180 results on '"Hess, Jay L."'
Search Results
152. The same site on the integrase-binding domain of lens epithelium-derived growth factor is a therapeutic target for MLL leukemia and HIV.
- Author
-
Murai, Marcelo J., Pollock, Jonathan, Shihan He, Hongzhi Miao, Purohit, Trupta, Yokom, Adam, Hess, Jay L., Muntean, Andrew G., Grembecka, Jolanta, and Cierpicki, Tomasz
- Subjects
- *
INTEGRASE inhibitors , *MOLECULAR structure of chromatin , *LEUKEMIA treatment , *HIV integrase inhibitors , *EPITHELIAL tumors - Abstract
Lens epithelium-derived growth factor (LEDGF) is a chromatin-associated protein implicated in leukemia and HIV type 1 infection. LEDGF associates with mixed-lineage leukemia (MLL) fusion proteins and menin and is required for leukemic transformation. To better understand the molecular mechanism underlying the LEDGF integrase-binding domain (IBD) interaction with MLL fusion proteins in leukemia, we determined the solution structure of the MLL-IBD complex. We found a novel MLL motif, integrase domain binding motif 2 (IBM2), which binds to a well-defined site on IBD. Point mutations within IBM2 abolished leukemogenic transformation by MLL-AF9, validating that this newly identified motif is essential for the oncogenic activity of MLL fusion proteins. Interestingly, the IBM2 binding site on IBD overlaps with the binding site for the HIV integrase (IN), and IN was capable of efficiently sequestering IBD from the menin-MLL complex. A short IBM2 peptide binds to IBD directly and inhibits both the IBD-MLL/menin and IBD-IN interactions. Our findings show that the same site on IBD is involved in binding to MLL and HIV-IN, revealing an attractive approach to simultaneously target LEDGF in leukemia and HIV. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
153. C/EBPα is an essential collaborator in Hoxa9/Meis1 -mediated leukemogenesis.
- Author
-
Collins, Cailin, Jingya Wang, Hongzhi Miao, Bronstein, Joel, Nawer, Humaira, Tao Xu, Figueroa, Maria, Muntean, Andrew G., and Hess, Jay L.
- Subjects
- *
LEUKEMIA etiology , *HOMEOBOX genes , *TRANSCRIPTION factors , *HEMATOPOIETIC stem cells , *LABORATORY mice - Abstract
Homeobox A9 (HOXA9) is a homeodomain-containing transcription factor that plays a key role in hematopoietic stem cell expansion and is commonly deregulated in human acute leukemias. A variety of upstream genetic alterations in acute myeloid leukemia (AML) lead to overexpression of HOXA9, almost always in association with overexpression of its cofactor meis homeobox 1 (MEIS1). A wide range of data suggests that HOXA9 and MEIS1 play a synergistic causative role in AML, although the molecular mechanisms leading to transformation by HOXA9 and MEIS1 remain elusive. In this study, we identify CCAAT/enhancer binding protein alpha (C/EBPα) as a critical collaborator required for Hoxa9/Meis1-mediated leukemogenesis. We show that C/EBPα is required for the proliferation of Hoxa9/Meis1 -transformed cells in culture and that loss of C/EBPα greatly improves survival in both primary and secondary murine models of Hoxa9/Meis1-induced leukemia. Over 50% of Hoxa9 genome-wide binding sites are cobound by C/EBPα, which coregulates a number of downstream target genes involved in the regulation of cell proliferation and differentiation. Finally, we show that Hoxa9 represses the locus of the cyclin-dependent kinase inhibitors Cdkn2afo in concert with C/EBPα to overcome a block in G1 cell cycle progression. Together, our results suggest a previously unidentified role for C/EBPα in maintaining the proliferation required for Hoxa9/Meis1-mediated leukemogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
154. Targeting Recruitment of Disruptor of Telomeric Silencing 1-like (DOT1L).
- Author
-
Chenxi Shen, Jo, Stephanie Y., Chenzhong Liao, Hess, Jay L., and Nikolovska-Coleska, Zaneta
- Subjects
- *
TELOMERES , *GENE silencing , *AMINO acids , *LEUKEMIA , *PEPTIDES - Abstract
The MLL fusion proteins, AF9 and ENL, activate target genes in part via recruitment of the histone methyltransf erase DOT1L (disruptor of telomeric silencing 1-like). Here we report biochemical, biophysical, and functional characterization of the interaction between DOT1L and MLL fusion proteins, AF9/ ENL. The AF9/ENL-binding site in human DOT1L was mapped, and the interaction site was identified to a 10-amino acid region (DOT1L865-874). This region is highly conserved in DOT1L from a variety of species. Alanine scanning mutagenesis analysis shows that four conserved hydrophobic residues from the identified binding motif are essential for the interactions with AF9/ ENL. Binding studies demonstrate that the entire intact C-terminal domain of AF9/ENL is required for optimal interaction with DOT1L. Functional studies show that the mapped AF9/ ENL interacting site is essential for immortalization by MLL-AF9, indicating that DOT1L interaction with MLL-AF9 and its recruitment are required for transformation by MLL-AF9. These results strongly suggest that disruption of interaction between DOT1L and AF9/ENL is a promising therapeutic strategy with potentially fewer adverse effects than enzymatic inhibition of DOT1L for MLL fusion protein-associated leukemia. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
155. A Subset of Mixed Lineage Leukemia Proteins Has Plant Homeodomain (PHD)-mediated E3 Ligase Activity.
- Author
-
Jingya Wang, Muntean, Andrew G., Wu, Laura, and Hess, Jay L.
- Subjects
- *
LEUKEMIA , *UBIQUITIN ligases , *GENETIC mutation , *PROTEINS , *GENES , *ENZYMES - Abstract
The mixed lineage leukemia protein MLL1 contains four highly conserved plant homeodomain (PHD) fingers, which are invariably deleted in oncogenic MLL1 fusion proteins in human leukemia. Here we show that the second PHD finger (PHD2) of MLL1 is an E3 ubiquitin ligase in the presence of the E2-conjugating enzyme CDC34. This activity is conserved in the second PHD finger of MLL4, the closest homolog to MLL1 but not in MLL2 or MLL3. Mutation of PHD2 leads to MLL1 stabilization, as well as increased transactivation ability and MLL1 recruitment to the target gene loci, suggesting that PHD2 negatively regulates MLL1 activity. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
156. Identification and characterization of Hoxa9 binding sites in hematopoietic cells.
- Author
-
Yongsheng Huang, Sitwala, Kajal, Bronstein, Joel, Sanders, Daniel, Dandekar, Monisha, Collins, Cailin, Robertson, Gordon, MacDonald, James, Cezard, Timothee, Bilenky, Misha, Thiessen, Nina, Yongjun Zhao, Zeng, Thomas, Hirst, Martin, Hero, Alfred, Jones, Steven, and Hess, Jay L.
- Subjects
- *
HEMATOPOIETIC stem cells , *BINDING sites , *GENETIC transcription , *PROTO-oncogenes , *GENE transfection , *GENE enhancers - Abstract
The clustered homeobox proteins play crucial roles in development, hematopoiesis and leukemia yet the targets they regulate and their mechanisms of action are poorly understood. Here, we identified the binding sites for Hoxa9 and the Hox cofactor Meis1 on a genome-wide level and profiled their associated epigenetic modifications and transcriptional targets. Hoxa9 and the Hox cofactor Meis1 co-bind at hundreds of highly evolutionarily-conserved sites, most of which are distant from transcription start sites. These sites show high levels of histone H3K4 monomethylation and CBP/P300 binding characteristic of enhancers. Furthermore, a subset of these sites shows enhancer activity in transient transfection assays. Many Hoxa9 and Meis1 binding sites are also bound by PU.1 and other lineage-restricted transcription factors previously implicated in establishment of myeloid enhancers. Conditional Hoxa9 activation is associated with CBP/P300 recruitment, histone acetylation and transcriptional activation of a network of proto-oncogenes including Erg, Flt3, Lmo2, Myb and Sox4. Collectively this work suggests that Hoxa9 regulates transcription by interacting with enhancers of genes important for hematopoiesis and leukemia. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
157. CBX8, a Polycomb Group Protein, Is Essential for MLL-AF9-Induced Leukemogenesis
- Author
-
Tan, Jiaying, Jones, Morgan, Koseki, Haruhiko, Nakayama, Manabu, Muntean, Andrew G., Maillard, Ivan, and Hess, Jay L.
- Subjects
- *
POLYCOMB group proteins , *LEUKEMIA etiology , *GENETIC transcription regulation , *GENETIC mutation , *ABLATION techniques - Abstract
Summary: Chromosomal translocations involving the mixed lineage leukemia (MLL) gene lead to the development of acute leukemias. Constitutive HOX gene activation by MLL fusion proteins is required for MLL-mediated leukemogenesis; however, the underlying mechanisms remain elusive. Here, we show that chromobox homolog 8 (CBX8), a Polycomb Group protein that interacts with MLL-AF9 and TIP60, is required for MLL-AF9-induced transcriptional activation and leukemogenesis. Conversely, both CBX8 ablation and specific disruption of the CBX8 interaction by point mutations in MLL-AF9 abrogate HOX gene upregulation and abolish MLL-AF9 leukemic transformation. Surprisingly, Cbx8-deficient mice are viable and display no apparent hematopoietic defects. Together, our findings demonstrate that CBX8 plays an essential role in MLL-AF9 transcriptional regulation and leukemogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
158. The PAF Complex Synergizes with MLL Fusion Proteins at HOX Loci to Promote Leukemogenesis
- Author
-
Muntean, Andrew G., Tan, Jiaying, Sitwala, Kajal, Huang, Yongsheng, Bronstein, Joel, Connelly, James A., Basrur, Venkatesha, Elenitoba-Johnson, Kojo S.J., and Hess, Jay L.
- Subjects
- *
LEUKEMIA etiology , *TUMOR proteins , *GENE fusion , *CHROMOSOMES , *GENE rearrangement , *RNA polymerases , *GENE expression , *PROMOTERS (Genetics) - Abstract
Summary: MLL is involved in chromosomal rearrangements that generate fusion proteins with deregulated transcriptional activity. The mechanisms of MLL fusion protein-mediated transcriptional activation are poorly understood. Here we show MLL interacts directly with the polymerase associated factor complex (PAFc) through sequences flanking the CxxC domain. PAFc interacts with RNA polymerase II and stimulates posttranslational histone modifications. PAFc augments MLL and MLL-AF9 mediated transcriptional activation of Hoxa9. Conversely, knockdown of PAFc disrupts MLL fusion protein-mediated transcriptional activation and MLL recruitment to target loci. PAFc gene expression is downregulated during hematopoiesis and likely serves to regulate MLL function. Deletions of MLL that abolish interactions with PAFc also eliminate MLL-AF9 mediated immortalization indicating an essential function for this interaction in leukemogenesis. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
159. The eleven-nineteen-leukemia protein ENL connects nuclear MLL fusion partners with chromatin.
- Author
-
Zeisig, Deniz T., Bittner, Claudia B., Zeisig, Bernd B., García-Cuéllar, Maria-Paz, Hess, Jay L., and Slany, Robert K.
- Subjects
- *
LEUKEMIA , *PROTEINS , *CHROMATIN , *NUCLEOPROTEINS , *CANCER - Abstract
Mixed lineage leukemia (MLL) fusion proteins are derived from translocations at 11q23 that occur in aggressive subtypes of leukemia. As a consequence, MLL is joined to different unrelated proteins to form oncogenic transcription factors. Here we demonstrate a direct interaction between several nuclear MLL fusion partners and present evidence for a role of these proteins in histone binding. In two-hybrid studies, ENL interacted with AF4 and AF5q31 as well as with a fragment of AF10. A structure–function analysis revealed that the AF4/AF5q31/AF10 binding domain in ENL coincided with the C-terminus that is essential for transformation by MLL-ENL. The ENL/AF4 association was corroborated by GST-pulldown experiments and by mutual coprecipitation. Both proteins colocalized in vivo in a nuclear speckled pattern. Moreover, AF4 and ENL coeluted on sizing columns together with the known ENL binding partner Polycomb3, suggesting the presence of a multiprotein complex. The overexpression of ENL alone activated a reporter construct and a mutational screen indicated the conserved YEATS domain as essential for this function. Overlay and pulldown-assays finally showed a specific and YEATS domain-dependent association of ENL with histones H3 and H1. In summary, our studies support a common role for nuclear MLL fusion partners in chromatin biology.Oncogene (2005) 24, 5525–5532. doi:10.1038/sj.onc.1208699; published online 25 April 2005 [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
160. Physical Association and Coordinate Function of the H3 K4 Methyltransferase MLL1 and the H4 K16 Acetyltransferase MOF.
- Author
-
Vail Dou, Mime, Thomas A., Tackett, Alan J., Smith, Edwin R., Fukuda, Aya, Wysocka, Joanna, Allis, C. David, Chait, Brian T., Hess, Jay L., and Roeder, Robert G.
- Subjects
- *
CELL culture , *METHYLATION , *CELL lines , *ZINC , *HEREDITY , *ALKYLATION - Abstract
A stable complex containing MLL1 and MOF has been immunoaffinity purified from a human cell line that stably expresses an epitope-tagged WDR5 subunit. Stable interactions between MLL1 and MOF were confirmed by reciprocal immunoprecipitation, cosedimentation, and cotransfection analyses, and interaction sites were mapped to MLLI C-terminal and MOF zinc finger domains. The purified complex has a robust MLL1-mediated histone methyltransferase activity that can effect mono-, di-, and trimethylation of H3 K4 and a MOF-mediated histone acetyltransferase activity that is specific for H4 K16. Importantly, both activities are required for optimal transcription activation on a chromatin template in vitro and on an endogenous MLL1 target gene, Hox a9, in vivo. These results indicate an activator-based mechanism for joint MLL1 and MOF recruitment and targeted methylation and acetylation and provide a molecular explanation for the closely correlated distribution of H3 K4 methylation and H4 K16 acetylation on active genes. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
161. Conditional MLL-CBP targets GMP and models therapy-related myeloproliferative disease.
- Author
-
Wang, Jing, Iwasaki, Hiromi, Krivtsov, Andrei, Febbo, Phillip G., Thorner, Aaron R., Ernst, Patricia, Anastasiadou, Ema, Kutok, Jeffery L., Kogan, Scott C., Zinkel, Sandra S., Fisher, Jill K., Hess, Jay L., Golub, Todd R., Armstrong, Scott A., Akashi, Koichi, and Korsmeyer, Stanley J.
- Subjects
- *
BONE marrow diseases , *LEUKEMIA , *ANEMIA , *THERAPEUTICS , *GENE expression , *GENETIC regulation - Abstract
Chromosomal translocations that fuse the mixed lineage leukemia (MLL) gene with multiple partners typify acute leukemias of infancy as well as therapy-related leukemias. We utilized a conditional knockin strategy to bypass the embryonic lethality caused by MLL-CBP expression and to assess the immediate effects of induced MLL-CBP expression on hematopoiesis. Within days of activating MLL-CBP, the fusion protein selectively expanded granulocyte/macrophage progenitors (GMP) and enhanced their self-renewal/proliferation. MLL-CBP altered the gene expression program of GMP, upregulating a subset of genes including Hox a9. Inhibition of Hox a9 expression by RNA interference demonstrated that MLL-CBP required Hox a9 for its enhanced cell expansion. Following exposure to sublethal?-irradiation or N-ethyl-N-nitrosourea (ENU), MLL-CBP mice developed myelomonocytic hyperplasia and progressed to fatal myeloproliferative disorders. These represented the spectrum of therapy-induced acute myelomonocytic leukemia/chronic myelomonocytic leukemia/myelodysplastic/myeloproliferative disorder similar to that seen in humans possessing the t(11;16). This model of MLL-CBP therapy-related myeloproliferative disease demonstrates the selectivity of this MLL fusion for GMP cells and its ability to initiate leukemogenesis in conjunction with cooperating mutations. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
162. Dimerization of MLL fusion proteins immortalizes hematopoietic cells
- Author
-
Martin, Mary Ellen, Milne, Thomas A., Bloyer, Sebastien, Galoian, Karine, Shen, Weiping, Gibbs, Denise, Brock, Hugh W., Slany, Robert, and Hess, Jay L.
- Subjects
- *
PROTEINS , *BONE marrow , *MYELOID metaplasia - Abstract
MLL fusion proteins are leukemogenic, but their mechanism is unclear. Induced dimerization of a truncated MLL immortalizes bone marrow and imposes a reversible block on myeloid differentiation associated with upregulation of Hox a7, a9, and Meis1. Both dimerized MLL and exon-duplicated MLL are potent transcriptional activators, suggesting a link between dimerization and partial tandem duplication of DNA binding domains of MLL. Dimerized MLL binds with higher affinity than undimerized MLL to a CpG island within the Hox a9 locus. However, MLL-AF9 is not dimerized in vivo. The data support a model in which either MLL dimerization/exon duplication or fusion to a transcriptional activator results in Hox gene upregulation and ultimately transformation. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
163. CONTRIBUTORS
- Author
-
Abkowitz, Janis L., Abrams, Charles S., Abrey, Lauren E., Ajioka, Richard S., Alexandrescu, Doru T., Alter, Harvey J., Alvandi, Firoozeh, Araten, David J., Arceci, Robert J., Armstrong, Elina, Bain, Barbara J., Barrett, John, Behringer, Karolin, Bennett, Joel S., Berger, Nathan A., Berliner, Nancy, Beutler, Ernest, Bloomfield, Clara D., Bolton-Maggs, Paula H.B., Bontempo, Franklin A., Bredenfeld, Henning, Brittenham, Gary M., Brown, Kevin E., Brown, Margaret, Burks, Eric J., Bussel, James B., Byrd, John C., Caligiuri, Michael A., Carlson, Katrin M., Carter, Melody C., Cawley, J.C., Cazzola, Mario, Choi, John K., Cines, Douglas B., Cohen, Jeffrey I., Comenzo, Raymond L., Cook, James R., Cooper, Megan A., Crowther, Mark A., Cunningham, Melody J., Dale, David C., Dave, Sandeep S., Leval, Laurence de, Diehl, Volker, Döhner, Hartmut, Döhner, Konstanze, Dokal, Inderjeet, Dunbar, Cynthia E., Dutcher, Janice P., Edelson, Richard, El-Shami, Khaled, Endy, Timothy P., Esteban, Juan I., Estey, Elihu H., Falanga, Anna, Felix, Carolyn A., Ferrara, James L.M., Ferreri, Andrés J.M., Ferri, Clodoveo, Fleisher, Thomas A., Fox, Lindy P., Francis, Charles W., Gaines, Peter, Gallagher, Patrick G., Ganser, Arnold, George, James N., Gewirtz, Alan M., Gewirtz, Amy S., Goldman, John M., Gregg, Xylina T., Grever, Michael R., Hajjar, Katherine A., Harrington, William J., Jr., Harris, Nancy Lee, Heit, John A., Hess, Jay L., Heuser, Michael, Horne, III, McDonald K., Hostetter, Richard B., Hsu, Lewis L., Invernizzi, Rosangela, Jin, David K., Johnson, Carol L., Joyce, Robin M., Kalayanarooj, Siripen, Kaplan, Karen L., Kazura, James W., Keel, Siobán, King, Karen E., King, May-Jean, Klein, Harvey G., Klimm, Beate, Kobbervig, Catie E., Koç, Omer N., Konkle, Barbara A., Kruskall, Margot S., Kushner, James P., Lazarus, Hillard M., Le Beau, Michelle M., Lee, Agnes Y.Y., Levi, Marcel, Lichtin, Alan, Little, Richard F., Liu, Johnson M., Loren, Alison W., LoRusso, Karen L., Loughran, Thomas P., Jr., Luppi, Mario, Luzzatto, Lucio, Ma, Alice D., Maciejewski, Jaroslaw P., Macik, B. Gail, Malech, Harry L., Mattei, Peter, Max, Edward E., McVey, John H., Means, Robert T., Jr., Metcalfe, Dean D., Middeldorp, Saskia, Mikhael, Joseph, Mohandas, Narla, Mrózek, Krzysztof, Myint, Khin Saw Aye, Ness, Paul M., Neufeld, Ellis J., Oliveira, João B., Owen, Roger G., Palmblad, Jan E.W., Parmar, Simrit, Perez, Kimberly, Petersdorf, Effie W., Peterson, LoAnn C., Pileri, Stefano A., Poncz, Mortimer, Powell, Jonathan, Prchal, Josef T., Pui, Ching-Hon, Ragni, Margaret V., Rao, V. Koneti, Reid, Marion E., Remick, Scot C., Remuzzi, Giuseppe, Ribeiro, Raul C., Rick, Margaret E., Rodgers, Griffin P., Rosenblum, Daniel, Rouault, Tracey, Roubey, Robert A.S., Russell-Jones, Robin, Sadler, J. Evan, Saint-Remy, Jean-Marie, Sallah, Sabah, Sassa, Shigeru, Schechter, Geraldine P., Schecter, Alan N., Schieppati, Arrigo, Schnipper, Lowell, Schwartzentruber, Douglas J., Scott, J. Paul, Seligman, Paul A., Siegel, Robert S., Sloand, Elaine M., Spivak, Jerry L., Stabler, Sally P., Stadtmauer, Edward A., Staudt, Louis M., Stetler-Stevenson, Maryalice, Stewart, A. Keith, Straus, Stephen E., Tallman, Martin S., Tefferi, Ayalew, Thompson, Arthur R., Tisdale, John F., Toh, Cheng Hock, Torelli, Giuseppe, Tosato, Giovanna, Tzachanis, Dimitrios, Uhl, Lynne, Valdez, Riccardo, Warkentin, Theodore E., Watanabe, Jill M., Watzke, Herbert H., Wayne, Alan S., Weiss, Mitchell J., Whittaker, Sean, Wiernik, Peter H., Wilson, Wyndham H., Wright, Daniel G., Yanik, Gregory, Yarchoan, Robert, Young, Neal S., Zheng, X. Long, Zieger, Barbara, and Zignego, Anna Linda
- Full Text
- View/download PDF
164. Medical School Without Walls: 50 Years of Regional Campuses at Indiana University School of Medicine.
- Author
-
Wallach PM, Birnbaum DR, Ryan ER, Pieczko BT, and Hess JL
- Subjects
- Humans, Schools, Medical, Universities, Indiana, Medicine, Education, Medical
- Abstract
The history of Indiana University School of Medicine (IUSM) dates to 1871, when Indiana Medical College entered into an affiliation with Indiana University in Bloomington to offer medical education. In 1971, the Indiana General Assembly passed a bill to create and fund a distributed model for medical education for which IUSM was responsible, an innovative approach to implementing a statewide medical education program. IUSM became one of the first U.S. medical schools to implement what is today known as a regional medical campus model. This regional medical campus system has permitted IUSM to expand enrollment based on national and local concerns about physician shortages, increase access to care locally, support expansion of graduate medical education, and provide opportunities for research and scholarship by faculty and students statewide. This effort was made possible by partnerships with other universities and health care systems across the state and the support of local community and state leaders. The model is a forward-thinking and cost-effective way to educate physicians for service in the state of Indiana and is applicable to others. This article highlights milestones in IUSM's 50-year history of regional medical education, describes the development of the regional medical campus model, recognizes significant achievements over the years, shares lessons learned, and discusses considerations for the future of medical education., (Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the Association of American Medical Colleges.)
- Published
- 2022
- Full Text
- View/download PDF
165. Erratum for Caslini et al., "MLL Associates with Telomeres and Regulates Telomeric Repeat-Containing RNA Transcription".
- Author
-
Caslini C, Connelly JA, Serna A, Broccoli D, and Hess JL
- Published
- 2021
- Full Text
- View/download PDF
166. Twenty-first century pathology sign-out.
- Author
-
Tomlins S, Robinson D, Penny RJ, and Hess JL
- Subjects
- Humans, Molecular Diagnostic Techniques methods, Pathology, Clinical methods, Precision Medicine methods
- Abstract
It is difficult to imagine a field that is changing as rapidly as pathology. A convergence of factors including not only scientific and technological advances but also changes in business models is transforming the field, particularly in the area of cancer diagnostics. The authors examine 8 themes, or "forces of change," in pathology and speculate on how these will affect pathology sign-out and the future role of pathologists in patient care., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
167. ECSASB2 mediates MLL degradation during hematopoietic differentiation.
- Author
-
Wang J, Muntean AG, and Hess JL
- Subjects
- Cell Differentiation genetics, Cells, Cultured, Cullin Proteins chemistry, Cullin Proteins genetics, Cullin Proteins metabolism, Cullin Proteins physiology, Elongin, HEK293 Cells, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells physiology, Histone-Lysine N-Methyltransferase, Humans, K562 Cells, Leukemia etiology, Leukemia genetics, Leukemia metabolism, Multiprotein Complexes genetics, Multiprotein Complexes metabolism, Multiprotein Complexes physiology, Myeloid-Lymphoid Leukemia Protein chemistry, Myeloid-Lymphoid Leukemia Protein genetics, Protein Processing, Post-Translational genetics, Protein Processing, Post-Translational physiology, SKP Cullin F-Box Protein Ligases chemistry, SKP Cullin F-Box Protein Ligases genetics, SKP Cullin F-Box Protein Ligases metabolism, Suppressor of Cytokine Signaling Proteins chemistry, Suppressor of Cytokine Signaling Proteins genetics, Suppressor of Cytokine Signaling Proteins metabolism, Transcription Factors chemistry, Transcription Factors genetics, Transcription Factors metabolism, Transcription Factors physiology, Transfection, Hematopoiesis genetics, Hematopoiesis physiology, Myeloid-Lymphoid Leukemia Protein metabolism, Proteolysis, SKP Cullin F-Box Protein Ligases physiology, Suppressor of Cytokine Signaling Proteins physiology
- Abstract
Mixed lineage leukemia (MLL) is a key epigenetic regulator of normal hematopoietic development and chromosomal translocations involving MLL are one of the most common genetic alterations in human leukemia. Here we show that ASB2, a component of the ECS(ASB) E3 ubiquitin ligase complex, mediates MLL degradation through interaction with the PHD/Bromodomain region of MLL. Forced expression of ASB2 degrades MLL and reduces MLL transactivation activity. In contrast, the MLL-AF9 fusion protein does not interact with ASB2 and is resistant to ASB2 mediated degradation. Increased expression of ASB2 during hematopoietic differentiation is associated with decreased levels of MLL protein and down-regulation of MLL target genes. Knockdown of ASB2 leads to increased expression of HOXA9 and delayed cell differentiation. Our data support a model whereby ASB2 contributes to hematopoietic differentiation, in part, through MLL degradation and HOX gene down-regulation. Moreover, deletion of the PHD/Bromo region renders MLL fusion proteins resistant to ASB2-mediated degradation and may contribute to leukemogenesis.
- Published
- 2012
- Full Text
- View/download PDF
168. Identification and characterization of Hoxa9 binding sites in hematopoietic cells.
- Author
-
Huang Y, Sitwala K, Bronstein J, Sanders D, Dandekar M, Collins C, Robertson G, MacDonald J, Cezard T, Bilenky M, Thiessen N, Zhao Y, Zeng T, Hirst M, Hero A, Jones S, and Hess JL
- Subjects
- Acetylation, Animals, Binding Sites, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Blotting, Western, Bone Marrow Cells metabolism, Chromatin Immunoprecipitation, Enhancer Elements, Genetic, Epigenomics, Female, Gene Expression Profiling, Leukemia metabolism, Mice, Mice, Inbred C57BL, Myeloid Ecotropic Viral Integration Site 1 Protein, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Oligonucleotide Array Sequence Analysis, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Transcription Factors genetics, Transcription Factors metabolism, Gene Expression Regulation, Leukemic, Hematopoiesis physiology, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Leukemia genetics
- Abstract
The clustered homeobox proteins play crucial roles in development, hematopoiesis, and leukemia, yet the targets they regulate and their mechanisms of action are poorly understood. Here, we identified the binding sites for Hoxa9 and the Hox cofactor Meis1 on a genome-wide level and profiled their associated epigenetic modifications and transcriptional targets. Hoxa9 and the Hox cofactor Meis1 cobind at hundreds of highly evolutionarily conserved sites, most of which are distant from transcription start sites. These sites show high levels of histone H3K4 monomethylation and CBP/P300 binding characteristic of enhancers. Furthermore, a subset of these sites shows enhancer activity in transient transfection assays. Many Hoxa9 and Meis1 binding sites are also bound by PU.1 and other lineage-restricted transcription factors previously implicated in establishment of myeloid enhancers. Conditional Hoxa9 activation is associated with CBP/P300 recruitment, histone acetylation, and transcriptional activation of a network of proto-oncogenes, including Erg, Flt3, Lmo2, Myb, and Sox4. Collectively, this work suggests that Hoxa9 regulates transcription by interacting with enhancers of genes important for hematopoiesis and leukemia.
- Published
- 2012
- Full Text
- View/download PDF
169. Requirement for Dot1l in murine postnatal hematopoiesis and leukemogenesis by MLL translocation.
- Author
-
Jo SY, Granowicz EM, Maillard I, Thomas D, and Hess JL
- Subjects
- Animals, Base Sequence, Cell Cycle, DNA Primers genetics, Gene Expression, Gene Knockout Techniques, Hematopoiesis genetics, Hematopoietic Stem Cells pathology, Histone Methyltransferases, Histone-Lysine N-Methyltransferase metabolism, Humans, Leukemia, Experimental genetics, Leukemia, Experimental metabolism, Leukemia, Experimental pathology, Male, Methyltransferases deficiency, Methyltransferases genetics, Mice, Mice, Knockout, Mice, Transgenic, Oncogene Proteins, Fusion genetics, Oncogenes, Pancytopenia etiology, Hematopoiesis physiology, Leukemia, Experimental etiology, Methyltransferases physiology, Myeloid-Lymphoid Leukemia Protein genetics, Translocation, Genetic
- Abstract
Disruptor of telomeric silencing 1-like (Dot1l) is a histone 3 lysine 79 methyltransferase. Studies of constitutive Dot1l knockout mice show that Dot1l is essential for embryonic development and prenatal hematopoiesis. DOT1L also interacts with translocation partners of Mixed Lineage Leukemia (MLL) gene, which is commonly translocated in human leukemia. However, the requirement of Dot1l in postnatal hematopoiesis and leukemogenesis of MLL translocation proteins has not been conclusively shown. With a conditional Dot1l knockout mouse model, we examined the consequences of Dot1l loss in postnatal hematopoiesis and MLL translocation leukemia. Deletion of Dot1l led to pancytopenia and failure of hematopoietic homeostasis, and Dot1l-deficient cells minimally reconstituted recipient bone marrow in competitive transplantation experiments. In addition, MLL-AF9 cells required Dot1l for oncogenic transformation, whereas cells with other leukemic oncogenes, such as Hoxa9/Meis1 and E2A-HLF, did not. These findings illustrate a crucial role of Dot1l in normal hematopoiesis and leukemogenesis of specific oncogenes.
- Published
- 2011
- Full Text
- View/download PDF
170. Loss-of-function Additional sex combs like 1 mutations disrupt hematopoiesis but do not cause severe myelodysplasia or leukemia.
- Author
-
Fisher CL, Pineault N, Brookes C, Helgason CD, Ohta H, Bodner C, Hess JL, Humphries RK, and Brock HW
- Subjects
- Animals, B-Lymphocytes cytology, B-Lymphocytes metabolism, Cell Count, Cell Lineage, Cells, Cultured, Flow Cytometry, Gene Targeting, Hematopoietic Stem Cells metabolism, Mice, Mice, Mutant Strains, Myeloid Cells pathology, Splenomegaly pathology, T-Lymphocytes cytology, T-Lymphocytes metabolism, Thymus Gland cytology, Hematopoiesis genetics, Leukemia genetics, Mutation genetics, Myelodysplastic Syndromes genetics, Repressor Proteins genetics
- Abstract
The Additional sex combs like 1 (Asxl1) gene is 1 of 3 mammalian homologs of the Additional sex combs (Asx) gene of Drosophila. Asx is unusual because it is required to maintain both activation and silencing of Hox genes in flies and mice. Asxl proteins are characterized by an amino terminal homology domain, by interaction domains for nuclear receptors, and by a C-terminal plant homeodomain protein-protein interaction domain. A recent study of patients with myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) revealed a high incidence of truncation mutations that would delete the PHD domain of ASXL1. Here, we show that Asxl1 is expressed in all hematopoietic cell fractions analyzed. Asxl1 knockout mice exhibit defects in frequency of differentiation of lymphoid and myeloid progenitors, but not in multipotent progenitors. We do not detect effects on hematopoietic stem cells, or in peripheral blood. Notably, we do not detect severe myelodysplastic phenotypes or leukemia in this loss-of-function model. We conclude that Asxl1 is needed for normal hematopoiesis. The mild phenotypes observed may be because other Asxl genes have redundant function with Asxl1, or alternatively, MDS or oncogenic phenotypes may result from gain-of-function Asxl mutations caused by genomic amplification, gene fusion, or truncation of Asxl1.
- Published
- 2010
- Full Text
- View/download PDF
171. The role of menin in hematopoiesis.
- Author
-
Maillard I and Hess JL
- Subjects
- Animals, Histone-Lysine N-Methyltransferase, Humans, Leukemia physiopathology, Multiprotein Complexes metabolism, Myeloid-Lymphoid Leukemia Protein genetics, Myeloid-Lymphoid Leukemia Protein metabolism, Proto-Oncogene Proteins genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Hematopoiesis physiology, Proto-Oncogene Proteins metabolism
- Abstract
In the hematopoietic system, menin was found to interact with MLL, a large protein encoded by the mixed linage leukemia gene that acts as a histone H3 methyltransferase. The MLL gene is a recurrent target for translocations in both acute myeloid and acute lymphoid leukemias. MLL gene rearrangements involve a variety of translocation partners, giving rise to MLL fusion proteins whose transforming ability is mediated through upregulated expression of Homeobox (Hox) genes as well as other targets. Recent work indicates that menin is an essential partner of MLL fusion proteins in leukemic cells and that it regulates normal hematopoiesis. In the absence of menin, steady-state hematopoiesis is largely preserved; however, menin-deficient hematopoietic stem cells are markedly deficient in situations of hematopoietic stress, such as during recovery after bone marrow transplantation. In leukemias driven by MLL fusion proteins, menin is essential for transformation and growth of the malignant cells. Thus, menin-MLL interactions represent a promising therapeutic target in leukemias with MLL rearrangements.
- Published
- 2009
- Full Text
- View/download PDF
172. Chromatin immunoprecipitation (ChIP) for analysis of histone modifications and chromatin-associated proteins.
- Author
-
Milne TA, Zhao K, and Hess JL
- Subjects
- Cross-Linking Reagents pharmacology, DNA Primers, Epigenesis, Genetic, Humans, Sequence Analysis, DNA, Chromatin genetics, Chromatin Immunoprecipitation methods, DNA-Binding Proteins analysis, DNA-Binding Proteins metabolism, Histones metabolism, Polymerase Chain Reaction methods
- Abstract
Disruption of epigenetic regulators of transcription is a central mechanism of oncogenesis. Many of the advances in the understanding of these mechanisms are attributable to the successful development of chromatin immunoprecipitation (ChIP) for in vivo detection of histone modifications as well as chromatin binding regulatory proteins. This is a powerful technique for analyzing histone modifications as well as binding sites for proteins that bind either directly or indirectly to DNA. Here we present two ChIP protocols. The first is particularly useful for identifying histone modifications or binding at specific, known genomic sites. The second, employing serial analysis of gene expression, is particularly powerful for the discovery of previously unidentified sites of modification or binding.
- Published
- 2009
- Full Text
- View/download PDF
173. MLL-AF9 leukemia stem cells: hardwired or taking cues from the microenvironment?
- Author
-
Muntean AG and Hess JL
- Subjects
- Animals, Apoptosis, Bone Marrow metabolism, Cell Culture Techniques, Cell Line, Transformed, Cell Proliferation, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Environment, Fetal Stem Cells immunology, Fetal Stem Cells metabolism, Gene Expression Regulation, Leukemic, Genotype, Humans, Intercellular Signaling Peptides and Proteins metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Multipotent Stem Cells immunology, Multipotent Stem Cells metabolism, Myeloid-Lymphoid Leukemia Protein genetics, Neoplastic Stem Cells immunology, Neoplastic Stem Cells metabolism, Oncogene Proteins, Fusion genetics, Phenotype, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Signal Transduction, Time Factors, rac GTP-Binding Proteins metabolism, Cell Lineage, Cell Transformation, Neoplastic pathology, Fetal Stem Cells pathology, Leukemia, Myeloid, Acute pathology, Multipotent Stem Cells pathology, Myeloid-Lymphoid Leukemia Protein metabolism, Neoplastic Stem Cells pathology, Oncogene Proteins, Fusion metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology
- Abstract
MLL rearrangements in humans lead to both acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL). While AML has been successfully produced in mice, modeling ALL has been more difficult. In this issue of Cancer Cell, Wei et al. (2008) describe generation of AML, ALL, and biphenotypic leukemia by manipulating the cytokine milieu of human progenitor cells expressing MLL-AF9. They demonstrate that both multipotent and lineage-restricted progenitors are targeted by MLL-AF9 fusion proteins and that Rac signaling is crucial for survival. This study demonstrates the heterogeneity of MLL-AF9 leukemic stem cells and the importance of the microenvironment in determining lineage outcome.
- Published
- 2008
- Full Text
- View/download PDF
174. HOX proteins and leukemia.
- Author
-
Sitwala KV, Dandekar MN, and Hess JL
- Abstract
HOX and three amino acid loop extension (TALE) proteins cooperate to induce transformation in mouse leukemia models, and are dysregulated in a variety of human leukemias. Despite decades of research, the mechanism of action for Hox proteins in embryogenesis and hematopoiesis remains unclear. Recent studies on the roles of Hoxa9 and Meis1 in leukemia has led to a wealth of new data, but their molecular mechanisms of action and synergy remain obscure. Advances in genome-wide technologies offer new avenues for understanding how homeodomain-containing transcription factors exert their programs in normal and neoplastic development.
- Published
- 2008
175. Mechanisms of transcriptional regulation by MLL and its disruption in acute leukemia.
- Author
-
Dou Y and Hess JL
- Subjects
- Animals, DNA Methylation, Disease Models, Animal, Histone-Lysine N-Methyltransferase, Homeodomain Proteins genetics, Homeodomain Proteins physiology, Humans, Mice, Mice, Knockout, Myeloid Ecotropic Viral Integration Site 1 Protein, Myeloid-Lymphoid Leukemia Protein physiology, Neoplasm Proteins genetics, Neoplasm Proteins physiology, Leukemia genetics, Leukopoiesis genetics, Myeloid-Lymphoid Leukemia Protein genetics, Transcription, Genetic genetics
- Abstract
Fusion of the mixed lineage leukemia protein (MLL) to one of over 50 different translocation partners converts it into a potent leukemogenic oncoprotein. The resulting fusion proteins transform primarily through upregulation of A-cluster Hox genes, including Hoxa9 and the Hox cofactor Meis1. Considerable progress has been made in delineating the differences between normal Hox gene regulation by MLL and deregulated transcription in MLL-induced leukemias. Some MLL translocation partners dimerize the truncated MLL molecule. Other translocation partners appear to recruit nuclear coactivator complexes that have diverse enzymatic activities that impinge on transcriptional elongation pathways. These enzymatic activities, including RNA polymerase II phosphorylation as well as histone H3 lysine 79 methylation present attractive targets for the development of future MLL-directed therapy.
- Published
- 2008
- Full Text
- View/download PDF
176. The tumor suppressor menin regulates hematopoiesis and myeloid transformation by influencing Hox gene expression.
- Author
-
Chen YX, Yan J, Keeshan K, Tubbs AT, Wang H, Silva A, Brown EJ, Hess JL, Pear WS, and Hua X
- Subjects
- Animals, Blotting, Western, Cell Line, Transformed, Cell Proliferation, Chromatin Immunoprecipitation, DNA metabolism, DNA Methylation, DNA-Binding Proteins metabolism, Exons, Flow Cytometry, Genotype, Histones chemistry, Homeodomain Proteins metabolism, Homozygote, Leukemia metabolism, Lysine chemistry, Methylation, Mice, Mice, Knockout, Mice, Transgenic, Models, Genetic, Models, Statistical, Myeloid Cells metabolism, Proto-Oncogene Proteins metabolism, Retroviridae genetics, Reverse Transcriptase Polymerase Chain Reaction, Stem Cells, Time Factors, Transgenes, Gene Expression Regulation, Hematopoiesis, Homeodomain Proteins physiology, Proto-Oncogene Proteins physiology
- Abstract
Menin is the product of the tumor suppressor gene Men1 that is mutated in the inherited tumor syndrome multiple endocrine neoplasia type 1 (MEN1). Menin has been shown to interact with SET-1 domain-containing histone 3 lysine 4 (H3K4) methyltransferases including mixed lineage leukemia proteins to regulate homeobox (Hox) gene expression in vitro. Using conditional Men1 knockout mice, we have investigated the requirement for menin in hematopoiesis and myeloid transformation. Men1 excision causes reduction of Hoxa9 expression, colony formation by hematopoietic progenitors, and the peripheral white blood cell count. Menin directly activates Hoxa9 expression, at least in part, by binding to the Hoxa9 locus, facilitating methylation of H3K4, and recruiting the methylated H3K4 binding protein chd1 to the locus. Consistent with signaling downstream of menin, ectopic expression of both Hoxa9 and Meis1 rescues colony formation defects in Men1-excised bone marrow. Moreover, Men1 excision also suppresses proliferation of leukemogenic mixed lineage leukemia-AF9 fusion-protein-transformed myeloid cells and Hoxa9 expression. These studies uncover an important role for menin in both normal hematopoiesis and myeloid transformation and provide a mechanistic understanding of menin's function in these processes that may be used for therapy.
- Published
- 2006
- Full Text
- View/download PDF
177. MLL associates specifically with a subset of transcriptionally active target genes.
- Author
-
Milne TA, Dou Y, Martin ME, Brock HW, Roeder RG, and Hess JL
- Subjects
- Animals, Cell Line, Chromatin Immunoprecipitation, Glutathione Transferase, Mice, Mice, Inbred C57BL, Polymerase Chain Reaction, Gene Expression Regulation, Genes, Homeobox genetics, Myeloid-Lymphoid Leukemia Protein metabolism, RNA Polymerase II metabolism, Transcription, Genetic genetics
- Abstract
MLL (mixed-lineage leukemia) is a histone H3 Lys-4 specific methyltransferase that is a positive regulator of Hox expression. MLL rearrangements and amplification are common in acute lymphoid and myeloid leukemias and myelodysplastic disorders and are associated with abnormal up-regulation of Hox gene expression. Although MLL is expressed throughout hematopoiesis, Hox gene expression is sharply down-regulated during differentiation, suggesting that either the activity of MLL or its association with target promoters must be regulated. Here we show that MLL associates with actively transcribed genes but does not remain bound after transcriptional down-regulation. Surprisingly, MLL is associated not only with promoter regions but also is distributed across the entire coding regions of genes. MLL interacts with RNA polymerase II (pol II) and colocalizes with RNA pol II at a subset of actively transcribed target in vivo. Loss of function Mll results in defects in RNA pol II distribution. Together the results suggest that an intimate association between MLL and RNA pol II occurs at MLL target genes in vivo that is required for normal initiation and/or transcriptional elongation.
- Published
- 2005
- Full Text
- View/download PDF
178. Menin and MLL cooperatively regulate expression of cyclin-dependent kinase inhibitors.
- Author
-
Milne TA, Hughes CM, Lloyd R, Yang Z, Rozenblatt-Rosen O, Dou Y, Schnepp RW, Krankel C, Livolsi VA, Gibbs D, Hua X, Roeder RG, Meyerson M, and Hess JL
- Subjects
- Carrier Proteins genetics, Cell Cycle Proteins genetics, Cell Line, Cell Proliferation, Cyclin-Dependent Kinase Inhibitor p18, Cyclin-Dependent Kinase Inhibitor p27, DNA-Binding Proteins genetics, Histone-Lysine N-Methyltransferase, Humans, Intracellular Signaling Peptides and Proteins genetics, Myeloid-Lymphoid Leukemia Protein, Open Reading Frames, Promoter Regions, Genetic, Proto-Oncogenes genetics, Transcription Factors genetics, Transcriptional Activation, Transfection, Tumor Suppressor Proteins genetics, Cyclin-Dependent Kinases antagonists & inhibitors, DNA-Binding Proteins physiology, Gene Expression Regulation, Proto-Oncogene Proteins physiology, Proto-Oncogenes physiology, Transcription Factors physiology
- Abstract
Mutations in the MEN1 gene are associated with the multiple endocrine neoplasia syndrome type 1 (MEN1), which is characterized by parathyroid hyperplasia and tumors of the pituitary and pancreatic islets. The mechanism by which MEN1 acts as a tumor suppressor is unclear. We have recently shown that menin, the MEN1 protein product, interacts with mixed lineage leukemia (MLL) family proteins in a histone methyltransferase complex including Ash2, Rbbp5, and WDR5. Here, we show that menin directly regulates expression of the cyclin-dependent kinase inhibitors p27Kip1 and p18Ink4c. Menin activates transcription by means of a mechanism involving recruitment of MLL to the p27Kip1 and p18Ink4c promoters and coding regions. Loss of function of either MLL or menin results in down-regulation of p27Kip1 and p18Ink4c expression and deregulated cell growth. These findings suggest that regulation of cyclin-dependent kinase inhibitor transcription by cooperative interaction between menin and MLL plays a central role in menin's activity as a tumor suppressor.
- Published
- 2005
- Full Text
- View/download PDF
179. MLL: a histone methyltransferase disrupted in leukemia.
- Author
-
Hess JL
- Subjects
- Gene Amplification, Gene Duplication, Histone Methyltransferases, Humans, Models, Genetic, Myeloid-Lymphoid Leukemia Protein, Protein Methyltransferases, Recombinant Fusion Proteins genetics, Transcriptional Activation, DNA-Binding Proteins genetics, Histone-Lysine N-Methyltransferase genetics, Leukemia genetics, Proto-Oncogenes genetics, Transcription Factors genetics
- Abstract
Rearrangements of the MLL gene, which is located at chromosome 11q23, are associated with aggressive acute leukemias in both children and adults. MLL regulates Hox gene expression through direct promoter binding and histone modification. MLL rearrangements occurring in leukemia include MLL fusion genes, partial tandem duplications of MLL and MLL amplification. MLL fusions and amplification upregulate Hox expression, apparently resulting in a block of hematopoietic differentiation. Future therapies for MLL-associated leukemia might involve blocking Hox gene upregulation by using fusion proteins or inhibiting the activity of Hox proteins themselves.
- Published
- 2004
- Full Text
- View/download PDF
180. Effects of the proteasome inhibitor PS-341 on tumor growth in HTLV-1 Tax transgenic mice and Tax tumor transplants.
- Author
-
Mitra-Kaushik S, Harding JC, Hess JL, and Ratner L
- Subjects
- Animals, Bortezomib, Cell Division drug effects, Crosses, Genetic, Cysteine Endopeptidases, DNA Replication drug effects, HTLV-I Infections immunology, Humans, Interleukin-10 metabolism, Interleukin-6 metabolism, Leukemia, T-Cell immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Proteasome Endopeptidase Complex, Antineoplastic Agents pharmacology, Boronic Acids pharmacology, Genes, pX, HTLV-I Infections pathology, Leukemia, T-Cell pathology, Multienzyme Complexes antagonists & inhibitors, Protease Inhibitors pharmacology, Pyrazines pharmacology
- Abstract
Recent studies have shown that the transcription factor nuclear factor kappaB (NF-kappaB) regulates critical survival pathways in a variety of cancers, including human T-cell leukemia/lymphotrophic virus 1 (HTLV-1)-transformed CD4 T cells. The activation of NF-kappaB is controlled by proteasome-mediated degradation of the inhibitor of nuclear factor kappaBalpha (IkappaBalpha). We investigated the effects of PS-341, a peptide boronate inhibitor of the proteasome in HTLV-1 Tax transgenic tumors in vitro and in vivo. In Tax transgenic mice, PS-341 administered thrice weekly inhibited tumor-associated NF-kappaB activity. Quantitation of proliferation, apoptosis, and interleukin 6 (IL-6) and IL-10 secretion by tumor cells in culture revealed that the effects of PS-341 on cell growth largely correlated with inhibition of pathways mediated by NF-kappaB. However, the effect of PS-341 on the growth of tumors in Tax transgenic mice revealed heterogeneity in drug responsiveness. The tumor tissues treated with PS-341 show no consistent inhibition of NFkappaB activation in vivo. Annexin V staining indicated that PS-341 response in vivo correlated with sensitivity to apoptosis induced by gamma irradiation. On the other hand, transplanted Tax tumors in Rag-1 mice showed consistent inhibition of tumor growth and prolonged survival in response to the same drug regimen. TUNEL staining indicated that PS-341 treatment sensitizes Tax tumors to DNA fragmentation.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.