Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Patricia E. Berg is active.

Publication


Featured researches published by Patricia E. Berg.


Molecular and Cellular Biology | 2004

Molecular and Cellular Determinants of Estrogen Receptor α Expression

Joseph J. Pinzone; Holly Stevenson; Jeannine S. Strobl; Patricia E. Berg

Critical roles for estrogens in growth and development and in pathological conditions of bone, breast, and uterus are well established. Estrogens and estrogen receptor modulators bind to estrogen receptor α (ERα) and/or ERβ to form discrete molecular complexes that exert pleiotropic tissue-specific effects by modulating the expression of target genes. Ligand-bound ER functions as a key transcription factor in various molecular pathways, and modulation of ER expression levels is important in determining cellular growth potential. Recent advances have begun to illuminate the mechanisms by which cells control ER expression. Kos et al. reviewed the genomic organization of the human ERα gene promoter region (31). The human ERα gene, located on chromosome 6, was cloned in 1986 and spans approximately 300 kb; its eight coding regions are transcribed from at least seven promoters (31). Considering the complexity of the ER promoter, it is not surprising that numerous factors affect ERα expression. The aim of this paper is to review the molecular mechanisms by which various cellular factors modulate ERα expression. We have focused on highlighting the major players, including effectors of chromatin structure, hormones, and other relevant agents, and limited our discussion to findings in human systems. There are vast qualitative and quantitative data regarding whether or not, and to what extent, ERα is expressed in normal and neoplastic tissues; this topic is beyond the scope of this review and will not be covered. Moreover, little is currently known about the role of specific transcription factors in ER expression, and these factors will be mentioned only briefly. Henceforth, the terms ERα and ER will be used interchangeably; expression of ERβ will not be discussed.


Leukemia | 2000

BP1, a new homeobox gene, is frequently expressed in acute leukemias

Susanne B. Haga; Sidney W. Fu; Judith E. Karp; D. D. Ross; Donna M. Williams; W. D. Hankins; F. Behm; F. W. Ruscetti; Myron Chang; B D Smith; D. Becton; S. C. Raimondi; Patricia E. Berg

Aberrant expression of homeobox genes has been described in primary leukemia blasts. We recently cloned a new cDNA, BP1, which is a member of the homeobox gene family. BP1 expression was investigated in bone marrow samples from acute myeloid leukemia (AML), acute T cell lymphocytic leukemia (ALL) and pre-B cell ALL. Expression levels of two apparent isoforms of BP1, DLX7 and DLX4, were measured in the same samples. They are weakly if at all detectable in normal bone marrow, PHA-stimulated T cells or B cells. BP1 RNA was highly expressed in 63% of AML cases, including 81% of the pediatric and 47% of the adult cases, and in 32% of T-ALL cases, but was not found in any of the pre-B ALL cases. Co-expression of BP1, DLX7 and DLX4 occurred in a significant number of leukemias. Our data, including co-expression of BP1 with c-myb and GATA-1, markers of early progenitors, suggest that BP1 expression occurs in primitive cells in AML. Analysis of CD34+ and CD34− normal bone marrow cells revealed BP1 is expressed in CD34− cells and virtually extinguished in CD34+ cells. Ectopic expression of BP1 in the leukemia cell line K562 increased clonogenicity, consistent with a role for BP1 in leukemogenesis. The presence of BP1 RNA in leukemic blasts may therefore be a molecular marker for primitive cells and/or may indicate that BP1 is an important upstream factor in an oncogenic pathway.


IEEE Transactions on Biomedical Circuits and Systems | 2010

Fabrication and Characterization of a Surface-Acoustic-Wave Biosensor in CMOS Technology for Cancer Biomarker Detection

Onur Tigli; Louis Bivona; Patricia E. Berg; Mona E. Zaghloul

Design, fabrication, and characterization of a novel surface acoustic wave (SAW) biosensor in complementary metal-oxide semiconductor (CMOS) technology are introduced. The biosensor employs a streptavidin/biotin-based five-layer immunoassay for detecting a prominent breast cancer biomarker, mammoglobin (hMAM). There is a growing demand to develop a sensitive and specific assay to detect biomarkers in serum that could be used in the early detection of breast cancer, determining prognosis and monitoring therapy. CMOS-SAW devices present a viable alternative to the existing biosensor technologies by providing higher sensitivity levels and better performance at low costs. Two architectures (circular and rectangular) were developed and respective tests were presented for performance comparison. The sensitivities of the devices were analyzed primarily based on center frequency shifts. A frequency sensitivity of 8.704 pg/Hz and a mass sensitivity of 2810.25 m2 /kg were obtained. Selectivity tests were carried out against bovine serum albumin. Experimental results indicate that it is possible to attach cancer biomarkers to functionalized CMOS-SAW sensor surfaces and selectively detect hMAM antigens with improved sensitivities, lowered costs, and increased repeatability of fabrication.


Molecular and Cellular Biology | 2002

BP1, a Homeodomain-Containing Isoform of DLX4, Represses the β-Globin Gene

Michael B. Chase; Sidong Fu; Susanne B. Haga; Gregory J Davenport; Holly Stevenson; Khanh Do; Doris Morgan; Alex L. Mah; Patricia E. Berg

ABSTRACT In earlier studies we identified a putative repressor of the human β-globin gene, termed beta protein 1 (BP1), which binds to two silencer DNA sequences upstream of the adult human β-globin gene and to a negative control region upstream of the adult δ-globin gene. Further studies demonstrated an inverse correlation between the binding affinity of the BP1 protein for the distal β-globin silencer sequence and the severity of sickle cell anemia, suggesting a possible role for BP1 in determining the production of hemoglobin S. We have now cloned a cDNA expressing the BP1 protein. Sequencing revealed that BP1 is a member of the homeobox gene family and belongs to the subfamily called Distal-less (DLX), genes important in early development. Further analysis showed that BP1 is an isoform of DLX4. BP1 protein has repressor function towards the β-globin promoter, acting through the two β-globin DNA silencers, demonstrated in transient transfection assays. Strong BP1 expression is restricted to placenta and kidney tissue, with no expression in 48 other human tissues. BP1 exhibits regulated expression in the human erythroid cell line MB-02, where its expression decreases upon induction of the β-globin gene. BP1 is thus the first member of the DLX family with known DNA binding sites and a function in globin gene regulation.


Breast Cancer Research and Treatment | 2005

Expression of BP1, a novel homeobox gene, correlates with breast cancer progression and invasion.

Yan-gao Man; Sidney W. Fu; Arnold M. Schwartz; Joseph J. Pinzone; Samuel J. Simmens; Patricia E. Berg

SummaryBackgroundOur previous studies revealed that the mRNA encoded by BP1, a member of the homeobox gene superfamily of transcription factors, was expressed in leukemia and infiltrating breast ductal carcinoma (IDC). This study investigated the immunohistochemical profile of BP1, to determine whether the expression of BP1 protein correlated with breast tumor progression and invasion and whether BP1 was co-localized with erbB2.DesignParaffin sections from normal reduction mammoplasties (n = 34) and a variety of in situand invasive breast cancers (n = 270) were either singly immunostained for BP1, or doubly immunostained for BP1 plus either erbB2 or Ki-67.ResultsThe prevalence of BP1 positive cells and the intensity of BP1 immunoreactivity increased with the extent of ductal proliferation and carcinogenesis. BP1 expression was barely detectable in normal reduction mammoplasties compared to distinct staining in 21, 46, and 81% of hyperplastic, in situ, and infiltrating lesions, respectively. In cases with co-existing normal, hyperplastic, in situ, and invasive lesions, the tumor cells of the invasive lesions consistently showed the highest frequency and the highest intensity of BP1 immunostaining, followed by in situ tumor cells. Double immunostaining revealed that BP1 co-localized with a subset of erbB2 positive cells in all 15 in situ and IDC tumors examined, and that BP1 positive cells had a substantially higher proliferation rate than morphologically similar cells without BP1 expression.ConclusionThese findings suggest that BP1 is an important upstream factor in an oncogenic pathway, and that expression of BP1 may reliably reflect or directly contribute to tumor progression and/or invasion.


Breast Cancer Research | 2003

Correlation of expression of BP1, a homeobox gene, with estrogen receptor status in breast cancer.

Sidney W. Fu; Arnold M. Schwartz; Holly Stevenson; Joseph J. Pinzone; Gregory J Davenport; Jan Marc Orenstein; Peter L. Gutierrez; Samuel J. Simmens; Jessy Abraham; Indira Poola; Dietrich A. Stephan; Patricia E. Berg

BackgroundBP1 is a novel homeobox gene cloned in our laboratory. Our previous studies in leukemia demonstrated that BP1 has oncogenic properties, including as a modulator of cell survival. Here BP1 expression was examined in breast cancer, and the relationship between BP1 expression and clinicopathological data was determined.MethodsTotal RNA was isolated from cell lines, tumors, and matched normal adjacent tissue or tissue from autopsy. Reverse transcription polymerase chain reaction was performed to evaluate BP1 expression. Statistical analysis was accomplished with SAS.ResultsAnalysis of 46 invasive ductal breast tumors demonstrated BP1 expression in 80% of them, compared with a lack of expression in six normal breast tissues and low-level expression in one normal breast tissue. Remarkably, 100% of tumors that were negative for the estrogen receptor (ER) were BP1-positive, whereas 73% of ER-positive tumors expressed BP1 (P = 0.03). BP1 expression was also associated with race: 89% of the tumors of African American women were BP1-positive, whereas 57% of those from Caucasian women expressed BP1 (P = 0.04). However, there was no significant difference in BP1 expression between grades I, II, and III tumors. Interestingly, BP1 mRNA expression was correlated with the ability of malignant cell lines to cause breast cancer in mice.ConclusionBecause BP1 is expressed abnormally in breast tumors, it could provide a useful target for therapy, particularly in patients with ER-negative tumors. The frequent expression of BP1 in all tumor grades suggests that activation of BP1 is an early event.


Breast Cancer Research | 2007

BP1 transcriptionally activates bcl-2 and inhibits TNFα-induced cell death in MCF7 breast cancer cells

Holly Stevenson; Sidney W. Fu; Joseph J. Pinzone; Jinguen Rheey; Samuel J. Simmens; Patricia E. Berg

IntroductionWe have previously shown that the Beta Protein 1 (BP1) homeodomain protein is expressed in 81% of invasive ductal breast carcinomas, and that increased BP1 expression correlates with tumor progression. The purpose of our current investigation was to determine whether elevated levels of BP1 in breast cancer cells are associated with increased cell survival.MethodsEffects on cell viability and apoptosis of MCF7 cells stably overexpressing BP1 were determined using MTT and Annexin V assays, and through examination of caspase activation. TNFα was used to induce apoptosis. The potential regulation of apoptosis-associated genes by BP1 was studied using real-time PCR and western blot analyses. Electrophoretic mobility shift assays, site-directed mutagenesis, and transient assays were performed to specifically characterize the interaction of BP1 with the promoter of the bcl-2 gene.ResultsStable overexpression of BP1 led to inhibition of apoptosis in MCF7 breast cancer cells challenged with TNFα. Increased BP1 resulted in reduced processing and activation of caspase-7, caspase-8, and caspase-9, and inactivation of the caspase substrate Poly(ADP-Ribose) Polymerase (PARP). Increased levels of full-length PARP and a decrease in procaspase-8 were also associated with BP1 overexpression. The bcl-2 gene is a direct target of BP1 since: (i) BP1 protein bound to a consensus binding sequence upstream of the bcl-2 P1 promoter in vitro. (ii) MCF7 cells overexpressing BP1 showed increased levels of bcl-2 mRNA and protein. (iii) Transient assays indicated that increased bcl-2 promoter activity is due to direct binding and modulation by BP1 protein. BP1 expression also prevented TNFα-mediated downregulation of bcl-2 mRNA and protein.ConclusionThese findings suggest mechanisms by which increased BP1 may impart a survival advantage to breast cancer cells, which could lead to increased resistance to therapeutic agents in patients.


Modern Pathology | 2009

BP1, a homeoprotein, is significantly expressed in prostate adenocarcinoma and is concordant with prostatic intraepithelial neoplasia.

Arnold M. Schwartz; Yan-gao Man; M. Katayoon Rezaei; Samuel J. Simmens; Patricia E. Berg

BP1 is a member of the homeobox gene superfamily of transcription factors that are essential for early development. Significant mRNA expression and immunohistochemical reactivity of BP1 is present in a majority of breast cancers and in all cases of inflammatory breast cancer. This study attempts to determine whether BP1 expression is detectable in prostate cancer, another hormone dependent solid tumor, and whether this expression correlates with histopathologic and prognostic factors. Paraffin sections from radical prostatectomy cancer specimens and from tissue microarray sections of prostate cancer, obtained from the Prostate Cancer Tissue Registry (NIH), were assayed for BP1 immunoreactivity. Immunoreactivity scoring by two independent pathologists, using a three-tiered system (0, 1+, 2+), was recorded and correlated with Gleason scoring and prostatic specific antigen (PSA) biochemical recurrence. Ki-67 (MIB-1) immunoreactivity was performed to assess proliferation. Kappa and Cochran–Mantel–Haenszel statistical analyses were used to assess interobserver agreement and pathobiologic correlations. Significant BP1 immunoreactivity (2+) was identified in approximately 70% of prostatic adenocarcinomas, whether the analysis was performed on tissue sections (50 cases) or tissue microarray platforms (123 cases). BP1 immunoreactivity was seen in <5% of normal acinar cells. The agreement between two separate observers was very good, with kappa-statistics >0.7. In tissue sections, 12 cases with paired carcinoma and prostatic intraepithelial neoplasia (PIN) showed concordance with strong immunoreactivity. Gleason scores or prostatic specific antigen (PSA) biochemical recurrences were not correlated with strong BP1 immunoreactivity. Tumor proliferation, assayed with Ki-67 (MIB-1) immunoreactivity, was higher in cancer cells that were BP1 immunoreactive, relative to those that were BP1 non-reactive. These findings suggest that BP1 is an important upstream factor in the carcinogenic pathway of prostate cancer and that the expression of BP1 may reflect or directly contribute to tumor progression and/or invasion.


Experimental Cell Research | 1984

Fate and structure of DNA microinjected into mouse TK-L cells.

Michael Huberman; Patricia E. Berg; M. Joan Curcio; Judith M. Dipietro; Ann S. Henderson; W. French Anderson

Co-microinjection of single linearized molecules of plasmids containing the human beta-globin gene (pRK1) and the herpes simplex virus (HSV) type I thymidine kinase gene (pX1) into the mouse TK-L cell nucleus results in covalent linkage between these (or derived) molecules within the nucleus as revealed by Southern blotting, plasmid rescue, and recovery of plasmid-derived DNA from a Charon 4A phage library of cellular DNA. The microinjected DNA is predominantly found as high molecular weight DNA as determined by Hirt fractionation. Southern blotting data and recombinants from the Charon 4A library suggest that the plasmid DNA is in the form of a head-to-tail linear concatamer of up to 80 copies. Passage of these microinjected cells in selective medium (HAT) results in coordinate amplification of both plasmids, which are maintained in an approx. 3:1 molar ratio of pRK1 to pX1-derived molecules. Hybridization in situ shows the DNA to be integrated on a translocation chromosome, t(4;4). Integration does not appear to be site-specific, since plasmid DNA from another microinjected cell line, C2B, appears on a different translocation chromosome, t(8?;14). Plasmid rescue experiments confirm a previous finding that passage of pBR322 DNA through eukaryotic cells may result in deletions of normally stable plasmid DNA upon subsequent transformation of E. coli. These deletions appear to occur in the bacteria, and originate in a 128 bp region between the Sal I and Hae II sites of pBR322.


Annals of Hematology | 2008

Overexpression of BP1, a homeobox gene, is associated with resistance to all-trans retinoic acid in acute promyelocytic leukemia cells

Rania T. Awwad; Khanh Tu Do; Holly Stevenson; Sidney W. Fu; Francesco Lo-Coco; Maura Costello; Cassandra L. Campbell; Patricia E. Berg

BP1, a homeobox gene, is overexpressed in the bone marrow of 63% of acute myeloid leukemia patients. In this study, we compared the growth-inhibitory and cyto-differentiating activities of all-trans retinoic acid (ATRA) in NB4 (ATRA-responsive) and R4 (ATRA-resistant) acute promyelocytic leukemia (APL) cells relative to BP1 levels. Expression of two oncogenes, bcl-2 and c-myc, was also assessed. NB4 and R4 cells express BP1, bcl-2, and c-myc; the expression of all three genes was repressed after ATRA treatment of NB4 cells but not R4 cells. To determine whether BP1 overexpression affects sensitivity to ATRA, NB4 cells were transfected with a BP1-expressing plasmid and treated with ATRA. In cells overexpressing BP1: (1) proliferation was no longer inhibited; (2) differentiation was reduced two- to threefold; (3) c-myc was no longer repressed. These and other data suggest that BP1 may regulate bcl-2 and c-myc expression. Clinically, BP1 levels were elevated in all pretreatment APL patients tested, while BP1 expression was decreased in 91% of patients after combined ATRA and chemotherapy treatment. Two patients underwent disease relapse during follow-up; one patient exhibited a 42-fold increase in BP1 expression, while the other showed no change. This suggests that BP1 may be part of a pathway involved in resistance to therapy. Taken together, our data suggest that BP1 is a potential therapeutic target in APL.

Collaboration


Dive into the Patricia E. Berg's collaboration.

Top Co-Authors

Avatar

Arnold M. Schwartz

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Sidney W. Fu

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Holly Stevenson

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Yan-gao Man

Armed Forces Institute of Pathology

View shared research outputs
Top Co-Authors

Avatar

Samuel J. Simmens

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Saurabh Kirolikar

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Joseph J. Pinzone

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jinguen Rheey

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Sanket Awate

George Washington University

View shared research outputs
Researchain Logo
Decentralizing Knowledge