72 results on '"Daniel R, Carrasco"'
Search Results
2. Data from BCL9 Promotes Tumor Progression by Conferring Enhanced Proliferative, Metastatic, and Angiogenic Properties to Cancer Cells
- Author
-
Daniel R. Carrasco, Kenneth C. Anderson, Sanford D. Markowitz, Lois L. Myeroff, Monica Bertagnolli, Victor Pena-Cruz, Felipe Diaz-Griffero, Hiroshi Ikeda, Jui Dutta-Simmons, Moshe E. Gatt, Kohichi Takada, Yunyu Zhang, Daniel E. Carrasco, and Mala Mani
- Abstract
Several components of the Wnt signaling cascade have been shown to function either as tumor suppressor proteins or as oncogenes in multiple human cancers, underscoring the relevance of this pathway in oncogenesis and the need for further investigation of Wnt signaling components as potential targets for cancer therapy. Here, using expression profiling analysis as well as in vitro and in vivo functional studies, we show that the Wnt pathway component BCL9 is a novel oncogene that is aberrantly expressed in human multiple myeloma as well as colon carcinoma. We show that BCL9 enhances β-catenin–mediated transcriptional activity regardless of the mutational status of the Wnt signaling components and increases cell proliferation, migration, invasion, and the metastatic potential of tumor cells by promoting loss of epithelial and gain of mesenchymal-like phenotype. Most importantly, BCL9 knockdown significantly increased the survival of xenograft mouse models of cancer by reducing tumor load, metastasis, and host angiogenesis through down-regulation of c-Myc, cyclin D1, CD44, and vascular endothelial growth factor expression by tumor cells. Together, these findings suggest that deregulation of BCL9 is an important contributing factor to tumor progression. The pleiotropic roles of BCL9 reported in this study underscore its value as a drug target for therapeutic intervention in several malignancies associated with aberrant Wnt signaling. [Cancer Res 2009;69(19):7577–86]
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 3 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Author
-
Glenn Dranoff, Martin C. Mihm, Matthew Johnson, Nicholas Souders, Dobrin Draganov, Daniel R. Carrasco, Yukoh Nakazaki, and Masahisa Jinushi
- Abstract
Supplementary Figure 3 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figures 1-2 from Expression of p16Ink4a Compensates for p18Ink4c Loss in Cyclin-Dependent Kinase 4/6–Dependent Tumors and Tissues
- Author
-
Norman E. Sharpless, Yue Xiong, Keith L. Ligon, Daniel R. Carrasco, Weili Lin, Chad Torrice, Xin-Hai Pei, Janakiraman Krishnamurthy, and Matthew R. Ramsey
- Abstract
Supplementary Figures 1-2 from Expression of p16Ink4a Compensates for p18Ink4c Loss in Cyclin-Dependent Kinase 4/6–Dependent Tumors and Tissues
- Published
- 2023
- Full Text
- View/download PDF
5. Data from Expression of p16Ink4a Compensates for p18Ink4c Loss in Cyclin-Dependent Kinase 4/6–Dependent Tumors and Tissues
- Author
-
Norman E. Sharpless, Yue Xiong, Keith L. Ligon, Daniel R. Carrasco, Weili Lin, Chad Torrice, Xin-Hai Pei, Janakiraman Krishnamurthy, and Matthew R. Ramsey
- Abstract
Cell cycle progression from G1 to S phase depends on phosphorylation of pRb by complexes containing a cyclin (D type or E type) and cyclin-dependent kinase (e.g., cdk2, cdk4, or cdk6). Ink4 proteins function to oppose the action of cdk4/6-cyclin D complexes by inhibiting cdk4/6. We employed genetic and pharmacologic approaches to study the interplay among Ink4 proteins and cdk4/6 activity in vivo. Mouse embryo fibroblasts (MEF) lacking p16Ink4a and p18Ink4c showed similar growth kinetics as wild-type MEFs despite increased cdk4 activity. In vivo, germline deficiency of p16Ink4a and p18Ink4c resulted in increased proliferation in the intermediate pituitary and pancreatic islets of adult mice, and survival of p16Ink4a−/−;p18Ink4c−/− mice was significantly reduced due to aggressive pituitary tumors. Compensation among the Ink4 proteins was observed both in vivo in p18Ink4c−/− mice and in MEFs from p16Ink4a−/−, p18Ink4c−/−, or p16Ink4a−/−;p18Ink4c−/− mice. Treatment with PD 0332991, a specific cdk4/6 kinase inhibitor, abrogated proliferation in those compartments where Ink4 deficiency was associated with enhanced proliferation (i.e., islets, pituitary, and B lymphocytes) but had no effect on proliferation in other tissues such as the small bowel. These data suggest that p16Ink4a and p18Ink4c coordinately regulate the in vivo catalytic activity of cdk4/6 in specific compartments of adult mice. [Cancer Res 2007;67(10):4732–41]
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 2 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Author
-
Glenn Dranoff, Martin C. Mihm, Matthew Johnson, Nicholas Souders, Dobrin Draganov, Daniel R. Carrasco, Yukoh Nakazaki, and Masahisa Jinushi
- Abstract
Supplementary Figure 2 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figures 1-8, Methods from BCL9 Promotes Tumor Progression by Conferring Enhanced Proliferative, Metastatic, and Angiogenic Properties to Cancer Cells
- Author
-
Daniel R. Carrasco, Kenneth C. Anderson, Sanford D. Markowitz, Lois L. Myeroff, Monica Bertagnolli, Victor Pena-Cruz, Felipe Diaz-Griffero, Hiroshi Ikeda, Jui Dutta-Simmons, Moshe E. Gatt, Kohichi Takada, Yunyu Zhang, Daniel E. Carrasco, and Mala Mani
- Abstract
Supplementary Figures 1-8, Methods from BCL9 Promotes Tumor Progression by Conferring Enhanced Proliferative, Metastatic, and Angiogenic Properties to Cancer Cells
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 4 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Author
-
Glenn Dranoff, Martin C. Mihm, Matthew Johnson, Nicholas Souders, Dobrin Draganov, Daniel R. Carrasco, Yukoh Nakazaki, and Masahisa Jinushi
- Abstract
Supplementary Figure 4 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Author
-
Glenn Dranoff, Martin C. Mihm, Matthew Johnson, Nicholas Souders, Dobrin Draganov, Daniel R. Carrasco, Yukoh Nakazaki, and Masahisa Jinushi
- Abstract
The pathogenesis of malignant melanoma involves the interplay of tumor cells with normal host elements, but the underlying mechanisms are incompletely understood. Here, we show that milk fat globule EGF-8 (MFG-E8), a secreted protein expressed at high levels in the vertical growth phase of melanoma, promotes disease progression through coordinated αvβ3 integrin signaling in the tumor microenvironment. In a murine model of melanoma, MFG-E8 enhanced tumorigenicity and metastatic capacity through Akt-dependent and Twist-dependent pathways. MFG-E8 augmented melanoma cell resistance to apoptosis, triggered an epithelial-to-mesenchymal transition (EMT), and stimulated invasion and immune suppression. In human melanoma cells, MFG-E8 knockdown attenuated Akt and Twist signaling and thereby compromised tumor cell survival, EMT, and invasive ability. MFG-E8–deficient human melanoma cells also showed increased sensitivity to small molecule inhibitors of insulin-like growth factor I receptor and c-Met. Together, these findings delineate pleiotropic roles for MFG-E8 in the tumor microenvironment and raise the possibility that systemic MFG-E8 blockade might prove therapeutic for melanoma patients. [Cancer Res 2008;68(21):8889–98]
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Table 1 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Author
-
Glenn Dranoff, Martin C. Mihm, Matthew Johnson, Nicholas Souders, Dobrin Draganov, Daniel R. Carrasco, Yukoh Nakazaki, and Masahisa Jinushi
- Abstract
Supplementary Table 1 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure Legends 1-4 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Author
-
Glenn Dranoff, Martin C. Mihm, Matthew Johnson, Nicholas Souders, Dobrin Draganov, Daniel R. Carrasco, Yukoh Nakazaki, and Masahisa Jinushi
- Abstract
Supplementary Figure Legends 1-4 from Milk Fat Globule EGF-8 Promotes Melanoma Progression through Coordinated Akt and Twist Signaling in the Tumor Microenvironment
- Published
- 2023
- Full Text
- View/download PDF
12. Multiplex flow cytometry barcoding and antibody arrays identify surface antigen profiles of primary and metastatic colon cancer cell lines.
- Author
-
Kumar Sukhdeo, Rosanto I Paramban, Jason G Vidal, Jeanne Elia, Jody Martin, Maricruz Rivera, Daniel R Carrasco, Awad Jarrar, Matthew F Kalady, Christian T Carson, Robert Balderas, Anita B Hjelmeland, Justin D Lathia, and Jeremy N Rich
- Subjects
Medicine ,Science - Abstract
Colon cancer is a deadly disease affecting millions of people worldwide. Current treatment challenges include management of disease burden as well as improvements in detection and targeting of tumor cells. To identify disease state-specific surface antigen signatures, we combined fluorescent cell barcoding with high-throughput flow cytometric profiling of primary and metastatic colon cancer lines (SW480, SW620, and HCT116). Our multiplexed technique offers improvements over conventional methods by permitting the simultaneous and rapid screening of cancer cells with reduced effort and cost. The method uses a protein-level analysis with commercially available antibodies on live cells with intact epitopes to detect potential tumor-specific targets that can be further investigated for their clinical utility. Multiplexed antibody arrays can easily be applied to other tumor types or pathologies for discovery-based approaches to target identification.
- Published
- 2013
- Full Text
- View/download PDF
13. Developmental Regulation of Mitochondrial Apoptosis by c-Myc Governs Age- and Tissue-Specific Sensitivity to Cancer Therapeutics
- Author
-
Daniel J. Murphy, Joseph R. Madsen, Daniel R. Carrasco, Cameron Fraser, Marti Goldenberg, Anthony Letai, Adam Cantlon, Sudeshna Fisch, Jing Deng, Wei-Ting Chang, Jeremy Ryan, Gail Golomb-Mello, Shenandoah Robinson, Kristopher A. Sarosiek, Chris Corbett, Dominic M. Walsh, Samuel K. McBrayer, John M. Sedivy, Ronglih Liao, Nathiya Muthalagu, Javid Moslehi, Brian Jian, and Patrick Bhola
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Somatic cell ,Apoptosis ,Biology ,Mitochondrion ,Article ,Proto-Oncogene Proteins c-myc ,Mice ,03 medical and health sciences ,Neoplasms ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Radiosensitivity ,bcl-2-Associated X Protein ,Age Factors ,Neurotoxicity ,Cell Biology ,medicine.disease ,Pediatric cancer ,Mitochondria ,3. Good health ,bcl-2 Homologous Antagonist-Killer Protein ,030104 developmental biology ,Oncology ,Doxorubicin ,Organ Specificity ,Immunology ,Cancer research - Abstract
It is not understood why healthy tissues can exhibit varying levels of sensitivity to the same toxic stimuli. Using BH3 profiling, we find that mitochondria of many adult somatic tissues, including brain, heart, and kidneys, are profoundly refractory to pro-apoptotic signaling, leading to cellular resistance to cytotoxic chemotherapies and ionizing radiation. In contrast, mitochondria from these tissues in young mice and humans are primed for apoptosis, predisposing them to undergo cell death in response to genotoxic damage. While expression of the apoptotic protein machinery is nearly absent by adulthood, in young tissues its expression is driven by c-Myc, linking developmental growth to cell death. These differences may explain why pediatric cancer patients have a higher risk of developing treatment-associated toxicities.
- Published
- 2017
- Full Text
- View/download PDF
14. TRIM13 (RFP2) downregulation decreases tumour cell growth in multiple myeloma through inhibition of NF Kappa B pathway and proteasome activity
- Author
-
Alexei Protopopov, Moshe E. Gatt, Daniel E. Carrasco, Bart Barlogie, Daniel R. Carrasco, John D. Shaughnessy, Teru Hideshima, Olle Sangfelt, Elena Ivanova, Kohichi Takada, Mala Mani, Dan Grandér, Kenneth C. Anderson, Mikael Lerner, and Marjorie Pick
- Subjects
Proteasome Endopeptidase Complex ,Cell Survival ,Down-Regulation ,Apoptosis ,Biology ,Article ,Gene product ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Humans ,Multiple myeloma ,Chromosomes, Human, Pair 13 ,Cell growth ,Gene Expression Profiling ,Tumor Suppressor Proteins ,Cell Cycle ,NF-kappa B ,Hematology ,Cell cycle ,medicine.disease ,NFKB1 ,Molecular biology ,DNA-Binding Proteins ,medicine.anatomical_structure ,Proteasome ,Gene Knockdown Techniques ,Bone marrow ,Chromosome Deletion ,Multiple Myeloma ,Cell Division - Abstract
Multiple myeloma (MM) is an incurable neoplasm caused by proliferation of malignant plasma cells in the bone marrow (BM). MM is characterized frequently by a complete or partial deletion of chromosome 13q14, seen in more than 50% of patients at diagnosis. Within this deleted region the tripartite motif containing 13 (TRIM13, also termed RFP2) gene product has been proposed to be a tumour suppressor gene (TSG). Here, we show that low expression levels of TRIM13 in MM are associated with chromosome 13q deletion and poor clinical outcome. We present a functional analysis of TRIM13 using a loss-of-function approach, and demonstrate that TRIM13 downregulation decreases tumour cell survival as well as cell cycle progression and proliferation of MM cells. In addition, we provide evidence for the involvement of TRIM13 downregulation in inhibiting the NF kappa B pathway and the activity of the 20S proteasome. Although this data does not support a role of TRIM13 as a TSG, it substantiates important roles of TRIM13 in MM tumour survival and proliferation, underscoring its potential role as a novel target for therapeutic intervention.
- Published
- 2013
- Full Text
- View/download PDF
15. PIK3CA Mutations in In situ and Invasive Breast Carcinomas
- Author
-
Sarah M. Kehoe, Alexander Miron, Hee Jung Kim, D. Craig Allred, So Yeon Park, Laura E. MacConaill, Deborah A. Dillon, J. Dirk Iglehart, Daniel R. Carrasco, Hailun Li, Lauren Luongo, Kornelia Polyak, Maria Varadi, Eun Yoon Cho, Gretchen Lewis, and Rebecca Gelman
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Class I Phosphatidylinositol 3-Kinases ,Breast Neoplasms ,Tumor initiation ,Biology ,Article ,Phosphatidylinositol 3-Kinases ,Breast cancer ,Biomarkers, Tumor ,medicine ,Humans ,Neoplasm Invasiveness ,skin and connective tissue diseases ,neoplasms ,Carcinoma in situ ,Cancer ,Exons ,Ductal carcinoma ,medicine.disease ,Oncology ,Tumor progression ,Mutation ,Female ,Breast disease ,Breast carcinoma ,Carcinoma in Situ - Abstract
The PIK3 signaling pathway has been identified as one of the most important and most frequently mutated pathways in breast cancer. Somatic mutations in the catalytic subunit of PIK3CA have been found in a significant fraction of breast carcinomas, and it has been proposed that mutant PIK3CA plays a role in tumor initiation. However, the majority of primary human tumors analyzed for genetic alterations in PIK3CA have been invasive breast carcinomas and the frequency of PIK3CA mutations in preinvasive lesions has not been explored. To investigate this, we sequenced exons 9 and 20 of PIK3CA in pure ductal carcinoma in situ (DCIS), DCIS adjacent to invasive carcinoma, and invasive ductal breast carcinomas. In a subset of cases, both in situ and invasive areas were analyzed from the same tumor. We found that the frequency of PIK3CA mutations was essentially the same (∼30%) in all three histologic groups. In some cases, in situ and invasive areas of the same tumor were discordant for PIK3CA status, and in two cases in which multiple invasive and adjacent in situ areas within the same tumor were analyzed independently, we detected intratumor heterogeneity for PIK3CA mutations. Our results suggest that mutation of PIK3CA is an early event in breast cancer that is more likely to play a role in breast tumor initiation than in invasive progression, although a potential role for exon 9 mutations in the progression of a subset of DCIS cases cannot be excluded. Cancer Res; 70(14); 5674–8. ©2010 AACR.
- Published
- 2010
- Full Text
- View/download PDF
16. BCL9 Promotes Tumor Progression by Conferring Enhanced Proliferative, Metastatic, and Angiogenic Properties to Cancer Cells
- Author
-
Monica M. Bertagnolli, Daniel E. Carrasco, Yunyu Zhang, Sanford D. Markowitz, Lois Myeroff, Moshe E. Gatt, Mala Mani, Daniel R. Carrasco, Hiroshi Ikeda, Kohichi Takada, Jui Dutta-Simmons, Kenneth C. Anderson, Felipe Diaz-Griffero, and Victor Pena-Cruz
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,Angiogenesis ,Cell Growth Processes ,medicine.disease_cause ,Article ,Cell Movement ,Cell Line, Tumor ,medicine ,Humans ,RNA, Messenger ,Neoplasm Metastasis ,Neovascularization, Pathologic ,Oncogene ,biology ,CD44 ,Wnt signaling pathway ,LRP5 ,Neoplasm Proteins ,Wnt Proteins ,Hyaluronan Receptors ,Oncology ,Tumor progression ,Colonic Neoplasms ,Cancer cell ,Disease Progression ,biology.protein ,Cancer research ,Multiple Myeloma ,Carcinogenesis ,Transcription Factors - Abstract
Several components of the Wnt signaling cascade have been shown to function either as tumor suppressor proteins or as oncogenes in multiple human cancers, underscoring the relevance of this pathway in oncogenesis and the need for further investigation of Wnt signaling components as potential targets for cancer therapy. Here, using expression profiling analysis as well as in vitro and in vivo functional studies, we show that the Wnt pathway component BCL9 is a novel oncogene that is aberrantly expressed in human multiple myeloma as well as colon carcinoma. We show that BCL9 enhances β-catenin–mediated transcriptional activity regardless of the mutational status of the Wnt signaling components and increases cell proliferation, migration, invasion, and the metastatic potential of tumor cells by promoting loss of epithelial and gain of mesenchymal-like phenotype. Most importantly, BCL9 knockdown significantly increased the survival of xenograft mouse models of cancer by reducing tumor load, metastasis, and host angiogenesis through down-regulation of c-Myc, cyclin D1, CD44, and vascular endothelial growth factor expression by tumor cells. Together, these findings suggest that deregulation of BCL9 is an important contributing factor to tumor progression. The pleiotropic roles of BCL9 reported in this study underscore its value as a drug target for therapeutic intervention in several malignancies associated with aberrant Wnt signaling. [Cancer Res 2009;69(19):7577–86]
- Published
- 2009
- Full Text
- View/download PDF
17. Aurora kinase A is a target of Wnt/β-catenin involved in multiple myeloma disease progression
- Author
-
Daniel R. Carrasco, Yunyu Zhang, Moshe E. Gatt, Kohichi Takada, Gullu Gorgun, Anthony Letai, Teru Hideshima, Jui Dutta-Simmons, Kenneth C. Anderson, Mala Mani, Noopur Raje, Daniel E. Carrasco, Nicole Carlson, and Yu-Tzu Tai
- Subjects
Transplantation, Heterologous ,Immunology ,Aurora inhibitor ,Mice, Transgenic ,Mice, SCID ,Protein Serine-Threonine Kinases ,Biology ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Mice ,Aurora kinase ,Aurora Kinases ,Mice, Inbred NOD ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,beta Catenin ,Multiple myeloma ,Aurora Kinase A ,Cell Proliferation ,Cell Cycle ,Wnt signaling pathway ,Cancer ,Cell Biology ,Hematology ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Wnt Proteins ,Drug Resistance, Neoplasm ,Catenin ,Disease Progression ,Cancer research ,Multiple Myeloma ,Monoclonal gammopathy of undetermined significance - Abstract
Multiple myeloma (MM) is a cancer of plasma cells with complex molecular characteristics that evolves from monoclonal gammopathy of undetermined significance, a highly prevalent premalignant condition. MM is the second most frequent hematologic cancer in the United States, and it remains incurable, thereby highlighting the need for new therapeutic approaches, particularly those targeting common molecular pathways involved in disease progression and maintenance, shared across different MM subtypes. Here we report that Wnt/β-catenin is one such pathway. We document the involvement of β-catenin in cell-cycle regulation, proliferation, and invasion contributing to enhanced proliferative and metastatic properties of MM. The pleiotropic effects of β-catenin in MM correlate with its transcriptional function, and we demonstrate regulation of a novel target gene, Aurora kinase A, implicating β-catenin in G2/M regulation. β-catenin and Aurora kinase A are present in most MM but not in normal plasma cells and are expressed in a pattern that parallels progression from monoclonal gammopathy of undetermined significance to MM. Our data provide evidence for a novel functional link between β-catenin and Aurora kinase A, underscoring a critical role of these pathways in MM disease progression.
- Published
- 2009
- Full Text
- View/download PDF
18. Epithelial and Stromal Cathepsin K and CXCL14 Expression in Breast Tumor Progression
- Author
-
Rebecca Gelman, Daniel R. Carrasco, Laura C. Collins, Ian E. Krop, Min Hu, Jun Yao, Yu-Hui Chen, Noga Bloushtain-Qimron, Pedram Argani, Kornelia Polyak, Michael S. Sabel, Stine Kathrein Kraeft, Stuart J. Schnitt, and Celina G. Kleer
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Stromal cell ,Cathepsin K ,Gene Expression ,Breast Neoplasms ,Biology ,medicine.disease_cause ,Article ,Epithelium ,Biopsy ,medicine ,Humans ,CXCL14 ,medicine.diagnostic_test ,Ductal carcinoma ,Cathepsins ,Oncology ,Tumor progression ,Disease Progression ,Immunohistochemistry ,Female ,Stromal Cells ,Carcinogenesis ,Chemokines, CXC - Abstract
Purpose: To evaluate the expression of cathepsin K (CTSK) and CXCL14 in stromal and epithelial cells in human breast tumor progression. Experimental Design: We did immunohistochemical analyses of CTSK and CXCL14 expression in normal breast tissue, biopsy sites, benign lesions, ductal carcinoma in situ, and invasive breast tumors of different stages. Expression patterns were related to histopathologic characteristics of the tumors and clinical outcome. The effect of CTSK+ breast stromal fibroblasts on CTSK- breast cancer cells was assessed in coculture. Results: Epithelial expression of CTSK was rarely detected in any of the tissue samples analyzed, whereas CXCL14-positive epithelial cells were found in all tissue types. The expression of CXCL14 was not associated with any tumor or patient characteristics analyzed. Stromal CTSK expression was significantly higher in invasive compared with in situ carcinomas, and in one of the two data sets analyzed, it correlated with higher tumor stage. Among all samples examined, the highest stromal CTSK levels were detected in biopsy sites. Neither epithelial nor stromal expression of CTSK was significantly associated with recurrence-free or overall survival. Coculture of CTSK+ fibroblasts enhanced the invasion of CTSK- breast tumor epithelial cells and this was blocked by CTSK inhibitors. Conclusions: CTSK may function as a paracrine factor in breast tumorigenesis. CTSK+ fibroblasts may play a role in tumor progression by promoting the invasiveness of tumor epithelial cells. The possibility that CTSK inhibitors may have a clinical role in decreasing the risk of tumor progression merits further investigation.
- Published
- 2008
- Full Text
- View/download PDF
19. Regulation of In Situ to Invasive Breast Carcinoma Transition
- Author
-
Yuri Nikolsky, Erica L. Bauerlein, Daniel R. Carrasco, Kornelia Polyak, Jun Yao, Haiyan Chen, Min Qi Hu, William C. Hahn, Stanislawa Weremowicz, Shelia M. Violette, Craig Allred, Andrea S. Richardson, Danielle K. Carroll, Stuart J. Schnitt, Rebecca Gelman, Mina J. Bissell, and Tatiana Nikolskaya
- Subjects
Cancer Research ,Breast Neoplasms ,CELLCYCLE ,Biology ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,Cell Adhesion ,Carcinoma ,medicine ,Homeostasis ,Humans ,Neoplasm Invasiveness ,skin and connective tissue diseases ,Cell adhesion ,Oligonucleotide Array Sequence Analysis ,030304 developmental biology ,0303 health sciences ,Myoepithelial cell ,Cell Biology ,Transforming growth factor beta ,DNA Methylation ,Ductal carcinoma ,Cell cycle ,medicine.disease ,3. Good health ,Gene Expression Regulation, Neoplastic ,Carcinoma, Intraductal, Noninfiltrating ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,DNA methylation ,Immunology ,Cancer research ,biology.protein ,Female ,CELLBIO ,Tumor Suppressor Protein p53 - Abstract
The transition of ductal carcinoma in situ (DCIS) to invasive carcinoma is a key event in breast tumor progression that is poorly understood. Comparative molecular analysis of tumor epithelial cells from in situ and invasive tumors has failed to identify consistent tumor stage-specific differences. However, the myoepithelial cell layer, present only in DCIS, is a key distinguishing and diagnostic feature. To determine the contribution of non-epithelial cells to tumor progression, we analyzed the role of myoepithelial cells and fibroblasts in the progression of in situ carcinomas using a xenograft model of human DCIS. Progression to invasion was promoted by fibroblasts, but inhibited by normal myoepithelial cells. The invasive tumor cells from these progressed lesions formed DCIS rather than invasive cancers when re-injected into naive mice. Molecular profiles of myoepithelial and epithelial cells isolated from primary normal and cancerous human breast tissue samples corroborated findings obtained in the xenograft model. These results provide the proof of principle that breast tumor progression could occur in the absence of additional genetic alterations and that tumor growth and progression could be controlled by replacement of normal myoepithelial inhibitory signals.
- Published
- 2008
- Full Text
- View/download PDF
20. MHC class I chain-related protein A antibodies and shedding are associated with the progression of multiple myeloma
- Author
-
Daniel R. Carrasco, Glenn Dranoff, Nikhil C. Munshi, Yu-Tzu Tai, Jerome Ritz, Kenneth C. Anderson, Donna Neuberg, Matthew Vanneman, Rao Prabhala, and Masahisa Jinushi
- Subjects
medicine.drug_class ,Antineoplastic Agents ,Biology ,Plasma cell ,Ligands ,Monoclonal antibody ,Bortezomib ,Cross-Priming ,Isoantibodies ,Cell Line, Tumor ,hemic and lymphatic diseases ,MHC class I ,Tumor Cells, Cultured ,medicine ,Humans ,Multidisciplinary ,Histocompatibility Antigens Class I ,Dendritic Cells ,Dendritic cell ,Biological Sciences ,Prognosis ,medicine.disease ,NKG2D ,Boronic Acids ,stomatognathic diseases ,medicine.anatomical_structure ,Pyrazines ,Immunology ,Disease Progression ,Proteasome inhibitor ,biology.protein ,Multiple Myeloma ,CD8 ,Monoclonal gammopathy of undetermined significance ,medicine.drug - Abstract
Monoclonal gammopathy of undetermined significance (MGUS) is a common disorder of aging and a precursor lesion to full-blown multiple myeloma (MM). The mechanisms underlying the progression from MGUS to MM are incompletely understood but include the suppression of innate and adaptive antitumor immunity. Here, we demonstrate that NKG2D, an activating receptor on natural killer (NK) cells, CD8 + T lymphocytes, and MHC class I chain-related protein A (MICA), an NKG2D ligand induced in malignant plasma cells through DNA damage, contribute to the pathogenesis of MGUS and MM. MICA expression is increased on plasma cells from MGUS patients compared with normal donors, whereas MM patients display intermediate MICA levels and a high expression of ERp5, a protein disulfide isomerase linked to MICA shedding (sMICA). MM, but not MGUS, patients harbor circulating sMICA, which triggers the down-regulation of NKG2D and impaired lymphocyte cytotoxicity. In contrast, MGUS, but not MM, patients generate high-titer anti-MICA antibodies that antagonize the suppressive effects of sMICA and stimulate dendritic cell cross-presentation of malignant plasma cells. Bortezomib, a proteasome inhibitor with anti-MM clinical efficacy, activates the DNA damage response to augment MICA expression in some MM cells, thereby enhancing their opsonization by anti-MICA antibodies. Together, these findings reveal that the alterations in the NKG2D pathway are associated with the progression from MGUS to MM and raise the possibility that anti-MICA monoclonal antibodies might prove therapeutic for these disorders.
- Published
- 2008
- Full Text
- View/download PDF
21. Expression of p16Ink4a Compensates for p18Ink4c Loss in Cyclin-Dependent Kinase 4/6–Dependent Tumors and Tissues
- Author
-
Norman E. Sharpless, Keith L. Ligon, Janakiraman Krishnamurthy, Chad Torrice, Daniel R. Carrasco, Xin Hai Pei, Yue Xiong, Matthew R. Ramsey, and Weili Lin
- Subjects
endocrine system ,Cancer Research ,endocrine system diseases ,Pyridines ,Mice, Transgenic ,Cell Growth Processes ,Biology ,Piperazines ,Mice ,In vivo ,medicine ,Animals ,Cyclin-Dependent Kinase Inhibitor p18 ,Pituitary Neoplasms ,neoplasms ,Cyclin-Dependent Kinase Inhibitor p16 ,Cyclin ,integumentary system ,Kinase ,Cyclin-dependent kinase 4 ,Pancreatic islets ,Pituitary tumors ,Cyclin-dependent kinase 2 ,Cyclin-Dependent Kinase 5 ,Cyclin-Dependent Kinase 6 ,Fibroblasts ,Embryo, Mammalian ,medicine.disease ,Molecular biology ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Cyclin-dependent kinase 6 ,biological phenomena, cell phenomena, and immunity - Abstract
Cell cycle progression from G1 to S phase depends on phosphorylation of pRb by complexes containing a cyclin (D type or E type) and cyclin-dependent kinase (e.g., cdk2, cdk4, or cdk6). Ink4 proteins function to oppose the action of cdk4/6-cyclin D complexes by inhibiting cdk4/6. We employed genetic and pharmacologic approaches to study the interplay among Ink4 proteins and cdk4/6 activity in vivo. Mouse embryo fibroblasts (MEF) lacking p16Ink4a and p18Ink4c showed similar growth kinetics as wild-type MEFs despite increased cdk4 activity. In vivo, germline deficiency of p16Ink4a and p18Ink4c resulted in increased proliferation in the intermediate pituitary and pancreatic islets of adult mice, and survival of p16Ink4a−/−;p18Ink4c−/− mice was significantly reduced due to aggressive pituitary tumors. Compensation among the Ink4 proteins was observed both in vivo in p18Ink4c−/− mice and in MEFs from p16Ink4a−/−, p18Ink4c−/−, or p16Ink4a−/−;p18Ink4c−/− mice. Treatment with PD 0332991, a specific cdk4/6 kinase inhibitor, abrogated proliferation in those compartments where Ink4 deficiency was associated with enhanced proliferation (i.e., islets, pituitary, and B lymphocytes) but had no effect on proliferation in other tissues such as the small bowel. These data suggest that p16Ink4a and p18Ink4c coordinately regulate the in vivo catalytic activity of cdk4/6 in specific compartments of adult mice. [Cancer Res 2007;67(10):4732–41]
- Published
- 2007
- Full Text
- View/download PDF
22. Targeting the β-catenin/TCF transcriptional complex in the treatment of multiple myeloma
- Author
-
Kumar Sukhdeo, Yunyu Zhang, Mei Zheng, Melissa Rooney, Haiying He, Daniel R. Carrasco, Jui Dutta, Daniel E. Carrasco, Constantine S. Mitsiades, Kenneth C. Anderson, Mala Mani, Hiroshi Yasui, and Yu-Tzu Tai
- Subjects
Beta-catenin ,Transcription, Genetic ,Transplantation, Heterologous ,Apoptosis ,Mice, SCID ,Biology ,TCF/LEF family ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Perylene ,beta Catenin ,Regulation of gene expression ,Multidisciplinary ,Interleukin-6 ,Gene Expression Profiling ,Wnt signaling pathway ,Biological Sciences ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Wnt Proteins ,medicine.anatomical_structure ,Cell culture ,Catenin ,biology.protein ,Cancer research ,Bone marrow ,Signal transduction ,Multiple Myeloma ,TCF Transcription Factors ,Protein Binding ,Signal Transduction - Abstract
Multiple myeloma (MM) is an invariably fatal form of cancer characterized by clonal proliferation of malignant plasma cells in the bone marrow. The canonical Wnt signaling pathway is activated in MM cells through constitutively active β-catenin, a messenger molecule relevant to growth, survival, and migration of MM cells. The identification of a number of small molecular compounds, such as PKF115–584, which disrupt the interaction of the transcriptionally active β-catenin/TCF protein complex, provides valuable new therapeutic tools to target an alternative pathway in MM independent of the proteasome. Here we evaluated the transcriptional, proteomic, signaling changes, and biological sequelae associated with the inhibition of Wnt signaling in MM by PKF115–584. The compound blocks expression of Wnt target genes and induces cytotoxicity in both patient MM cells and MM cell lines without a significant effect in normal plasma cells. In xenograft models of human MM, PKF115–584 inhibits tumor growth and prolongs survival. Taken together, these data demonstrate the efficacy of disrupting the β-catenin/TCF transcriptional complex to exploit tumor dependence on Wnt signaling as a therapeutic approach in the treatment of MM.
- Published
- 2007
- Full Text
- View/download PDF
23. Novel etodolac analog SDX-308 (CEP-18082) induces cytotoxicity in multiple myeloma cells associated with inhibition of β-catenin/TCF pathway
- Author
-
Paul G. Richardson, Yutaka Okawa, KC Anderson, D Chauhan, Paola Neri, Hiroshi Yasui, Daniel R. Carrasco, Hiroshi Ikeda, Kumar Sukhdeo, Sonia Vallet, Tanyel Kiziltepe, Teru Hideshima, Enrique M. Ocio, Klaus Podar, and Noopur Raje
- Subjects
Cancer Research ,Stromal cell ,Antineoplastic Agents ,Apoptosis ,Cysteine Proteinase Inhibitors ,Biology ,chemistry.chemical_compound ,Cell Line, Tumor ,Survivin ,medicine ,Humans ,Insulin-Like Growth Factor I ,Etodolac ,Cytotoxicity ,beta Catenin ,Interleukin-6 ,Bortezomib ,Hematology ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,Cell culture ,Cancer research ,Bone marrow ,Drug Screening Assays, Antitumor ,Poly(ADP-ribose) Polymerases ,Growth inhibition ,Multiple Myeloma ,TCF Transcription Factors ,Heterocyclic Compounds, 3-Ring ,Signal Transduction ,medicine.drug - Abstract
We have reported previously that R-enantiomer of etodolac (R-etodolac), which is under investigation in phase 2 clinical trials in chronic lymphocytic leukemia, induces potent cytotoxicity at clinically relevant concentrations in multiple myeloma (MM) cells. In this study, we demonstrated that SDX-308 (CEP-18082), a novel analog of etodolac, has more potent cytotoxicity than R-etodolac against both MM cell lines and patient MM cells, including tumor cells resistant to conventional (dexamethasone, doxorubicine, melphalan) and novel (bortezomib) therapies. SDX-308-induced cytotoxicity is triggered by caspase-8/9/3 activation and poly (ADP-ribose) polymerase cleavage, followed by apoptosis. SDX-308 significantly inhibits beta-catenin/T-cell factor pathway by inhibiting nuclear translocation of beta-catenin, thereby downregulating transcription and expression of downstream target proteins including myc and survivin. Neither interleukin-6 nor insulin-like growth factor-1 protect against growth inhibition triggered by SDX-308. Importantly, growth of MM cells adherent to bone marrow (BM) stromal cells is also significantly inhibited by SDX-308. Our data therefore indicate that the novel etodolac analog SDX-308 can target MM cells in the BM milieu.
- Published
- 2007
- Full Text
- View/download PDF
24. FoxOs Are Lineage-Restricted Redundant Tumor Suppressors and Regulate Endothelial Cell Homeostasis
- Author
-
Gerald C. Chu, Daniel R. Carrasco, Diego H. Castrillon, Zuzana Tothova, Ronald A. DePinho, James W. Horner, Yonghong Xiao, Zhihu Ding, Hongkai Ji, Wing Hung Wong, Ramya Kollipara, D. Gary Gilliland, Lili Miao, Lynda Chin, Shan Jiang, and Jihye Paik
- Subjects
Cell type ,endocrine system ,Lymphoma ,Somatic cell ,Cellular differentiation ,Endothelial cell morphogenesis ,Neovascularization, Physiologic ,Cell Cycle Proteins ,Nerve Tissue Proteins ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Phosphatidylinositol 3-Kinases ,Animals ,Drosophila Proteins ,Homeostasis ,Cell Lineage ,Transcription factor ,PI3K/AKT/mTOR pathway ,Homeodomain Proteins ,Mice, Knockout ,Forkhead Box Protein O1 ,Biochemistry, Genetics and Molecular Biology(all) ,Tumor Suppressor Proteins ,fungi ,Forkhead Box Protein O3 ,Pre-B-Cell Leukemia Transcription Factor 1 ,FOXO Family ,Endothelial Cells ,Cell Differentiation ,Forkhead Transcription Factors ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,FOXO4 ,embryonic structures ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Hemangioma ,Proto-Oncogene Proteins c-akt ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,Transcription Factors - Abstract
SummaryActivated phosphoinositide 3-kinase (PI3K)-AKT signaling appears to be an obligate event in the development of cancer. The highly related members of the mammalian FoxO transcription factor family, FoxO1, FoxO3, and FoxO4, represent one of several effector arms of PI3K-AKT signaling, prompting genetic analysis of the role of FoxOs in the neoplastic phenotypes linked to PI3K-AKT activation. While germline or somatic deletion of up to five FoxO alleles produced remarkably modest neoplastic phenotypes, broad somatic deletion of all FoxOs engendered a progressive cancer-prone condition characterized by thymic lymphomas and hemangiomas, demonstrating that the mammalian FoxOs are indeed bona fide tumor suppressors. Transcriptome and promoter analyses of differentially affected endothelium identified direct FoxO targets and revealed that FoxO regulation of these targets in vivo is highly context-specific, even in the same cell type. Functional studies validated Sprouty2 and PBX1, among others, as FoxO-regulated mediators of endothelial cell morphogenesis and vascular homeostasis.
- Published
- 2007
- Full Text
- View/download PDF
25. The PTEN and INK4A/ARF tumor suppressors maintain myelolymphoid homeostasis and cooperate to constrain histiocytic sarcoma development in humans
- Author
-
Daniel R, Carrasco, Tim, Fenton, Kumar, Sukhdeo, Marina, Protopopova, Miriam, Enos, Mingjian J, You, Dolores, Di Vizio, Dolores, Divicio, Cristina, Nogueira, Jayne, Stommel, Geraldine S, Pinkus, Christopher, Fletcher, Jason L, Hornick, Webster K, Cavenee, Frank B, Furnari, and Ronald A, Depinho
- Subjects
Histiocytic Disorders, Malignant ,Cancer Research ,CELLCYCLE ,02 engineering and technology ,Histiocytic sarcoma ,medicine.disease_cause ,Methylation ,Immunophenotyping ,law.invention ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,law ,Ink4a arf ,Tumor Suppressor Protein p14ARF ,medicine ,Animals ,Homeostasis ,Humans ,PTEN ,Myeloid Cells ,Lymphocytes ,Epigenetics ,Extracellular Signal-Regulated MAP Kinases ,Cyclin-Dependent Kinase Inhibitor p16 ,030304 developmental biology ,0303 health sciences ,Mutation ,biology ,PTEN Phosphohydrolase ,Sarcoma ,Cell Biology ,Cell cycle ,021001 nanoscience & nanotechnology ,medicine.disease ,Phenotype ,Enzyme Activation ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Cancer research ,Suppressor ,0210 nano-technology ,Proto-Oncogene Proteins c-akt - Abstract
SummaryHistiocytic sarcoma (HS) is a rare malignant proliferation of histiocytes of uncertain molecular pathogenesis. Here, genetic analysis of coincident loss of Pten and Ink4a/Arf tumor suppressors in the mouse revealed a neoplastic phenotype dominated by a premalignant expansion of biphenotypic myelolymphoid cells followed by the development of HS. Pten protein loss occurred only in the histiocytic portion of tumors, suggesting a stepwise genetic inactivation in the generation of HS. Similarly, human HS showed genetic or epigenetic inactivation of PTEN, p16INK4A, and p14ARF, supporting the relevance of this genetically engineered mouse model of HS. These genetic and translational observations establish a cooperative role of Pten and Ink4a/Arf in the development of HS and provide mechanistic insights into the pathogenesis of human HS.
- Published
- 2006
- Full Text
- View/download PDF
26. High-resolution genomic profiles define distinct clinico-pathogenetic subgroups of multiple myeloma patients
- Author
-
Kenneth C. Anderson, Daniel R. Carrasco, Giovanni Tonon, Alexei Protopopov, Deepak B. Khatry, Ronald A. DePinho, Ichiro Hanamura, John D. Shaughnessy, Lynda Chin, Yunyu Zhang, Raktim Sinha, Fenghuang Zhan, Bin Feng, Marina Protopopova, Kumar Sukhdeo, Yongsheng Huang, Owen W. Stephens, James P. Stewart, Cameron Brennan, Bart Barlogie, Carrasco, Dr, Tonon, G, Huang, Y, Zhang, Y, Sinha, R, Feng, B, Stewart, Jp, Zhan, F, Khatry, D, Protopopova, M, Protopopov, A, Sukhdeo, K, Hanamura, I, Stephens, O, Barlogie, B, Anderson, Kc, Chin, L, Shaughnessy, Jr., Jd, and Brennan, C and DePinho RA
- Subjects
Cancer Research ,Gene Dosage ,Genomics ,CELLCYCLE ,Biology ,Gene dosage ,Genome ,Disease-Free Survival ,medicine ,Chromosomes, Human ,Humans ,Multiple myeloma ,Genetics ,Regulation of gene expression ,Genome, Human ,Gene Expression Profiling ,Cell Biology ,medicine.disease ,Prognosis ,Diploidy ,Human genetics ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Oncology ,Multiple Myeloma ,Comparative genomic hybridization - Abstract
SummaryTo identify genetic events underlying the genesis and progression of multiple myeloma (MM), we conducted a high-resolution analysis of recurrent copy number alterations (CNAs) and expression profiles in a collection of MM cell lines and outcome-annotated clinical specimens. Attesting to the molecular heterogeneity of MM, unsupervised classification using nonnegative matrix factorization (NMF) designed for array comparative genomic hybridization (aCGH) analysis uncovered distinct genomic subtypes. Additionally, we defined 87 discrete minimal common regions (MCRs) within recurrent and highly focal CNAs. Further integration with expression data generated a refined list of MM gene candidates residing within these MCRs, thereby providing a genomic framework for dissection of disease pathogenesis, improved clinical management, and initiation of targeted drug discovery for specific MM patients.
- Published
- 2006
- Full Text
- View/download PDF
27. Cytotoxic activity of the maytansinoid immunoconjugate B-B4–DM1 against CD138+ multiple myeloma cells
- Author
-
Antonella Gozzini, Victor S. Goldmacher, Nikhil C. Munshi, Pierfrancesco Tassone, Paola Neri, Linda L. Hylander-Gans, Charles K. Allam, Daniel R. Carrasco, Teru Hideshima, Jialan Shi, Reshma Shringarpure, Kathleen R. Whiteman, Kenneth C. Anderson, John Wijdenes, Salvatore Venuta, and Masood A. Shammas
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Immunoconjugates ,Syndecans ,Stromal cell ,Cell Survival ,Transplantation, Heterologous ,Immunology ,Antineoplastic Agents ,Bone Marrow Cells ,Mice, SCID ,Maytansinoid ,Biochemistry ,Mice ,chemistry.chemical_compound ,In vivo ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Maytansine ,Cell Proliferation ,Membrane Glycoproteins ,business.industry ,Antimicrotubule agent ,Cell Biology ,Hematology ,Immunoconjugate ,Survival Rate ,medicine.anatomical_structure ,chemistry ,Cell culture ,Case-Control Studies ,Cancer research ,Proteoglycans ,Syndecan-1 ,Bone marrow ,Multiple Myeloma ,business ,Neoplasm Transplantation - Abstract
We tested the in vitro and in vivo antitumor activity of the maytansinoid DM1 (N2′-deacetyl-N2′-(3-mercapto-1-oxopropyl)-maytansine), a potent antimicrotubule agent, covalently linked to the murine monoclonal antibody (mAb) B-B4 targeting syndecan-1 (CD138). We evaluated the in vitro activity of B-B4–DM1 against a panel of CD138+ and CD138- cell lines, as well as CD138+ patient multiple myeloma (MM) cells. Treatment with B-B4–DM1 selectively decreased growth and survival of MM cell lines, patient MM cells, and MM cells adherent to bone marrow stromal cells. We further examined the activity of B-B4–DM1 in 3 human MM models in mice: (1) severe combined immunodeficient (SCID) mice bearing subcutaneous xenografts; (2) SCID mice bearing green fluorescent protein–positive (GFP+) xenografts; and (3) SCID mice implanted with human fetal bone (SCID-hu) and subsequently injected with patient MM cells. Tumor regression and inhibition of tumor growth, improvement in overall survival, and reduction in levels of circulating human paraprotein were observed in mice treated with B-B4–DM1. Although immunohistochemical analysis demonstrates restricted CD138 expression in human tissues, the lack of B-B4 reactivity with mouse tissues precludes evaluation of its toxicity in these models. In conclusion, B-B4–DM1 is a potent anti-MM agent that kills cells in an antigen-dependent manner in vitro and mediates in vivo antitumor activity at doses that are well tolerated, providing the rationale for clinical trials of this immunoconjugate in MM.
- Published
- 2004
- Full Text
- View/download PDF
28. Loss of p16Ink4a with retention of p19Arf predisposes mice to tumorigenesis
- Author
-
James W. Horner, Andrew J. Aguirre, Diego H. Castrillon, Kee Ho Lee, Daniel R. Carrasco, Ronald A. DePinho, Norman E. Sharpless, Nabeel Bardeesy, and Emily A. Wu
- Subjects
Male ,Senescence ,Tumor suppressor gene ,Cell division ,9,10-Dimethyl-1,2-benzanthracene ,T-Lymphocytes ,Thymus Gland ,Biology ,medicine.disease_cause ,Urethane ,Mice ,Neoplasms ,Tumor Suppressor Protein p14ARF ,medicine ,Animals ,Genetic Predisposition to Disease ,neoplasms ,Cells, Cultured ,Mice, Knockout ,Mutation ,Multidisciplinary ,Kinase ,Genes, p16 ,Proteins ,Gene targeting ,Fibroblasts ,Embryo, Mammalian ,Cell Transformation, Neoplastic ,Gene Targeting ,Knockout mouse ,Carcinogens ,Cancer research ,Female ,Carcinogenesis ,Cell Division ,Gene Deletion - Abstract
The cyclin-dependent kinase inhibitor p16INK4a can induce senescence of human cells, and its loss by deletion, mutation or epigenetic silencing is among the most frequently observed molecular lesions in human cancer. Overlapping reading frames in the INK4A/ARF gene encode p16INK4a and a distinct tumour-suppressor protein, p19ARF (ref. 3). Here we describe the generation and characterization of a p16Ink4a-specific knockout mouse that retains normal p19Arf function. Mice lacking p16Ink4a were born with the expected mendelian distribution and exhibited normal development except for thymic hyperplasia. T cells deficient in p16Ink4a exhibited enhanced mitogenic responsiveness, consistent with the established role of p16Ink4a in constraining cellular proliferation. In contrast to mouse embryo fibroblasts (MEFs) deficient in p19Arf (ref. 4), p16Ink4a-null MEFs possessed normal growth characteristics and remained susceptible to Ras-induced senescence. Compared with wild-type MEFs, p16Ink4a-null MEFs exhibited an increased rate of immortalization, although this rate was less than that observed previously for cells null for Ink4a/Arf, p19Arf or p53 (refs 4, 5). Furthermore, p16Ink4a deficiency was associated with an increased incidence of spontaneous and carcinogen-induced cancers. These data establish that p16Ink4a, along with p19Arf, functions as a tumour suppressor in mice.
- Published
- 2001
- Full Text
- View/download PDF
29. Antigen-Induced Eosinophilic Lung Inflammation Develops in Mice Deficient in Chemokine Eotaxin
- Author
-
Rolf-Peter Ryseck, Daniel R. Carrasco, Yi Yang, James Loy, and Rodrigo Bravo
- Subjects
Eotaxin ,Chemokine CCL11 ,Chemokine ,Ovalbumin ,Recombinant Fusion Proteins ,Immunology ,Inflammation ,Mice, Transgenic ,Biology ,Biochemistry ,Mice ,Bacterial Proteins ,Genes, Reporter ,medicine ,Escherichia coli ,Eosinophilia ,Animals ,Antigens ,Pulmonary Eosinophilia ,Mice, Knockout ,Chemotaxis ,Dextrans ,Cell Biology ,Hematology ,Eosinophil ,respiratory system ,beta-Galactosidase ,Microspheres ,respiratory tract diseases ,Chemotaxis, Leukocyte ,medicine.anatomical_structure ,Phenotype ,Organ Specificity ,Chemokines, CC ,Thioglycolates ,Gene Targeting ,biology.protein ,Cytokines ,medicine.symptom - Abstract
The mechanisms that regulate the selective infiltration of eosinophils in certain allergic diseases are still poorly understood. The CC chemokine eotaxin is a potent chemoattractant, highly specific for eosinophils. Recent studies have implicated that eotaxin plays an important role in the recruitment of eosinophils in different inflammation processes. A number of other chemokines, cytokines, and chemoattractants also have chemotactic activities for eosinophils and some of them present high selectivity for eosinophils. To further study the role of eotaxin in inflammation, we generated mutant mice with the eotaxin gene disrupted and replaced by the Escherichia coliβ-galactosidase gene. These mice developed normally and had no histologic or hematopoietic abnormalities. Furthermore, our studies showed that the lack of eotaxin did not affect the recruitment of eosinophils in the inflammation models induced by Sephadex beads and thioglycollate, as well as in an experimental lung eosinophilia model induced by ovalbumin aerosol challenge, even at the onset of the inflammatory response. The replacement of the eotaxin gene by the β-galactosidase gene provided a useful marker to monitor the activity of the eotaxin promoter under normal conditions and after antigen challenges. Immunohistochemical staining suggested that endothelial cells were the major sources of eotaxin expression.
- Published
- 1998
- Full Text
- View/download PDF
30. Gastric Hyperplasia and Increased Proliferative Responses of Lymphocytes in Mice Lacking the COOH-terminal Ankyrin Domain of NF-κB2
- Author
-
Hideaki Ishikawa, Rolf-Peter Ryseck, Daniel R. Carrasco, Rodrigo Bravo, and Estefania Claudio
- Subjects
Ankyrins ,genetic structures ,T-Lymphocytes ,Protein subunit ,medicine.medical_treatment ,Blotting, Western ,Immunology ,Lymphocyte proliferation ,Biology ,Lymphocyte Activation ,Article ,Mice ,NF-kappa B p52 Subunit ,Proto-Oncogene Proteins ,medicine ,Animals ,Immunology and Allergy ,Ankyrin ,RNA, Messenger ,Protein Precursors ,In Situ Hybridization ,DNA Primers ,Sequence Deletion ,Mice, Knockout ,chemistry.chemical_classification ,Hyperplasia ,Stomach ,NF-kappa B ,Gene targeting ,Articles ,Flow Cytometry ,NFKB1 ,Molecular biology ,Proto-Oncogene Proteins c-rel ,DNA-Binding Proteins ,Phenotype ,Cytokine ,chemistry ,Gene Targeting ,Cytokines ,I-kappa B Proteins ,Ankyrin repeat ,Dimerization ,Cell Division - Abstract
The nfkb2 gene encodes the p100 precursor which produces the p52 protein after proteolytic cleavage of its COOH-terminal domain. Although the p52 product can act as an alternative subunit of NF-kappaB, the p100 precursor is believed to function as an inhibitor of Rel/NF-kappaB activity by cytoplasmic retention of Rel/NF-kappaB complexes, like other members of the IkappaB family. However, the physiological relevance of the p100 precursor as an IkappaB molecule has not been understood. To assess the role of the precursor in vivo, we generated, by gene targeting, mice lacking p100 but still containing a functional p52 protein. Mice with a homozygous deletion of the COOH-terminal ankyrin repeats of NF-kappaB2 (p100(-/-)) had marked gastric hyperplasia, resulting in early postnatal death. p100(-/-) animals also presented histopathological alterations of hematopoietic tissues, enlarged lymph nodes, increased lymphocyte proliferation in response to several stimuli, and enhanced cytokine production in activated T cells. Dramatic induction of nuclear kappaB-binding activity composed of p52-containing complexes was found in all tissues examined and also in stimulated lymphocytes. Thus, the p100 precursor is essential for the proper regulation of p52-containing Rel/NF-kappaB complexes in various cell types and its absence cannot be efficiently compensated for by other IkappaB proteins.
- Published
- 1997
- Full Text
- View/download PDF
31. IκBα Overexpression Delays Tumor Formation in v-rel Transgenic Mice
- Author
-
Anne Lewin, Paloma Perez, Daniel R. Carrasco, and Rodrigo Bravo
- Subjects
Genetically modified mouse ,animal structures ,P50 ,medicine.diagnostic_test ,T cell ,Immunology ,Biology ,medicine.disease ,Oncogene Proteins v-rel ,Molecular biology ,Lymphoma ,Flow cytometry ,IκBα ,medicine.anatomical_structure ,embryonic structures ,medicine ,Immunology and Allergy ,CD8 - Abstract
Summary We have previously shown that transgenic mice expressing the oncoprotein v-Rel under the control of a T cell‐specific promoter develop T cell lymphomas. Tumor formation was correlated with the presence of p50/v-Rel and v-Rel/v-Rel nuclear k B-binding activity. Since experimental evidence has led to the suggestion of a potential tumor suppressor activity for I k B a , we have studied the role of I k B a in the transforming activity of v-Rel by overexpressing I k B a in v- rel transgenic mice. Overexpression of I k B a in v- rel transgenic mice resulted in an extended survival, and the development of cutaneous T cell lymphomas of CD8 1 CD4 2 phenotype. These phenotypic alterations were associated with a dramatic reduction of p50/v-Rel, but not v-Rel/v-Rel nuclear DNA binding activity and an increased expression of the intercellular adhesion molecule 1. Our results indicate that v-Rel homodimers are active in transformation and that the capacity of v-Rel‐containing complexes to escape the inhibitory effect of I k B a may be a key element in its transforming capability.
- Published
- 1997
- Full Text
- View/download PDF
32. The v-rel oncogene promotes malignant T-cell leukemia/lymphoma in transgenic mice
- Author
-
Dorfman K, Daniel R. Carrasco, Rodrigo Bravo, and Cheryl A. Rizzo
- Subjects
Genetically modified mouse ,animal structures ,General Immunology and Microbiology ,Oncogene ,General Neuroscience ,Transgene ,T-cell leukemia ,Biology ,medicine.disease ,Oncogene Proteins v-rel ,Molecular biology ,General Biochemistry, Genetics and Molecular Biology ,Lymphoma ,Leukemia ,embryonic structures ,medicine ,Molecular Biology ,CD8 - Abstract
The oncogene product from the avian reticuloendotheliosis virus strain T, v-Rel, is a member of the Rel/ NF-kappa B family of transcription factors. The mechanism by which v-Rel induces oncogenic transformation remains unclear. Several attempts to transform mammalian cells with v-Rel have failed, suggesting that v-Rel transformation may be a species-specific event. However, here we demonstrate that v-Rel, but not a truncated c-Rel, expressed under the control of the lck promoter, efficiently induced malignancies in transgenic mice. Most of the animals died before 10 months of age and developed immature, multicentric aggressive T-cell leukemia/lymphomas. Most tumors contain CD4+CD8+ cells or CD4-CD8+ cells, which have an immature rather than a mature peripheral phenotype. No tumor development was observed in control littermates and transgenic mice expressing a truncated form of c-Rel. Tumor formation was correlated with the presence of constitutive p50/v-Rel DNA binding activity and overexpression of several kappa B-regulated genes in v-rel transgenic thymocytes. However, v-Rel is also transforming in transgenic thymocytes lacking p50, indicating that p50/v-Rel heterodimer formation is not essential for the transforming activity of v-Rel. The transforming activity of v-Rel in p50 null mice has been identified as v-Rel/v-Rel homodimers. Since tumors represent immature T-lymphocytes, constitutive v-Rel expression appears to be leukemogenic at earlier stages of T-cell development. These v-Rel mice should aid in the study of lymphoma development, T-cell development and NF-kappa B regulation.
- Published
- 1996
- Full Text
- View/download PDF
33. Targeted disruption of the BCL9/β-catenin complex inhibits oncogenic Wnt signaling
- Author
-
Ramesh A. Shivdasani, Di Zhu, Nikhil C. Munshi, Andrew L. Kung, Daniel R. Carrasco, Madeleine E. Lemieux, Jeremy Ryan, Daniel E. Carrasco, Gregory H. Bird, Kohichi Takada, David Horst, Wenqing Xu, Mala Mani, Loren D. Walensky, Jianjun Zhao, Kumar Sukhdeo, and Mariateresa Fulciniti
- Subjects
Transcription, Genetic ,Angiogenesis ,Molecular Sequence Data ,Biology ,Protein Structure, Secondary ,Article ,Mice ,BCL9 ,Transcription (biology) ,Cell Movement ,Cell Line, Tumor ,Animals ,Humans ,Amino Acid Sequence ,Intestinal Mucosa ,Transcription factor ,Wnt Signaling Pathway ,beta Catenin ,Cell Proliferation ,Neovascularization, Pathologic ,Protein Stability ,Wnt signaling pathway ,LRP6 ,LRP5 ,General Medicine ,Oncogenes ,Molecular biology ,Xenograft Model Antitumor Assays ,Cell biology ,Neoplasm Proteins ,Gene Targeting ,Catenin complex ,Colorectal Neoplasms ,Peptides ,TCF Transcription Factors ,Protein Binding ,Transcription Factors - Abstract
Deregulated Wnt/β-catenin signaling underlies the pathogenesis of a broad range of human cancers, yet the development of targeted therapies to disrupt the resulting aberrant transcription has proved difficult because the pathway comprises large protein interaction surfaces and regulates many homeostatic functions. Therefore, we have directed our efforts toward blocking the interaction of β-catenin with B cell lymphoma 9 (BCL9), a co-activator for β-catenin–mediated transcription that is highly expressed in tumors but not in the cells of origin. BCL9 drives β-catenin signaling through direct binding mediated by its α-helical homology domain 2. We developed a stabilized α helix of BCL9 (SAH-BCL9), which we show targets β-catenin, dissociates native β-catenin/BCL9 complexes, selectively suppresses Wnt transcription, and exhibits mechanism-based antitumor effects. SAH-BCL9 also suppresses tumor growth, angiogenesis, invasion, and metastasis in mouse xenograft models of Colo320 colorectal carcinoma and INA-6 multiple myeloma. By inhibiting the BCL9–β-catenin interaction and selectively suppressing oncogenic Wnt transcription, SAH-BCL9 may serve as a prototype therapeutic agent for cancers driven by deregulated Wnt signaling.
- Published
- 2012
34. Bruton tyrosine kinase inhibition is a novel therapeutic strategy targeting tumor in the bone marrow microenvironment in multiple myeloma
- Author
-
Chirag Acharya, Yu-Tzu Tai, Sun-Young Kong, Antonia Cagnetta, Yiguo Hu, Jianjun Zhao, Betty Y. Chang, Laurence Elias, Michele Cea, Mike Y Zhong, Michael A. Sellitto, Yolanda Calle, Jianhong Lin, Kenneth C. Anderson, Mariateresa Fulciniti, Daniel R. Carrasco, Steven P. Treon, Paul G. Richardson, Nikhil C. Munshi, Joseph J. Buggy, Qiuju Wang, William Matsui, and Guang Yang
- Subjects
Osteolysis ,medicine.medical_treatment ,Gene Expression ,Osteoclasts ,Mice, SCID ,Biochemistry ,Mice ,Piperidines ,Bone Marrow ,Agammaglobulinaemia Tyrosine Kinase ,Tumor Microenvironment ,Tumor ,Reverse Transcriptase Polymerase Chain Reaction ,Hematology ,Protein-Tyrosine Kinases ,medicine.anatomical_structure ,Cytokine ,RANKL ,Cytokines ,Chemokines ,Multiple Myeloma ,Stromal cell ,Cell Survival ,Immunoblotting ,Immunology ,Down-Regulation ,Biology ,SCID ,Bone resorption ,Cell Line ,Cell Line, Tumor ,medicine ,Bruton's tyrosine kinase ,Animals ,Humans ,Cell Proliferation ,Cell growth ,Adenine ,Coculture Techniques ,Pyrazoles ,Pyrimidines ,Stromal Cells ,Xenograft Model Antitumor Assays ,Cell Biology ,medicine.disease ,Molecular biology ,biology.protein ,Bone marrow - Abstract
Bruton tyrosine kinase (Btk) has a well-defined role in B-cell development, whereas its expression in osteoclasts (OCs) further suggests a role in osteoclastogenesis. Here we investigated effects of PCI-32765, an oral and selective Btk inhibitor, on osteoclastogenesis as well as on multiple myeloma (MM) growth within the BM microenvironment. PCI-32765 blocked RANKL/M-CSF–induced phosphorylation of Btk and downstream PLC-γ2 in OCs, resulting in diminished TRAP5b (ED50 = 17nM) and bone resorption activity. PCI-32765 also inhibited secretion of multiple cytokines and chemokines from OC and BM stromal cell cultures from both normal donors (ED50 = 0.5nM) and MM patients. It decreased SDF-1–induced migration of MM cells, and down-regulated MIP1-α/CCL3 in MM cells. It also blocked MM cell growth and survival triggered by IL-6 or coculture with BM stromal cells or OCs in vitro. Importantly, PCI-32765 treatment significantly inhibits in vivo MM cell growth (P < .03) and MM cell–induced osteolysis of implanted human bone chips in SCID mice. Moreover, PCI-32765 prevents in vitro colony formation by stem-like cells from MM patients. Together, these results delineate functional sequelae of Btk activation mediating osteolysis and growth of MM cells, supporting evaluation of PCI-32765 as a novel therapeutic in MM.
- Published
- 2012
35. β-catenin is dynamically stored and cleared in multiple myeloma by the proteasome-aggresome-autophagosome-lysosome pathway
- Author
-
Daniel R. Carrasco, Ito S, KC Anderson, Pena-Cruz, Mendez G, Kumar Sukhdeo, Mala Mani, Kohichi Takada, and Teru Hideshima
- Subjects
Autophagosome ,Cancer Research ,Proteasome Endopeptidase Complex ,Beta-catenin ,Article ,hemic and lymphatic diseases ,Lysosome ,Phagosomes ,medicine ,Humans ,Multiple myeloma ,beta Catenin ,Phagosome ,biology ,Hematology ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,Aggresome ,Oncology ,Proteasome ,Catenin ,biology.protein ,Microscopy, Electron, Scanning ,Lysosomes ,Multiple Myeloma - Abstract
β-catenin is dynamically stored and cleared in multiple myeloma by the proteasome–aggresome–autophagosome–lysosome pathway
- Published
- 2011
36. Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy
- Author
-
Thanh Trang T. Vo, Daniel J. DeAngelo, Michelle S. Hirsch, Paul G. Richardson, Anupama Tammareddi, Daniel R. Carrasco, Jeremy Ryan, Kristopher A. Sarosiek, Victoria Del Gaizo Moore, Richard Stone, Stephen E. Sallan, Ursula A. Matulonis, Ronny Drapkin, Triona Ni Chonghaile, David J. McConkey, Constantine S. Mitsiades, Jing Deng, Kenneth C. Anderson, Anthony Letai, Lewis B. Silverman, and Yu-Tzu Tai
- Subjects
Adult ,Male ,Myeloid ,medicine.medical_treatment ,Priming (immunology) ,Antineoplastic Agents ,Apoptosis ,Mitochondrion ,Biology ,Disease-Free Survival ,Permeability ,Mice ,Cell Line, Tumor ,Neoplasms ,medicine ,Neoplasm ,Animals ,Humans ,Child ,Aged ,Cell Proliferation ,Membrane Potential, Mitochondrial ,Ovarian Neoplasms ,Chemotherapy ,Multidisciplinary ,Remission Induction ,Middle Aged ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Peptide Fragments ,Mitochondria ,Mice, Inbred C57BL ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,Drug Resistance, Neoplasm ,Immunology ,Cancer research ,Female ,Ovarian cancer ,Multiple Myeloma ,Signal Transduction - Abstract
Cytotoxic chemotherapy targets elements common to all nucleated human cells, such as DNA and microtubules, yet it selectively kills tumor cells. Here we show that clinical response to these drugs correlates with, and may be partially governed by, the pretreatment proximity of tumor cell mitochondria to the apoptotic threshold, a property called mitochondrial priming. We used BH3 profiling to measure priming in tumor cells from patients with multiple myeloma, acute myelogenous and lymphoblastic leukemia, and ovarian cancer. This assay measures mitochondrial response to peptides derived from proapoptotic BH3 domains of proteins critical for death signaling to mitochondria. Patients with highly primed cancers exhibited superior clinical response to chemotherapy. In contrast, chemoresistant cancers and normal tissues were poorly primed. Manipulation of mitochondrial priming might enhance the efficacy of cytotoxic agents.
- Published
- 2011
37. Expression of relB transcripts during lymphoid organ development: specific expression in dendritic antigen-presenting cells
- Author
-
Rodrigo Bravo, Daniel R. Carrasco, and Rolf-Peter Ryseck
- Subjects
Lymphoid Tissue ,Transcription Factor RelB ,Antigen-Presenting Cells ,Gene Expression ,Mice, Inbred Strains ,Spleen ,Dendritic cell differentiation ,Biology ,Polymerase Chain Reaction ,Mice ,Immune system ,Proto-Oncogene Proteins ,medicine ,Animals ,Antigen-presenting cell ,Molecular Biology ,In Situ Hybridization ,RELB ,Dendritic Cells ,Immunohistochemistry ,Cell biology ,Haematopoiesis ,Lymphatic system ,medicine.anatomical_structure ,Immunology ,Transcription Factors ,Developmental Biology - Abstract
We have studied the expression of the relB gene during mouse development using in situ hybridization and immunocytochemical analysis. The results show that the expression of the relB gene is highly restricted to a sub-population of cells that colonize the lymphoid tissues and that appear very late during the process of hemapoietic diversification. RNA transcripts of relB are very low or undetectable in early and late embryos. Low relB expression is observed in the thymus at late stages of embryogenesis but rapidly increases after birth. In adult lymphoid tissues, relB is detected in the medullary region of the thymus, the periarterial lymphatic sheaths of the spleen, and the deep cortex of the lymph nodes, which correspond to the regions where T cells of mature phenotype and interdigitating dendritic cells are present. Using double immunofluorescent labeling of thymic cell suspensions, we have identified the interdigitating dendritic cells as the target of RelB expression. These cells are part of a system of antigen-presenting cells that function in the induction of several immune responses, such as, tolerance, sensitization of MHC-restricted T cells, rejection of organ transplants and formation of T-dependent antibodies. Our observations indicate that RelB may play a particular role in the signal transduction pathways that regulate dendritic cell differentiation and its cellular responses.
- Published
- 1993
- Full Text
- View/download PDF
38. MicroRNAs 15a/16-1 function as tumor suppressor genes in multiple myeloma
- Author
-
Moshe E. Gatt, Sophia Adamia, Zhangbo Chu, Margaret S. Ebert, Stephane Minvielle, Mala Mani, Jui Dutta-Simmons, Yunyu Zhang, Jianjun Zhao, Yu-Tzu Tai, Daniel E. Carrasco, Roi Gazit, Kenneth C. Anderson, Nikhil C. Munshi, Hervé Avet-Loiseau, and Daniel R. Carrasco
- Subjects
Genetics ,JAG1 ,Angiogenesis ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,law.invention ,Cell biology ,Gene expression profiling ,law ,microRNA ,Suppressor ,Coding region ,Gene ,Chromosomal Deletion - Abstract
Multiple myeloma is characterized by frequent chromosomal alterations. Deletion of chr 13, especially band 13q14, is commonly observed in early stages of MM, suggesting the presence of tumor suppressor genes within this region. Here, we functionally validate the role of the microRNAs-15a/16-1 cluster, centered at the deleted region, as TSGs and delineate their downstream target genes in MM. Using "sponge" lentiviral vectors to competitive stably inhibit mature microRNAs in vitro and in vivo, we have documented enhanced proliferative and invasive capacity of cells with stably inhibition of miR-16. Importantly, miR-16 inhibition decreased animal survival in a xenograft model of MM by increasing tumor load and host angiogenesis. Expression profiling analysis of miR-16-deficient cells identified a large number of downstream target genes including FGFR1, PI3KCa, MDM4, VEGFa, as well as secondary affected genes such as JUN and Jag1. We validated designated genes showing binding sites within the conserved 3'-UTR and also within the mRNA coding region as direct miR-16 targets, thus indicating that the miRNAs may have many more targets than anticipated by conventional prediction methods. This loss-of-function system, which mimics the 13q chromosomal deletion, provides a valuable tool to investigate their function in MM pathogenesis and their potential use as therapeutic targets.
- Published
- 2010
39. A proto-oncogene BCL6 is up-regulated in the bone marrow microenvironment in multiple myeloma cells
- Author
-
Noopur Raje, Gullu Gorgun, Daniel R. Carrasco, Hiroshi Ikeda, Paul G. Richardson, Nikhil C. Munshi, Constantine S. Mitsiades, Teru Hideshima, Hiromasa Hideshima, Dharminder Chauhan, and Kenneth C. Anderson
- Subjects
STAT3 Transcription Factor ,Stromal cell ,Immunology ,Blotting, Western ,Bone Marrow Cells ,Biology ,Biochemistry ,Proto-Oncogene Mas ,Bone Marrow ,medicine ,Tumor Cells, Cultured ,Humans ,RNA, Messenger ,RNA, Small Interfering ,STAT3 ,Janus kinase inhibitor ,Cell Proliferation ,Janus Kinases ,Lymphoid Neoplasia ,Oncogene ,Cell growth ,Interleukin-6 ,Reverse Transcriptase Polymerase Chain Reaction ,NF-kappa B ,Cell Biology ,Hematology ,Molecular biology ,Antibodies, Neutralizing ,Up-Regulation ,DNA-Binding Proteins ,medicine.anatomical_structure ,Cell culture ,Cancer research ,biology.protein ,Proto-Oncogene Proteins c-bcl-6 ,Bone marrow ,Janus kinase ,Multiple Myeloma ,Signal Transduction - Abstract
Constitutive B-cell lymphoma 6 (Bcl-6) expression was undetectable in multiple myeloma (MM) cell lines, except U266 cells. However, it was up-regulated by coculture with bone marrow (BM) stromal cell-culture supernatant (SCCS). Bcl-6 expression in patient MM cells in the BM was positive. Anti–interleukin-6 (IL-6)–neutralizing antibody significantly blocked SCCS-induced Bcl-6 in MM cells. Indeed, IL-6 strongly triggered Bcl-6 expression in MM cells, whereas Janus kinase inhibitor and STAT3 siRNA down-regulated Bcl-6. Tumor necrosis factor-α (TNF-α) also triggered Bcl-6, but independently of STAT3, whereas IκB kinaseβ inhibitor down-regulated TNF-α–induced Bcl-6, indicating that the canonical nuclear factor-κB pathway mediates TNF-α–induced Bcl-6 expression. Importantly, down-regulation of Bcl-6 by shRNA significantly inhibited MM cell growth in the presence of SCCS. Our results therefore suggest that Bcl-6 expression in MM cells is modulated, at least in part, via Janus kinase/STAT3 and canonical nuclear factor-κB pathways and that targeting Bcl-6, either directly or via these cascades, inhibits MM cell growth in the BM milieu.
- Published
- 2010
40. Milk fat globule EGF-8 promotes melanoma progression through coordinated Akt and twist signaling in the tumor microenvironment
- Author
-
Daniel R. Carrasco, Martin C. Mihm, Masahisa Jinushi, Nicholas C. Souders, Yukoh Nakazaki, Matthew Johnson, Dobrin Draganov, and Glenn Dranoff
- Subjects
Cancer Research ,medicine.medical_treatment ,Integrin ,Apoptosis ,Mice ,Immune system ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Protein kinase B ,Melanoma ,DNA Primers ,Tumor microenvironment ,biology ,Base Sequence ,Growth factor ,Twist-Related Protein 1 ,Cancer ,Nuclear Proteins ,medicine.disease ,Milk Proteins ,Oncology ,Microscopy, Fluorescence ,Immunology ,Antigens, Surface ,biology.protein ,Cancer research ,Disease Progression ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The pathogenesis of malignant melanoma involves the interplay of tumor cells with normal host elements, but the underlying mechanisms are incompletely understood. Here, we show that milk fat globule EGF-8 (MFG-E8), a secreted protein expressed at high levels in the vertical growth phase of melanoma, promotes disease progression through coordinated αvβ3 integrin signaling in the tumor microenvironment. In a murine model of melanoma, MFG-E8 enhanced tumorigenicity and metastatic capacity through Akt-dependent and Twist-dependent pathways. MFG-E8 augmented melanoma cell resistance to apoptosis, triggered an epithelial-to-mesenchymal transition (EMT), and stimulated invasion and immune suppression. In human melanoma cells, MFG-E8 knockdown attenuated Akt and Twist signaling and thereby compromised tumor cell survival, EMT, and invasive ability. MFG-E8–deficient human melanoma cells also showed increased sensitivity to small molecule inhibitors of insulin-like growth factor I receptor and c-Met. Together, these findings delineate pleiotropic roles for MFG-E8 in the tumor microenvironment and raise the possibility that systemic MFG-E8 blockade might prove therapeutic for melanoma patients. [Cancer Res 2008;68(21):8889–98]
- Published
- 2008
41. Targeting NF-kappaB in Waldenstrom macroglobulinemia
- Author
-
Irene M. Ghobrial, Mena Farag, Evdoxia Hatjiharissi, Xavier Leleu, Feda Azab, Aldo M. Roccaro, Jérôme Eeckhoute, Judith Runnels, Antonio Sacco, Thomas E. Witzig, Abdel Kareem Azab, Hai T. Ngo, Kenneth C. Anderson, Xiaoying Jia, Myles Brown, Zachary R. Hunter, Teru Hideshima, Molly R. Melhem, Anne Moreau, Daniel R. Carrasco, Nicolas Burwick, and Steven P. Treon
- Subjects
Cell Survival ,Phosphorylcholine ,Immunology ,Pharmacology ,Biology ,Biochemistry ,Bortezomib ,chemistry.chemical_compound ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Cytotoxicity ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Neoplasia ,NF-kappa B ,Waldenstrom macroglobulinemia ,NF-κB ,Drug Synergism ,Cell Biology ,Hematology ,Perifosine ,medicine.disease ,Boronic Acids ,chemistry ,Pyrazines ,Cancer research ,Waldenstrom Macroglobulinemia ,Chromatin immunoprecipitation ,medicine.drug ,Signal Transduction - Abstract
The nuclear factor-κB (NF-κB) path-way has been implicated in tumor B-cell survival, growth, and resistance to therapy. Because tumor cells overcome single-agent antitumor activity, we hypothesized that combination of agents that target differentially NF-κB pathway will induce significant cytotoxicity. Therapeutic agents that target proteasome and Akt pathways should induce significant activity in B-cell malignancies as both pathways impact NF-κB activity. We demonstrated that perifosine and bortezomib both targeted NF-κB through its recruitment to the promoter of its target gene IκB using chromatin immunoprecipitation assay. This combination led to synergistic cytotoxicity in Waldenstrom macroglobulinemia (WM) cells that was mediated through a combined reduction of the PI3K/Akt and ERK signaling pathways, found to be critical for survival of WM cells. Moreover, a combination of these drugs with the CD20 monoclonal antibody rituximab further increased their cytotoxic activity. Thus, effective WM therapy may require combination regimens targeting the NF-κB pathway.
- Published
- 2008
42. Ink4a/Arf tumor suppressor does not modulate the degenerative conditions or tumor spectrum of the telomerase-deficient mouse
- Author
-
Marcus Bosenberg, Daniel R. Carrasco, Christine M. Khoo, Ronald A. DePinho, and Jihye Paik
- Subjects
Telomerase ,Time Factors ,Tumor suppressor gene ,Transgene ,Context (language use) ,Mice, Transgenic ,In situ hybridization ,Biology ,law.invention ,Mice ,law ,Neoplasms ,medicine ,Animals ,Genes, Tumor Suppressor ,Neoplasms, Glandular and Epithelial ,neoplasms ,Cyclin-Dependent Kinase Inhibitor p16 ,In Situ Hybridization, Fluorescence ,Multidisciplinary ,Cancer ,3T3 Cells ,Biological Sciences ,Telomere ,medicine.disease ,Genes, p53 ,Molecular biology ,Phenotype ,Cancer research ,Suppressor ,Tumor Suppressor Protein p53 - Abstract
The Rb/p16 Ink4a and p53/p19Arf tumor suppressor pathways have been linked to diverse cancer-relevant processes, including those governing the cellular responses to telomere dysfunction. In this study, we sought to provide direct genetic evidence of a role for the Ink4a/Arf tumor suppressor gene, encoding both p16 Ink4a and p19 Arf , in modulating the cellular and tissue phenotypes associated with telomere dysfunction by using the mTerc Ink4a/Arf mouse model. In contrast to the rescue associated with p53 deficiency, Ink4a/Arf deficiency did not attenuate the degenerative phenotypes elicited by telomere dysfunction in the late-generation mTerc −/− mice. Furthermore, in contrast to accelerated cancer onset and increased epithelial cancers of late-generation mTerc −/− p53 mutant mice, late-generation mTerc −/− Ink4a/Arf mutant mice experienced a delayed tumor onset and maintained the lymphoma and sarcoma spectrum. Consistent with the negligible role of Ink4a/Arf in the telomere checkpoint response in vivo , late-generation mTerc −/− Ink4a/Arf −/− tissues show activated p53, and derivative tumor cell lines sustain frequent loss of p53 function, whereas all early generation mTerc Ink4a/Arf −/− tumor cell lines remain intact for p53. In addition, the late-generation mTerc −/− Ink4a/Arf −/− tumors showed activation of the alternative lengthening of telomere mechanism, underscoring the need for adaptation to the presence of telomere dysfunction in the absence of p16 Ink4a and p19 Arf . These observations highlight the importance of genetic context in dictating whether telomere dysfunction promotes or suppresses age-related degenerative conditions as well as the rate of initiation and type of spontaneous cancers.
- Published
- 2007
43. The differentiation and stress response factor, XBP-1, drives multiple myeloma pathogenesis
- Author
-
Alexei Protopopov, Nikhil C. Munshi, Ronald A. DePinho, Raktim Sinha, James W. Horner, Mei Zheng, Elena Ivanova, Daniel R. Carrasco, Kenneth C. Anderson, Marina Protopopova, Joel M. Henderson, Giovanni Tonon, Daniel E. Carrasco, Mala Mani, Kumar Sukhdeo, Geraldine S. Pinkus, Miriam Enos, Carrasco, Dr, Sukhdeo, K, Protopopova, M, Sinha, R, Enos, M, Carrasco, De, Zheng, M, Mani, M, Henderson, J, Pinkus, G, Munshi, N, Horner, J, Ivanova, Ev, Protopopov, A, Anderson, Kc, Tonon, G, and Depinho, Ra
- Subjects
Male ,X-Box Binding Protein 1 ,Cancer Research ,Aging ,Transcription, Genetic ,Cellular differentiation ,Electrophoretic Mobility Shift Assay ,CELLCYCLE ,Endoplasmic Reticulum ,Malignant transformation ,Pathogenesis ,Mice ,0302 clinical medicine ,Hypergammaglobulinemia ,Multiple myeloma ,Cells, Cultured ,0303 health sciences ,B-Lymphocytes ,Nuclear Proteins ,Cell Differentiation ,Cell cycle ,Phenotype ,3. Good health ,DNA-Binding Proteins ,Oncology ,030220 oncology & carcinogenesis ,Female ,Kidney Diseases ,Bone Diseases ,Multiple Myeloma ,Genetically modified mouse ,Transgene ,RNA Splicing ,Plasma Cells ,Mice, Transgenic ,Regulatory Factor X Transcription Factors ,Biology ,Skin Diseases ,Article ,03 medical and health sciences ,medicine ,Animals ,Humans ,030304 developmental biology ,Dromaiidae ,Cell Biology ,medicine.disease ,Mice, Inbred C57BL ,Immunology ,Cancer research ,Transcription Factors - Abstract
Summary Multiple myeloma (MM) evolves from a highly prevalent premalignant condition termed MGUS. The factors underlying the malignant transformation of MGUS are unknown. We report a MGUS/MM phenotype in transgenic mice with Eμ-directed expression of the XBP-1 spliced isoform (XBP-1s), a factor governing unfolded protein/ER stress response and plasma-cell development. Eμ-XBP-1s elicited elevated serum Ig and skin alterations. With age, Eμ-xbp-1s transgenics develop features diagnostic of human MM, including bone lytic lesions and subendothelial Ig deposition. Furthermore, transcriptional profiles of Eμ-xbp-1s lymphoid and MM cells show aberrant expression of known human MM dysregulated genes. The similarities of this model with the human disease, coupled with documented frequent XBP-1s overexpression in human MM, serve to implicate XBP-1s dysregulation in MM pathogenesis.
- Published
- 2007
44. MFG-E8-mediated uptake of apoptotic cells by APCs links the pro- and antiinflammatory activities of GM-CSF
- Author
-
Masahisa Jinushi, Daniel R. Carrasco, Yukoh Nakazaki, Glenn Dranoff, Martin C. Mihm, and Michael Dougan
- Subjects
CD4-Positive T-Lymphocytes ,Cellular differentiation ,T cell ,medicine.medical_treatment ,Antigen-Presenting Cells ,Inflammation ,Apoptosis ,Biology ,Cancer Vaccines ,Mice ,Antigen ,Phagocytosis ,Neoplasms ,medicine ,Animals ,Homeostasis ,Antigen-presenting cell ,Mice, Knockout ,Granulocyte-Macrophage Colony-Stimulating Factor ,Cell Differentiation ,General Medicine ,Immunotherapy ,Milk Proteins ,Cell biology ,Mice, Inbred C57BL ,Granulocyte macrophage colony-stimulating factor ,medicine.anatomical_structure ,Gene Expression Regulation ,Antigens, Surface ,Interleukin 12 ,medicine.symptom ,medicine.drug ,Research Article - Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) enhances protection against tumors and infections, but GM-CSF-deficient mice develop inflammatory disease. Here we show that GM-CSF is required for the expression of milk fat globule EGF 8 (MFG-E8) in antigen-presenting cells, and that MFG-E8-mediated uptake of apoptotic cells is a key determinant of GM-CSF-triggered tolerance and immunity. Upon exposure to apoptotic cells, GM-CSF-deficient antigen-presenting cells (APCs) produce an altered cytokine profile that results in decreased Tregs and increased Th1 cells, whereas concurrent ablation of IFN-gamma promotes Th17 cells. In wild-type mice, MFG-E8 attenuates the vaccination activity of GM-CSF-secreting tumor cells through Treg induction, whereas a dominant-negative MFG-E8 mutant potentiates GM-CSF-stimulated tumor destruction through Treg inhibition. These findings clarify the immunoregulatory effects of apoptotic cells and suggest new therapeutic strategies to modulate CD4(+) T cell subsets in cancer and autoimmunity.
- Published
- 2006
45. In vivo assessment of RAS-dependent maintenance of tumor angiogenesis by real-time magnetic resonance imaging
- Author
-
Minjung Kim, Ralph Weissleder, Yi Tang, Lynda Chin, Andrew L. Kung, and Daniel R. Carrasco
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Melanoma, Experimental ,Mice, Transgenic ,Biology ,Neovascularization ,Mice ,In vivo ,medicine ,Animals ,Gene Silencing ,Cyclin-Dependent Kinase Inhibitor p16 ,Neovascularization, Pathologic ,Melanoma ,medicine.disease ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Genes, ras ,Oncology ,Tumor progression ,Apoptosis ,Cancer research ,ras Proteins ,medicine.symptom ,Preclinical imaging ,Magnetic Resonance Angiography ,Blood vessel - Abstract
New blood vessel formation is a prominent feature of human cancers and tumor progression and is frequently accompanied by the acquisition of an angiogenic phenotype associated with a switch in the balance of proangiogenic and antiangiogenic molecules. This study was designed to investigate the role of activated H-RAS on the angiogenic phenotype of melanoma that arises in the inducible Tyr/Tet-RAS Ink4a/Arf−/− model using in vivo imaging with histopathologic correlation. We show that loss of RAS activity in fully established melanomas led to a reduction in tumor volume, which was preceded by impairment of vascular function as determined by in vivo magnetic resonance imaging. This correlated with activation of apoptosis in host-derived endothelial cells as well as in tumor cells. Thus, real-time in vivo imaging provided evidence that maintenance of tumor angiogenesis requires activated RAS in this model system, and that loss of vascular integrity upon inactivation of RAS is an active process rather than a consequence of loss of tumor cell viability.
- Published
- 2005
46. A clinically relevant SCID-hu in vivo model of human multiple myeloma
- Author
-
Renate Burger, Nikhil C. Munshi, Vidit Munshi, Paola Neri, Laurence Catley, Victor S. Goldmacher, Salvatore Venuta, Daniel R. Carrasco, Robert J. Fram, Masood A. Shammas, Pierfrancesco Tassone, GS Jacob, and Kenneth C. Anderson
- Subjects
Pathology ,medicine.medical_specialty ,Stromal cell ,Immunology ,Green Fluorescent Proteins ,Transplantation, Heterologous ,Mice, SCID ,Biochemistry ,Dexamethasone ,Mice ,In vivo ,Fetal Tissue Transplantation ,Transduction, Genetic ,Atiprimod ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Spiro Compounds ,Multiple myeloma ,Severe combined immunodeficiency ,Bone Transplantation ,Neoplasia ,business.industry ,Immunotoxins ,Cell Biology ,Hematology ,medicine.disease ,Receptors, Interleukin-6 ,Recombinant Proteins ,Disease Models, Animal ,medicine.anatomical_structure ,Cell culture ,Interleukin-6 receptor ,Cancer research ,Bone marrow ,business ,Multiple Myeloma ,Neoplasm Transplantation - Abstract
We developed a novel in vivo multiple myeloma (MM) model by engrafting the interleukin 6 (IL-6)-dependent human MM cell line INA-6 into severe combined immunodeficiency (SCID) mice previously given implants of a human fetal bone chip (SCID-hu mice). INA-6 cells require either exogenous human IL-6 (huIL-6) or bone marrow stromal cells (BMSCs) to proliferate in vitro. In this model, we monitored the in vivo growth of INA-6 cells stably transduced with a green fluorescent protein (GFP) gene (INA-6GFP+ cells). INA-6 MM cells engrafted in SCID-hu mice but not in SCID mice that had not been given implants of human fetal bone. The level of soluble human IL-6 receptor (shuIL-6R) in murine serum and fluorescence imaging of host animals were sensitive indicators of tumor growth. Dexamethasone as well as experimental drugs, such as Atiprimod and B-B4-DM1, were used to confirm the utility of the model for evaluation of anti-MM agents. We report that this model is highly reproducible and allows for evaluation of investigational drugs targeting IL-6-dependent MM cells in the human bone marrow (huBM) milieu. (Blood. 2005;106:713-716)
- Published
- 2005
47. In vitro and in vivo activity of the maytansinoid immunoconjugate huN901-N2'-deacetyl-N2'-(3-mercapto-1-oxopropyl)-maytansine against CD56+ multiple myeloma cells
- Author
-
Daniel R. Carrasco, Pierfrancesco Tassone, Cheng Li, Charles K. Allam, Antonella Gozzini, Kenneth C. Anderson, Salvatore Venuta, Victor S. Goldmacher, Kathleen R. Whiteman, Nikhil C. Munshi, and Masood A. Shammas
- Subjects
Male ,Cancer Research ,Stromal cell ,Mice, SCID ,CD38 ,Maytansinoid ,chemistry.chemical_compound ,Mice ,In vivo ,Immunotoxin ,Cell Line, Tumor ,Cell Adhesion ,Medicine ,Animals ,Humans ,Maytansine ,Neural Cell Adhesion Molecules ,business.industry ,Immunotoxins ,Cell Cycle ,Antibodies, Monoclonal ,hemic and immune systems ,Molecular biology ,Xenograft Model Antitumor Assays ,CD56 Antigen ,Immunoconjugate ,stomatognathic diseases ,medicine.anatomical_structure ,Oncology ,chemistry ,Cell culture ,Bone marrow ,business ,Multiple Myeloma - Abstract
HuN901 is a humanized monoclonal antibody that binds with high affinity to CD56, the neuronal cell adhesion molecule. HuN901 conjugated with the maytansinoid N2′-deacetyl-N2′-(3-mercapto-1-oxopropyl)-maytansine (DM1), a potent antimicrotubular cytotoxic agent, may provide targeted delivery of the drug to CD56 expressing tumors. Based on gene expression profiles of primary multiple myeloma (MM) cells showing expression of CD56 in 10 out of 15 patients (66.6%) and flow cytometric profiles of MM (CD38brightCD45lo) cells showing CD56 expression in 22 out of 28 patients (79%), we assessed the efficacy of huN901-DM1 for the treatment of MM. We first examined the in vitro cytotoxicity and specificity of huN901-DM1 on a panel of CD56+ and CD56− MM cell lines, as well as a CD56− Waldenstrom’s macroglobulinemia cell line. HuN901-DM1 treatment selectively decreased survival of CD56+ MM cell lines and depleted CD56+ MM cells from mixed cultures with a CD56− cell line or adherent bone marrow stromal cells. In vivo antitumor activity of huN901-DM1 was then studied in a tumor xenograft model using a CD56+ OPM2 human MM cell line in SCID mice. We observed inhibition of serum paraprotein secretion, inhibition of tumor growth, and increase in survival of mice treated with huN901-DM1. Our data therefore demonstrate that huN901-DM1 has significant in vitro and in vivo antimyeloma activity at doses that are well tolerated in a murine model. Taken together, these data provide the framework for clinical trials of this agent to improve patient outcome in MM.
- Published
- 2004
48. Identification of genes modulated in multiple myeloma using genetically identical twin samples
- Author
-
Faith E. Davies, Lief Bergsagel, Rafael Fonseca, Nikhil C. Munshi, Daniel R. Carrasco, Masood A. Shammas, Ryung S. Kim, Daniel Auclair, S. Vincent Rajkumar, Teru Hideshima, Kenneth C. Anderson, Nicholas Mitsiades, Dharminder Chauhan, Cheng Li, and Constantine S. Mitsiades
- Subjects
Adult ,Telomerase ,Proteasome Endopeptidase Complex ,Syndecans ,Genotype ,Immunology ,Cell ,Blotting, Western ,Down-Regulation ,Antigens, CD34 ,Bone Marrow Cells ,In situ hybridization ,Biology ,Biochemistry ,Models, Biological ,Multienzyme Complexes ,Gene expression ,medicine ,Diseases in Twins ,Humans ,Gene ,In Situ Hybridization, Fluorescence ,Oligonucleotide Array Sequence Analysis ,Genetics ,Regulation of gene expression ,Membrane Glycoproteins ,Neovascularization, Pathologic ,Cell Biology ,Hematology ,Twins, Monozygotic ,Molecular biology ,Immunohistochemistry ,Up-Regulation ,Gene expression profiling ,Cysteine Endopeptidases ,medicine.anatomical_structure ,Gene Expression Regulation ,Female ,Proteoglycans ,Bone marrow ,Syndecan-1 ,Multiple Myeloma - Abstract
Genetic heterogeneity between individuals confounds the comparison of gene profiling of multiple myeloma (MM) cells versus normal plasma cells (PCs). To overcome this barrier, we compared the gene expression profile of CD138+ MM cells from a patient bone marrow (BM) sample with CD138+ PCs from a genetically identical twin BM sample using microarray profiling. Two hundred and ninety-six genes were up-regulated and 103 genes were down-regulated at least 2-fold in MM cells versus normal twin PCs. Highly expressed genes in MM cells included cell survival pathway genes such as mcl-1, dad-1, caspase 8, and FADD-like apoptosis regulator (FLIP); oncogenes/transcriptional factors such as Jun-D, Xbp-1, calmodulin, Calnexin, and FGFR-3; stress response and ubiquitin/proteasome pathway–related genes and various ribosomal genes reflecting increased metabolic and translational activity. Genes that were down-regulated in MM cells versus healthy twin PCs included RAD51, killer cell immunoglobulin-like receptor protein, and apoptotic protease activating factor. Microarray results were further confirmed by Western blot analyses, immunohistochemistry, fluorescent in situ hybridization (FISH), and functional assays of telomerase activity and bone marrow angiogenesis. This molecular profiling provides potential insights into mechanisms of malignant transformation in MM. For example, FGFR3, xbp-1, and both mcl-1 and dad-1 may mediate transformation, differentiation, and survival, respectively, and may have clinical implications. By identifying genes uniquely altered in MM cells compared with normal PCs in an identical genotypic background, the current study provides the framework to identify novel therapeutic targets.
- Published
- 2003
49. Loss of the Lkb1 tumour suppressor provokes intestinal polyposis but resistance to transformation
- Author
-
Massimo Loda, Ronald A. DePinho, Nabeel Bardeesy, Norman E. Sharpless, Aram F. Hezel, Sabina Signoretti, Daniel R. Carrasco, Manisha Sinha, and Nathaniel A. Hathaway
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Stromal cell ,Tumor suppressor gene ,Angiogenesis ,STK11 ,Peutz-Jeghers Syndrome ,Biology ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Malignant transformation ,Transcriptome ,Mice ,Germline mutation ,Animals ,Genes, Tumor Suppressor ,RNA, Messenger ,skin and connective tissue diseases ,Cellular Senescence ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,Multidisciplinary ,Intracellular Signaling Peptides and Proteins ,Intestinal Polyps ,Fibroblasts ,Phenotype ,Cell Transformation, Neoplastic ,Genes, ras ,Culture Media, Conditioned ,Immunology ,Gene Targeting ,Cancer research ,Carrier Proteins ,Gene Deletion - Abstract
Germline mutations in LKB1 (also known as STK11) are associated with Peutz-Jeghers syndrome (PJS), a disorder with predisposition to gastrointestinal polyposis and cancer. PJS polyps are unusual neoplasms characterized by marked epithelial and stromal overgrowth but have limited malignant potential. Here we show that Lkb1(+/-) mice develop intestinal polyps identical to those seen in individuals affected with PJS. Consistent with this in vivo tumour suppressor function, Lkb1 deficiency prevents culture-induced senescence without loss of Ink4a/Arf or p53. Despite compromised mortality, Lkb1(-/-) mouse embryonic fibroblasts show resistance to transformation by activated Ha-Ras either alone or with immortalizing oncogenes. This phenotype is in agreement with the paucity of mutations in Ras seen in PJS polyps and suggests that loss of Lkb1 function as an early neoplastic event renders cells resistant to subsequent oncogene-induced transformation. In addition, the Lkb1 transcriptome shows modulation of factors linked to angiogenesis, extracellular matrix remodelling, cell adhesion and inhibition of Ras transformation. Together, our data rationalize several features of PJS polyposis--notably its peculiar histopathological presentation and limited malignant potential--and place Lkb1 in a distinct class of tumour suppressors.
- Published
- 2002
50. Multiplex Flow Cytometry Barcoding and Antibody Arrays Identify Surface Antigen Profiles of Primary and Metastatic Colon Cancer Cell Lines
- Author
-
Jeanne Elia, Matthew F. Kalady, Kumar Sukhdeo, Rosanto I. Paramban, Jason G. Vidal, Christian T. Carson, Robert Balderas, Maricruz Rivera, Daniel R. Carrasco, Jody L. Martin, Anita B. Hjelmeland, Justin D. Lathia, Jeremy N. Rich, and Awad Jarrar
- Subjects
Pathology ,Colorectal cancer ,Fluorescent Antibody Technique ,lcsh:Medicine ,Epitope ,Metastasis ,0302 clinical medicine ,Molecular Cell Biology ,Gastrointestinal Cancers ,Basic Cancer Research ,Tumor Cells, Cultured ,Multiplex ,Neoplasm Metastasis ,lcsh:Science ,0303 health sciences ,Multidisciplinary ,medicine.diagnostic_test ,Colon Adenocarcinoma ,Flow Cytometry ,Immunohistochemistry ,3. Good health ,Oncology ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Medicine ,Membranes and Sorting ,Antibody ,Cancer Screening ,Research Article ,medicine.medical_specialty ,Colon ,Protein Array Analysis ,Gastroenterology and Hepatology ,Biology ,Flow cytometry ,03 medical and health sciences ,Antigen ,Cell Line, Tumor ,Gastrointestinal Tumors ,Cell Adhesion ,Cancer Detection and Diagnosis ,Early Detection ,Biomarkers, Tumor ,medicine ,Humans ,Antigens ,030304 developmental biology ,lcsh:R ,Cancers and Neoplasms ,Computational Biology ,medicine.disease ,High-Throughput Screening Assays ,Cancer cell ,Cancer research ,biology.protein ,lcsh:Q ,Cytometry - Abstract
Colon cancer is a deadly disease affecting millions of people worldwide. Current treatment challenges include management of disease burden as well as improvements in detection and targeting of tumor cells. To identify disease state-specific surface antigen signatures, we combined fluorescent cell barcoding with high-throughput flow cytometric profiling of primary and metastatic colon cancer lines (SW480, SW620, and HCT116). Our multiplexed technique offers improvements over conventional methods by permitting the simultaneous and rapid screening of cancer cells with reduced effort and cost. The method uses a protein-level analysis with commercially available antibodies on live cells with intact epitopes to detect potential tumor-specific targets that can be further investigated for their clinical utility. Multiplexed antibody arrays can easily be applied to other tumor types or pathologies for discovery-based approaches to target identification.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.