hosted by
publicationslist.org
    

Walter J Jessen

Biomarker Center of Excellence
Covance
671 S. Meridian
Greenfield, IN 46140
walter@walterjessen.com
Walter Jessen is a computational biologist with Covance's Biomarker Center of Excellence and is focused on biomarker discovery and prioritization. Walter is also a knowledge curator at Biomarker Commons, which aggregates the latest biomarker news and research focused on biomarker discovery, development and use: http://biomarkercommons.org

Journal articles

2012
Vincent W Keng, Eric P Rahrmann, Adrienne L Watson, Barbara R Tschida, Christopher L Moertel, Walter J Jessen, Tilat A Rizvi, Margaret H Collins, Nancy Ratner, David A Largaespada (2012)  PTEN and NF1 Inactivation in Schwann Cells Produces a Severe Phenotype in the Peripheral Nervous System That Promotes the Development and Malignant Progression of Peripheral Nerve Sheath Tumors.   Cancer Res 72: 13. 3405-3413 Jul  
Abstract: The genetic evolution from a benign neurofibroma to a malignant sarcoma in patients with neurofibromatosis type 1 (NF1) syndrome remains unclear. Schwann cells and/or their precursor cells are believed to be the primary pathogenic cell in neurofibromas because they harbor biallelic neurofibromin 1 (NF1) gene mutations. However, the phosphatase and tensin homolog (Pten) and neurofibromatosis 1 (Nf1) genes recently were found to be comutated in high-grade peripheral nerve sheath tumors (PNST) in mice. In this study, we created transgenic mice that lack both Pten and Nf1 in Schwann cells and Schwann cell precursor cells to validate the role of these two genes in PNST formation in vivo. Haploinsufficiency or complete loss of Pten dramatically accelerated neurofibroma development and led to the development of higher grade PNSTs in the context of Nf1 loss. Pten dosage, together with Nf1 loss, was sufficient for the progression from low-grade to high-grade PNSTs. Genetic analysis of human malignant PNSTs (MPNST) also revealed downregulation of PTEN expression, suggesting that Pten-regulated pathways are major tumor-suppressive barriers to neurofibroma progression. Together, our findings establish a novel mouse model that can rapidly recapitulate the onset of human neurofibroma tumorigenesis and the progression to MPNSTs. Cancer Res; 72(13); 3405-13. ©2012 AACR.
Notes:
Ami V Patel, David Eaves, Walter J Jessen, Tilat A Rizvi, Jeffrey A Ecsedy, Mark G Qian, Bruce Aronow, John Perentesis, Eduard Serra, Timothy Cripe, Shyra C Miller, Nancy Ratner (2012)  Ras-Driven Transcriptome Analysis Identifies Aurora Kinase A as a Potential Malignant Peripheral Nerve Sheath Tumor Therapeutic Target.   Clin Cancer Res Jul  
Abstract: PURPOSE: Patients with Neurofibromatosis Type 1 (NF1) develop malignant peripheral nerve sheath tumors (MPNST) which are often inoperable and do not respond well to current chemotherapies or radiation. The goal of this study was to utilize comprehensive gene expression analysis to identify novel therapeutic targets.EXPERIMENTAL DESIGN: Nerve Schwann cells and/or their precursors are the tumorigenic cell types in MPNST due to the loss of the NF1 gene, which encodes the RasGAP protein neurofibromin. Therefore, we created a transgenic mouse model, CNP-HRas12V, expressing constitutively-active HRas in Schwann cells and defined a Ras-induced gene expression signature to drive a Bayesian factor regression model analysis of differentially expressed genes in mouse and human neurofibromas and MPNSTs. We tested functional significance of Aurora kinase over-expression in MPNST in vitro and in vivo using Aurora kinase shRNAs and compounds that inhibit Aurora kinase. RESULTS: We identified 2000 genes with probability of linkage to nerve Ras signaling of which 339 were significantly differentially expressed in mouse and human NF1-related tumor samples relative to normal nerves, including Aurora kinase A (AURKA). AURKA was dramatically over-expressed and genomically amplified in MPNSTs but not neurofibromas. Aurora kinase shRNAs and Aurora kinase inhibitors blocked MPNST cell growth in vitro. Furthermore, an AURKA selective inhibitor, MLN8237, stabilized tumor volume and significantly increased survival of mice with MPNST xenografts. CONCLUSION: Integrative cross-species transcriptome analyses combined with preclinical testing has provided an effective method for identifying candidates for molecular-targeted therapeutics. Blocking Aurora kinases may be a viable treatment platform for MPNST.
Notes:
2011
Clay E S Comstock, Michael A Augello, Matthew J Schiewer, Jason Karch, Craig J Burd, Adam Ertel, Erik S Knudsen, Walter J Jessen, Bruce J Aronow, Karen E Knudsen (2011)  Cyclin D1 is a selective modifier of androgen-dependent signaling and androgen receptor function.   J Biol Chem 286: 10. 8117-8127 Mar  
Abstract: D-type cyclins regulate cellular outcomes in part through cyclin-dependent, kinase-independent mechanisms that modify transcription factor action, and recent in vivo studies showed that cyclin D1 associates with a large number of transcriptional regulators in cells of the retina and breast. Given the frequency of cyclin D1 alterations in cancer, it is imperative to delineate the molecular mechanisms by which cyclin D1 controls key transcription factor networks in human disease. Prostate cancer was used as a paradigm because this tumor type is reliant at all stages of the disease on androgen receptor (AR) signaling, and cyclin D1 has been shown to negatively modulate AR-dependent expression of prostate-specific antigen (KLK3/PSA). Strategies were employed to control cyclin D1 expression under conditions of hormone depletion, and the effect of cyclin D1 on subsequent androgen-dependent gene expression was determined using unbiased gene expression profiling. Modulating cyclin D1 conferred widespread effects on androgen signaling and revealed cyclin D1 to be a selective effector of hormone action. A subset of androgen-induced target genes, known to be directly regulated by AR, was strongly suppressed by cyclin D1. Analyses of AR occupancy at target gene regulatory loci of clinical relevance demonstrated that cyclin D1 limits AR residence after hormone stimulation. Together, these findings reveal a new function for cyclin D1 in controlling hormone-dependent transcriptional outcomes and demonstrate a pervasive role for cyclin D1 in regulating transcription factor dynamics.
Notes:
2010
T R Hummel, W J Jessen, S J Miller, L Kluwe, V F Mautner, M R Wallace, C Lázaro, G P Page, P F Worley, B J Aronow, E K Schorry, N Ratner (2010)  Gene expression analysis identifies potential biomarkers of neurofibromatosis type 1 including adrenomedullin.   Clin Cancer Res. 16: 20. 5048-57 Aug  
Abstract: PURPOSE: Plexiform neurofibromas (pNF) are Schwann cell tumors found in a third of individuals with neurofibromatosis type 1 (NF1). pNF can undergo transformation to malignant peripheral nerve sheath tumors (MPNST). There are no identified serum biomarkers of pNF tumor burden or transformation to MPNST. Serum biomarkers would be useful to verify NF1 diagnosis, monitor tumor burden, and/or detect transformation. EXPERIMENTAL DESIGN: We used microarray gene expression analysis to define 92 genes that encode putative secreted proteins in neurofibroma Schwann cells, neurofibromas, and MPNST. We validated differential expression by quantitative reverse transcription-PCR, Western blotting, and ELISA assays in cell conditioned medium and control and NF1 patient sera. RESULTS: Of 13 candidate genes evaluated, only adrenomedullin (ADM) was confirmed as differentially expressed and elevated in serum of NF1 patients. ADM protein concentrati on was further elevated in serum of a small sampling of NF1 patients with MPNST. MPNST cell conditioned medium, containing ADM and hepatocyte growth factor, stimulated MPNST migration and endothelial cell proliferation. CONCLUSIONS: Thus, microarray analysis identifies potential serum biomarkers for disease, and ADM is a serum biomarker of NF1. ADM serum levels do not seem to correlate with the presence of pNFs but may be a biomarker of transformation to MPNST.
Notes:
2009
S J Miller*, W J Jessen*, T Mehta, A Hardiman, E Sites, S Kaiser, A Jegga, H Li, M Upadhyaya, M Giovannini, D Muir, M R Wallace, E Lopez, E Serra, G P Nielsen, C Lazaro, A Stemmer-Rachamimov, G Page, B J Aronow, N Ratner (2009)  Integrative genomic analyses of neurofibromatosis tumours identify SOX9 as a biomarker and survival gene.   EMBO Mol Med 1: 4. 236-248 Jul  
Abstract: Understanding the biological pathways critical for common neurofibromatosis type 1 (NF1) peripheral nerve tumours is essential, as there is a lack of tumour biomarkers, prognostic factors and therapeutics. We used gene expression profiling to define transcriptional changes between primary normal Schwann cells (n = 10), NF1-derived primary benign neurofibroma Schwann cells (NFSCs) (n = 22), malignant peripheral nerve sheath tumour (MPNST) cell lines (n = 13), benign neurofibromas (NF) (n = 26) and MPNST (n = 6). Dermal and plexiform NFs were indistinguishable. A prominent theme in the analysis was aberrant differentiation. NFs repressed gene programs normally active in Schwann cell precursors and immature Schwann cells. MPNST signatures strongly differed; genes up-regulated in sarcomas were significantly enriched for genes activated in neural crest cells. We validated the differential expression of 82 genes including the neural crest transcription factor SOX9 and SOX9 predicted targets. SOX9 immunoreactivity was robust in NF and MPSNT tissue sections and targeting SOX9 - strongly expressed in NF1-related tumours - caused MPNST cell death. SOX9 is a biomarker of NF and MPNST, and possibly a therapeutic target in NF1.
Notes:
2008
Hui Shen, Nathan Powers, Nitin Saini, Clay E S Comstock, Ankur Sharma, Katherine Weaver, Monica P Revelo, William Gerald, Erin Williams, Walter J Jessen, Bruce J Aronow, Gary Rosson, Bernard Weissman, Christian Muchardt, Moshe Yaniv, Karen E Knudsen (2008)  The SWI/SNF ATPase Brm is a gatekeeper of proliferative control in prostate cancer.   Cancer Res 68: 24. 10154-10162 Dec  
Abstract: Factors that drive prostate cancer progression remain poorly defined, thus hindering the development of new therapeutic strategies. Disseminated tumors are treated through regimens that ablate androgen signaling, as prostate cancer cells require androgen for growth and survival. However, recurrent, incurable tumors that have bypassed the androgen requirement ultimately arise. This study reveals that the Brm ATPase, a component of selected SWI/SNF complexes, has significant antiproliferative functions in the prostate that protect against these transitions. First, we show that targeted ablation of Brm is causative for the development of prostatic hyperplasia in mice. Second, in vivo challenge revealed that Brm-/- epithelia acquire the capacity for lobe-specific, castration-resistant cellular proliferation. Third, investigation of human specimens revealed that Brm mRNA and protein levels are attenuated in prostate cancer. Fourth, Brm down-regulation was associated with an increased proliferative index, consistent with the mouse model. Lastly, gene expression profiling showed that Brm loss alters factors upstream of E2F1; this was confirmed in murine models, wherein Brm loss induced E2F1 deregulation in a tissue-specific manner. Combined, these data identify Brm as a major effector of serum androgen-induced proliferation in the prostate that is disrupted in human disease, and indicate that loss of Brm confers a proliferative advantage in prostate cancer.
Notes:
Xuesong Ouyang, Walter J Jessen, Hikmat Al-Ahmadie, Angel M Serio, Yong Lin, Weichung-Joseph Shih, Victor E Reuter, Peter T Scardino, Michael M Shen, Bruce J Aronow, Andrew J Vickers, William L Gerald, Cory Abate-Shen (2008)  Activator protein-1 transcription factors are associated with progression and recurrence of prostate cancer.   Cancer Res 68: 7. 2132-2144 Apr  
Abstract: To identify biomarkers that discriminate the aggressive forms of prostate cancer, we performed gene expression profiling of prostate tumors using a genetically engineered mouse model that recapitulates the stages of human prostate cancer, namely Nkx3.1; Pten mutant mice. We observed a significant deregulation of the epidermal growth factor and mitogen-activated protein kinase (MAPK) signaling pathways, as well as their major downstream effectors--the activator protein-1 transcription factors c-Fos and c-Jun. Forced expression of c-Fos and c-Jun in prostate cancer cells promotes tumorigenicity and results in activation of extracellular signal-regulated kinase (Erk) MAPK signaling. In human prostate cancer, up-regulation of c-Fos and c-Jun proteins occurs in advanced disease and is correlated with Erk MAPK pathway activation, whereas high levels of c-Jun expression are associated with disease recurrence. Our analyses reveal a hitherto unappreciated role for AP-1 transcription factors in prostate cancer progression and identify c-Jun as a marker of high-risk prostate cancer. This study provides a striking example of how accurate mouse models can provide insights on molecular processes involved in progression and recurrence of human cancer.
Notes:
2007
Sergio Kaiser, Young-Kyu Park, Jeffrey L Franklin, Richard B Halberg, Ming Yu, Walter J Jessen, Johannes Freudenberg, Xiaodi Chen, Kevin Haigis, Anil G Jegga, Sue Kong, Bhuvaneswari Sakthivel, Huan Xu, Timothy Reichling, Mohammad Azhar, Gregory P Boivin, Reade B Roberts, Anika C Bissahoyo, Fausto Gonzales, Greg C Bloom, Steven Eschrich, Scott L Carter, Jeremy E Aronow, John Kleimeyer, Michael Kleimeyer, Vivek Ramaswamy, Stephen H Settle, Braden Boone, Shawn Levy, Jonathan M Graff, Thomas Doetschman, Joanna Groden, William F Dove, David W Threadgill, Timothy J Yeatman, Robert J Coffey, Bruce J Aronow (2007)  Transcriptional recapitulation and subversion of embryonic colon development by mouse colon tumor models and human colon cancer.   Genome Biol 8: 7.  
Abstract: BACKGROUND: The expression of carcino-embryonic antigen by colorectal cancer is an example of oncogenic activation of embryonic gene expression. Hypothesizing that oncogenesis-recapitulating-ontogenesis may represent a broad programmatic commitment, we compared gene expression patterns of human colorectal cancers (CRCs) and mouse colon tumor models to those of mouse colon development embryonic days 13.5-18.5. RESULTS: We report here that 39 colon tumors from four independent mouse models and 100 human CRCs encompassing all clinical stages shared a striking recapitulation of embryonic colon gene expression. Compared to normal adult colon, all mouse and human tumors over-expressed a large cluster of genes highly enriched for functional association to the control of cell cycle progression, proliferation, and migration, including those encoding MYC, AKT2, PLK1 and SPARC. Mouse tumors positive for nuclear beta-catenin shifted the shared embryonic pattern to that of early development. Human and mouse tumors differed from normal embryonic colon by their loss of expression modules enriched for tumor suppressors (EDNRB, HSPE, KIT and LSP1). Human CRC adenocarcinomas lost an additional suppressor module (IGFBP4, MAP4K1, PDGFRA, STAB1 and WNT4). Many human tumor samples also gained expression of a coordinately regulated module associated with advanced malignancy (ABCC1, FOXO3A, LIF, PIK3R1, PRNP, TNC, TIMP3 and VEGF). CONCLUSION: Cross-species, developmental, and multi-model gene expression patterning comparisons provide an integrated and versatile framework for definition of transcriptional programs associated with oncogenesis. This approach also provides a general method for identifying pattern-specific biomarkers and therapeutic targets. This delineation and categorization of developmental and non-developmental activator and suppressor gene modules can thus facilitate the formulation of sophisticated hypotheses to evaluate potential synergistic effects of targeting within- and between-modules for next-generation combinatorial therapeutics and improved mouse models.
Notes:
Whitney Banach-Petrosky, Walter J Jessen, Xuesong Ouyang, Hui Gao, Jayashree Rao, John Quinn, Bruce J Aronow, Cory Abate-Shen (2007)  Prolonged exposure to reduced levels of androgen accelerates prostate cancer progression in Nkx3.1; Pten mutant mice.   Cancer Res 67: 19. 9089-9096 Oct  
Abstract: In this report, we have investigated the relationship between androgen levels and prostate tumorigenesis in Nkx3.1; Pten mutant mice, a genetically engineered mouse model of human prostate cancer. By experimentally manipulating serum levels of testosterone in these mice for an extended period (i.e., 7 months), we have found that prolonged exposure of Nkx3.1; Pten mutant mice to androgen levels that are 10-fold lower than normal (the "Low-T" group) resulted in a marked acceleration of prostate tumorigenesis compared with those exposed to androgen levels within the reference range (the "Normal-T" group). We found that prostate tumors from the Low-T mutant mice share a similar gene expression profile as androgen-independent prostate tumors from these mutant mice, which includes the deregulated expression of several genes that are up-regulated in human hormone-refractory prostate cancer, such as Vav3 and Runx1. We propose that exposure to reduced androgens may promote prostate tumorigenesis by selecting for molecular events that promote more aggressive, hormone-refractory tumors.
Notes:
Xiangdong Liu, Walter J Jessen, Siva Sivaganesan, Bruce J Aronow, Mario Medvedovic (2007)  Bayesian hierarchical model for transcriptional module discovery by jointly modeling gene expression and ChIP-chip data.   BMC Bioinformatics 8: 08  
Abstract: BACKGROUND: Transcriptional modules (TM) consist of groups of co-regulated genes and transcription factors (TF) regulating their expression. Two high-throughput (HT) experimental technologies, gene expression microarrays and Chromatin Immuno-Precipitation on Chip (ChIP-chip), are capable of producing data informative about expression regulatory mechanism on a genome scale. The optimal approach to joint modeling of data generated by these two complementary biological assays, with the goal of identifying and characterizing TMs, is an important open problem in computational biomedicine. RESULTS: We developed and validated a novel probabilistic model and related computational procedure for identifying TMs by jointly modeling gene expression and ChIP-chip binding data. We demonstrate an improved functional coherence of the TMs produced by the new method when compared to either analyzing expression or ChIP-chip data separately or to alternative approaches for joint analysis. We also demonstrate the ability of the new algorithm to identify novel regulatory relationships not revealed by ChIP-chip data alone. The new computational procedure can be used in more or less the same way as one would use simple hierarchical clustering without performing any special transformation of data prior to the analysis. The R and C-source code for implementing our algorithm is incorporated within the R package gimmR which is freely available at http://eh3.uc.edu/gimm. CONCLUSION: Our results indicate that, whenever available, ChIP-chip and expression data should be analyzed within the unified probabilistic modeling framework, which will likely result in improved clusters of co-regulated genes and improved ability to detect meaningful regulatory relationships. Given the good statistical properties and the ease of use, the new computational procedure offers a worthy new tool for reconstructing transcriptional regulatory networks.
Notes:
2006
Walter J Jessen, Scott A Hoose, Jessica A Kilgore, Michael P Kladde (2006)  Active PHO5 chromatin encompasses variable numbers of nucleosomes at individual promoters.   Nat Struct Mol Biol 13: 3. 256-263 Mar  
Abstract: Transcriptional activation is often associated with chromatin remodeling. However, little is known about the dynamics of remodeling of nucleosome arrays in vivo. Upon induction of Saccharomyces cerevisiae PHO5, a novel kinetic assay of DNA methyltransferase accessibility showed that nucleosomes adjacent to the histone-free upstream activating sequence (UASp1) are disrupted earlier and at higher frequency in the cell population than are those more distal. Individually cloned molecules, each representing the chromatin state of a full promoter from a single cell, revealed multiple promoter classes with either no remodeling or variable numbers of disrupted nucleosomes. Individual promoters in the remodeled fraction were highly enriched for contiguous blocks of disrupted nucleosomes, the majority of which overlapped the UAS region. These results support a probabilistic model in which chromatin remodeling at PHO5 spreads from sites of transactivator association with DNA and attenuates with distance.
Notes:
2004
Walter J Jessen, Archana Dhasarathy, Scott A Hoose, Christopher D Carvin, April L Risinger, Michael P Kladde (2004)  Mapping chromatin structure in vivo using DNA methyltransferases.   Methods 33: 1. 68-80 May  
Abstract: Cytosine-5 DNA methyltransferases (C5 DMTases) are effective reagents for analyzing chromatin and footprinting DNA-bound factors in vivo. Cytosine methylation in accessible regions is assayed positively by the PCR-based technique of bisulfite sequencing. In this article, we outline two complementary uses for the DNA methyltransferase CviPI (M.CviPI, GC specificity) in probing chromatin organization. First, we describe the use of the naturally occurring, free enzyme as a diffusible probe to map changes in nucleosome structure and to footprint factor interactions at cis-regulatory sequences. In a second application, termed targeted gene methylation (TAGM), the DMTase is targeted via in-frame fusion to a DNA-binding factor. The rapid accumulation of DNA methylation enables highly sensitive detection of factor binding. Both strategies can be applied with any C5 DMTase, such as M.SssI, which also possesses a short-recognition specificity (CG). A description of methods for constructing C5 DMTase-expressing strains of Saccharomyces cerevisiae and analyzing chromatin regions is provided. We also include comprehensive protocols for the isolation and bisulfite treatment of genomic DNA as well as the subsequent bisulfite sequencing steps. Data demonstrating the efficacy of both DMTase probing techniques, theoretical considerations, and experimental analyses are presented at GAL1 and PHO5.
Notes:
2003
Christopher D Carvin, Archana Dhasarathy, Laurie B Friesenhahn, Walter J Jessen, Michael P Kladde (2003)  Targeted cytosine methylation for in vivo detection of protein-DNA interactions.   Proc Natl Acad Sci U S A 100: 13. 7743-7748 Jun  
Abstract: We report a technique, named targeted gene methylation (TAGM), for identifying in vivo protein-binding sites in chromatin. M.CviPI, a cytosine-5 DNA methyltransferase recognizing GC sites, is fused to a DNA-binding factor enabling simultaneous detection of targeted methylation, factor footprints, and chromatin structural changes by bisulfite genomic sequencing. Using TAGM with the yeast transactivator Pho4, methylation enrichments of up to 34- fold occur proximal to native Pho4-binding sites. Additionally, significant selective targeting of methylation is observed several hundred nucleotides away, suggesting the detection of long-range interactions due to higher-order chromatin structure. In contrast, at an extragenic locus lacking Pho4-binding sites, methylation levels are at the detection limit at early times after Pho4 transactivation. Notably, substantial amounts of methylation are targeted by Pho4-M.CviPI under repressive conditions when most of the transactivator is excluded from the nucleus. Thus, TAGM enables rapid detection of DNA-protein interactions even at low occupancies and has potential for identifying factor targets at the genome-wide level. Extension of TAGM from yeast to vertebrates, which use methylation to initiate and propagate repressed chromatin, could also provide a valuable strategy for heritable inactivation of gene expression.
Notes:

Book chapters

2007
C E S Comstock, C J Burd, W J Jessen, K E Knudsen (2007)  Gene profiling analysis of androgen receptor mediated function.   In: Contemporary Endocrinology: Genomics in Endocrinology, DNA Microarray Analysis in Endocrine Health and Disease. Edited by:S Handwerger and B Aronow. 83-113 Humana Press Inc.  
Abstract: Androgens, particularly testosterone and its potent metabolite 5-α-dihydrotestosterone, serve as critical mediators in the development and maintenance of the male reproductive and non-reproductive systems. Androgen-dependent signaling is conveyed by the androgen receptor (AR), which is a member of the nuclear receptor superfamily. AR binding of androgen stimulates its ability to bind DNA and regulate gene transcription. The importance of the AR in human physiology is exemplified by the fact that disruption of this key receptor is causative for androgen insensitivity syndrome (AIS) and spinal and bulbar muscular atrophy (SBMA), also known as Kennedy’s disease. In contrast, AR activity is essential for benign prostatic hyperplasia (BPH) and the development and progression of prostate cancer. The advent of microarray technology has provided significant contributions toward the understanding of the underlying mechanisms that govern the AR function in these physiological and pathophysiological conditions. The aim of this chapter is to summarize the recent advances using genomics to study the role of androgens in developmental and reproductive processes, AIS, Kennedy’s disease, and prostate cancer. As the body of work relative to prostate cancer is large, a gene list comparison was performed with prostate cancer studies that utilized a well-characterized human prostatic adenocarcinoma cell line treated with physiological concentrations of androgen. The consensus targets were then evaluated on their physiological relevance as well as on their potential as direct AR targets. In addition, alternate models of AR action in prostate cancer were examined. These studies provide evidence for AR-dependent regulation of other identified genes involved in metabolism, transcription, and signaling pathways. Together, these collective observations reveal insightful information concerning androgen action in human health and disease.
Notes:
2006
J A Kilgore, S A Hoose, A Dhasarathy, D W Neef, W J Jessen, M P Kladde (2006)  Dynamic chromatin remodeling at the budding yeast PHO5 promoter.   In: DNA Structure, Chromatin and Gene Expression. Edited by:R Kiyama, M. Shimizu. 101-120 Transworld Research Network  
Abstract: Promoter transactivation is often associated with the disruption or remodeling of nucleosomes in chromatin. The budding yeast PHO5 promoter, which drives expression of the major secreted acid phosphatase, is a proven model system for investigating gene regulatory mechanisms in the context of chromatin. Under conditions of high external phosphate concentration, PHO5 transcription is repressed through an upstream signaling cascade and an array of nucleosomes that are well positioned and hinder access to crucial regulatory elements. Much recent progress has been made in elucidating the molecular steps that lead to promoter activation upon phosphate starvation. Inhibition of an upstream repressive signaling cascade enables DNA binding by site-specific activator proteins, which, in turn, lead to the direct recruitment of two histone modifying and two ATP-dependent chromatin remodeling complexes. Using a novel single-molecule assay for chromatin remodeling in vivo, we have shown that the number of nucleosomes disrupted from cell-to-cell is highly variable. Moreover, nucleosomes adjacent to activator binding sites are disrupted earlier and at higher frequency than are those more distal. The results suggest that the localized recruitment of chromatin modifying and remodeling complexes leads to nucleosome disruption that spreads from sites of transactivator association with DNA and attenuates with distance.
Notes:
Powered by PublicationsList.org.