociation into single cells. Accutase-dissociated single cells can be separated by FACS and proliferate as fully pluripotent hESCs. An Oct4-eGFP reporter construct engineered into hESCs was used to monitor the pluripotency of hESCs passaged with Accutase. Compared to collagenase-passaged hESCs, Accutase-treated cultures contained a larger proportion of undifferentiated (Oct4-positive) cells. Additionally, Accutase-passaged undifferentiated hESCs could be grown as monolayers without the need for monitoring and/or selection for quality hESC colonies.


Subject(s)
Cell Culture Techniques/methods , Cell Proliferation/drug effects , Embryonic Stem Cells/cytology , Enzymes/pharmacology , Pluripotent Stem Cells/cytology , Cell Survival/drug effects , Cells, Cultured , Embryonic Stem Cells/physiology , Humans , Pluripotent Stem Cells/physiology
14.
Transgenic Res ; 17(3): 403-15, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18027100

ABSTRACT

The genes that regulate the formation of blood vessels in adult tissues represent promising therapeutic targets because angiogenesis plays a role in many diseases, including cancer. We wished to develop a mouse model allowing characterization of gene function in adult angiogenic vasculature while minimizing effects on embryonic vasculature or adult quiescent vasculature. Here we describe a transgenic mouse model that allows expression of proteins in the endothelial cells of newly forming blood vessels in the adult using a selective retroviral gene delivery system. We generated transgenic mouse lines that express the TVA receptor for the RCAS avian-specific retrovirus from Flk1 gene regulatory elements that drive expression in proliferating endothelial cells. Several of these Flk1-TVA lines expressed TVA mRNA in the embryonic vasculature and TVA protein in new blood vessels growing into subcutaneous extracellular matrix implants in adult mice. In a Flk1-TVA line that was crossed with the MMTV-PyMT transgenic mammary tumor model, tumor endothelial cells also expressed the TVA protein. Furthermore, endothelial cells in extracellular matrix implants and the tumors of Flk1-TVA mice were susceptible to RCAS infection, as determined by expression of green fluorescent protein encoded by the virus. The Flk1-TVA mouse model in conjunction with the RCAS gene delivery system will be useful to study molecular mechanisms underlying adult forms of angiogenesis.


Subject(s)
Avian Proteins/genetics , Blood Vessels/metabolism , Gene Transfer Techniques , Receptors, Virus/genetics , Transgenes , Vascular Endothelial Growth Factor Receptor-2/genetics , Alpharetrovirus/genetics , Animals , Cells, Cultured , Cloning, Molecular , Disease Susceptibility , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Transgenic , Models, Biological , Neovascularization, Physiologic/genetics , Retroviridae Infections/genetics , Terminal Repeat Sequences/genetics
15.
J Biol Chem ; 281(45): 34374-80, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16959766

ABSTRACT

Stat5 and Stat3, two members of the Stat (signal transducer and activator of transcription) family, are known to play critical roles in mammopoiesis/lactogenesis and involution, respectively, in the mammary gland. Phosphotyrosine phosphatase Shp2 has been shown to dephosphorylate and thus inactivate both Stat5 and Stat3 in vitro. Paradoxically, cell culture studies also suggest a positive role of Shp2 in promoting prolactin-stimulated Stat5 activation. We have shown here that selective deletion of Shp2 in mouse mammary glands suppresses Stat5 activity during pregnancy and lactation, resulting in significant impairment of lobulo-alveolar outgrowth and lactation. In contrast, Stat3 activity was slightly up-regulated shortly before/at involution, leading to normal epithelial cell apoptosis/involution in Shp2-deficient mammary gland. Thus, Shp2 acts to promote Stat5 activation by the JAK2.prolactin receptor complex, while negatively modulating Stat3 activity before the onset of involution. This is the first demonstration that Shp2 manipulates Stat5 and Stat3 activities reciprocally in mammary epithelial cells, providing novel insight into the complex mechanisms for regulation of various Stat family members by a cytoplasmic tyrosine phosphatase.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Mammary Glands, Animal/growth & development , Protein Tyrosine Phosphatases/physiology , STAT5 Transcription Factor/metabolism , Animals , Apoptosis , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation , Genotype , Immunoenzyme Techniques , Intracellular Signaling Peptides and Proteins/genetics , Lactation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Polymerase Chain Reaction , Pregnancy , Prolactin/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , Sequence Deletion , Transgenes
16.
Cancer Res ; 65(12): 5325-36, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958580

ABSTRACT

The ErbB2 (Neu) receptor tyrosine kinase is frequently overexpressed in human breast cancers, and this phenotype correlates with a poor clinical prognosis. We examined the effects of the mammalian target of rapamycin inhibitor, rapamycin, on mammary tumorigenesis in transgenic mice bearing an activated ErbB2 (NeuYD) transgene in the absence or presence of a second transgene encoding vascular endothelial growth factor (VEGF). Treatment of NeuYD or NeuYD x VEGF mice with rapamycin dramatically inhibited tumor growth accompanied by a marked decrease in tumor vascularization. Two key events that may underlie the antitumor activity of rapamycin were decreased expression of ErbB3 and inhibition of hypoxia-inducible factor-1-dependent responses to hypoxic stress. Rapamycin exposure caused only a modest inhibition of the proliferation of tumor-derived cell lines in standard monolayer cultures, but dramatically inhibited the growth of the same cells in three-dimensional cultures, due in part to the induction of apoptotic cell death. These studies underscore the therapeutic potential of mammalian target of rapamycin inhibitors in ErbB2-positive breast cancers and indicate that, relative to monolayer cultures, three-dimensional cell cultures are more predictive in vitro models for studies of the antitumor mechanisms of rapamycin and related compounds.


Subject(s)
Breast Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Receptor, ErbB-2/biosynthesis , Sirolimus/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Cell Proliferation/drug effects , DNA-Binding Proteins/biosynthesis , Female , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Male , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nuclear Proteins/biosynthesis , Phosphorylation , Protein Kinases/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-3/biosynthesis , Spheroids, Cellular , TOR Serine-Threonine Kinases , Transcription Factors/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
17.
Cancer Res ; 64(1): 169-79, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14729621

ABSTRACT

The Neu (ErbB2, HER2) member of the epidermal growth factor receptor family is implicated in many human breast cancers. We have tested the importance of increased angiogenic signaling in the NeuYD [mouse mammary tumor virus (MMTV)-Neu(ndl)-YD5] mammary tumor model. Transgenic mice expressing vascular endothelial growth factor (VEGF)(164) from the MMTV promoter were generated. These mice expressed VEGF(164) RNA and protein at 20- to 40-fold higher levels throughout mammary gland development but exhibited normal mammary gland development and function. However, in combination with the NeuYD oncogene, VEGF(164) expression resulted in increased vascularization of hyperplastic mammary epithelium and dramatic acceleration of tumor appearance from 111 to 51 days. Gene expression profiling also indicated that the VEGF-accelerated tumors were substantially more vascularized and less hypoxic. The preferential vascularization of early hyperplastic portions of mammary epithelia in NeuYD;MMTV-VEGF animals was associated with NeuYD RNA expression, disorganization of the tight junctions, and overlapping transgenic VEGF expression. NeuYD;MMTV-VEGF(164) bigenic, tumor-bearing animals resulted in an average of 10 tumor cell colonies/lung lodged within vascular spaces. No similar lung colonies were found in control NeuYD mice with similar tumor burdens. Overall, these results demonstrate the angiogenic restriction of early hyperplastic mammary lesions. They also reinforce in vivo the importance of activated Neu in causing disorganization of mammary luminal epithelial cell junctions and provide support for an invasion-independent mechanism of metastasis.


Subject(s)
Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Neovascularization, Pathologic/pathology , Receptor, ErbB-2/physiology , Animals , Base Sequence , DNA Primers , Disease Progression , Mammary Tumor Virus, Mouse , Mice , Mice, Transgenic , Neoplasm Metastasis , Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
18.
Mol Cell Biol ; 23(23): 8614-25, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14612405

ABSTRACT

The Ets2 transcription factor is regulated by mitogen-activated protein (MAP) kinase phosphorylation of a single threonine residue. We generated by gene targeting a single codon mutation in Ets2 substituting Ala for the critical Thr-72 phosphorylation site (Ets2A72), to investigate the importance of MAP kinase activation of Ets2 in embryo and tumor development. Ets2(A72/A72) mice are viable and develop normally. However, combining the Ets2A72 allele with a deletion mutant of Ets2 results in lethality at E11.5 and shows that Ets2A72 is a hypomorphic allele. Mammary tumors caused by transgenic polyomavirus middle T antigen, activated Neu(Erbb2), or the combination of Neu and transgenic VEGF (Neu; VEGF-25) were all restricted in Ets2(A72/A72) females. The Ets2(A72/A72) restriction on Neu; VEGF-25 tumor growth was associated with increased p21Cip1 expression. The size of tumors transplanted into fat pads of mice with Ets2 targeted alleles was correlated directly with Ets2 activity and fewer stromal cells expressing matrix metalloproteinase 9 (MMP-9). Decreased MMP-3 and MMP-9 mRNAs were confirmed in Ets2(A72/A72) macrophages. Activation of Ets2 at Thr-72 acts in the stroma, downstream of vascular endothelial growth factor production, in part through the regulation of macrophage proteases to support the progression of Neu- and polyomavirus middle-T-initiated mammary tumors.


Subject(s)
DNA-Binding Proteins , Mammary Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins , Trans-Activators/metabolism , Transcription Factors , Alleles , Amino Acid Substitution , Animals , Base Sequence , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA, Complementary/genetics , Embryonic and Fetal Development , Female , Gene Expression , Gene Targeting , Heterozygote , Macrophages/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Phenotype , Phosphorylation , Pregnancy , Proto-Oncogene Protein c-ets-2 , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL