UB Vice President for Research and Economic Development markup
Rose-Anne Romano UB Profile
Oral Biology
Associate Professor
210 Foster Hall
Buffalo NY 14214
716-829-3665
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Jeremy Kiripolsky, Rose-Anne Romano, Eileen M. Kasperek, Guan Yu, Jill M. Kramer
PubMedId: 31993047
Journal-article: Frontiers in Immunology, vol. 10, 2020
Authors: Akinsola Oyelakin, Erich Horeth, Eun-Ah Christine Song, Sangwon Min, Monika Che, Brandon Marzullo, Christopher J Lessard, Astrid Rasmussen, Lida Radfar, R Hal Scofield, David M Lewis, Donald U Stone, Kiely Grundahl, Scott S De Rossi, Zoya Kurago, A Darise Farris, Kathy L Sivils, Satrajit Sinha, Jill M Kramer, Rose-Anne Romano
PubMedId: 33488608
1: Frontiers in immunology, vol. 11, 2020
Authors: Christian Gluck, Akinsola Oyelakin, Alexandra Glathar, Michael Buck, Rose-Anne Romano, Satrajit Sinha
Proceedings-article: Poster Presentations - Proffered Abstracts, 2020
Authors: Snahlata Singh, Emily Elenio, Nicolae A Leu, Rose-Anne Romano, Andrew E Vaughan, Jennifer DeRiso, Kameswaran Surendran, Rumela Chakrabarti
PubMedId: 31002388
1: FEBS letters, vol. 593, 2019
Authors: Petar Pajic, Pavlos Pavlidis, Kirsten Dean, Lubov Neznanova, Rose-Anne Romano, Danielle Garneau, Erin Daugherity, Anja Globig, Stefan Ruhl, Omer Gokcumen
PubMedId: 31084707
Journal-article: eLife, vol. 8, 2019
Authors: Xiying Fan, Dongmei Wang, Jeremy Evan Burgmaier, Yudong Teng, Rose-Anne Romano, Satrajit Sinha, Rui Yi
Journal-article: Developmental Cell, vol. 47, pp. 133, 2018
Authors: Sangwon Min, Eun-Ah Christine Song, Akinsola Oyelakin, Christian Gluck, Kirsten Smalley, Rose-Anne Romano
PubMedId: 29462154
Journal-article: PLOS ONE, vol. 13, pp. e0192775, 2018
Authors: Eun-Ah Christine Song, Sangwon Min, Akinsola Oyelakin, Kirsten Smalley, Jonathan E. Bard, Lan Liao, Jianming Xu, Rose-Anne Romano
PubMedId: 30232460
Journal-article: Scientific Reports, vol. 8, 2018
Authors: Xiying Fan, Dongmei Wang, Jeremy Evan Burgmaier, Yudong Teng, Rose-Anne Romano, Satrajit Sinha, Rui Yi
PubMedId: 30220568
1: Developmental cell, vol. 47, 2018
Authors: Akinsola Oyelakin, Christian Gluck, Satrajit Sinha, Rose-Anne Romano
Journal-article: Cancer Research, vol. 78, pp. 3350-3350, 2018
Authors: Christian Gluck, Sangwon Min, Akinsola Oyelakin, Kirsten Smalley, Satrajit Sinha, Rose-Anne Romano
PubMedId: 27852218
Journal Article: BMC genomics, vol. 17, 2016
Authors: Isha Sethi, Rose-Anne Romano, Christian Gluck, Kirsten Smalley, Borivoj Vojtesek, Michael J Buck, Satrajit Sinha
Abstract: The transcription factor p63 belongs to the p53/p63/p73 family and plays key functional roles during normal epithelial development and differentiation and in pathological states such as squamous cell carcinomas. The human TP63 gene, located on chromosome 3q28 is driven by two promoters that generate the full-length transactivating (TA) and N-terminal truncated (N) isoforms. Furthermore alternative splicing at the C-terminus gives rise to additional , , and likely several other minor variants. Teasing out the expression and biological function of each p63 variant has been both the focus of, and a cause for contention in the p63 field. Here we have taken advantage of a burgeoning RNA-Seq based genomic data-sets to examine the global expression profiles of p63 isoforms across commonly utilized human cell-lines and major tissues and organs. Consistent with earlier studies, we find Np63 transcripts, primarily that of the Np63 isoforms, to be expressed in most cells of epithelial origin such as those of skin and oral tissues, mammary glands and squamous cell carcinomas. In contrast, TAp63 is not expressed in the majority of normal cell-types and tissues; rather it is selectively expressed at moderate to high levels in a subset of Burkitt's and diffuse large B-cell lymphoma cell lines. We verify this differential expression pattern of p63 isoforms by Western blot analysis, using newly developed N and TA specific antibodies. Furthermore using unsupervised clustering of human cell lines, tissues and organs, we show that Np63 and TAp63 driven transcriptional networks involve very distinct sets of molecular players, which may underlie their different biological functions. In this study we report comprehensive and global expression profiles of p63 isoforms and their relationship to p53/p73 and other potential transcriptional co-regulators. We curate publicly available data generated in part by consortiums such as ENCODE, FANTOM and Human Protein Atlas to delineate the vastly different transcriptomic landscapes of Np63 and TAp63. Our studies help not only in dispelling prevailing myths and controversies on p63 expression in commonly used human cell lines but also augur new isoform- and cell type-specific activities of p63.
PubMedId: 26251276
Journal-article: BMC genomics, vol. 16, 2015
Authors: Jason M. Rizzo, Rose-Anne Romano, Jonathan Bard, Satrajit Sinha
PubMedId: 25202826
Journal-article: Journal of Investigative Dermatology, vol. 135, pp. 629-632, 2015
Authors: Lihua Hu, Jingpeng Liu, Zhi Li, Ferhat Ozturk, Channabasavaiah Gurumurthy, Rose-Anne Romano, Satrajit Sinha, Ali Nawshad
Abstract: The periderm is a flat layer of epithelium created during embryonic development. During palatogenesis, the periderm forms a protective layer against premature adhesion of the oral epithelia, including the palate. However, the periderm must be removed in order for the medial edge epithelia (MEE) to properly adhere and form a palatal seam. Improper periderm removal results in a cleft palate. Although the timing of transforming growth factor 3 (TGF3) expression in the MEE coincides with periderm degeneration, its role in periderm desquamation is not known. Interestingly, murine models of knockout (-/-) TGF3, interferon regulatory factor 6 (IRF6) (-/-), and truncated p63 (Np63) (-/-) are born with palatal clefts because of failure of the palatal shelves to adhere, suggesting that these genes regulate palatal epithelial differentiation. However, despite having similar phenotypes in null mouse models, no studies have analyzed the possible association between the TGF3 signaling cascade and the IRF6/Np63 genes during palate development. Recent studies indicate that regulation of Np63, which depends on IRF6, facilitates epithelial differentiation. We performed biochemical analysis, gene activity and protein expression assays with palatal sections of TGF3 (-/-), Np63 (-/-), and wild-type (WT) embryos, and primary MEE cells from WT palates to analyze the association between TGF3 and IRF6/Np63. Our results suggest that periderm degeneration depends on functional TGF3 signaling to repress Np63, thereby coordinating periderm desquamation. Cleft palate occurs in TGF3 (-/-) because of inadequate periderm removal that impedes palatal seam formation, while cleft palate occurs in Np63 (-/-) palates because of premature fusion.
PubMedId: 25358290
Journal-article: Journal of Cellular Physiology, vol. 230, pp. 1212-1225, 2015
Authors: Rumela Chakrabarti, Yong Wei, Julie Hwang, Xiang Hang, Mario Andres Blanco, Abrar Choudhury, Benjamin Tiede, Rose-Anne Romano, Christina DeCoste, Laura Mercatali, Toni Ibrahim, Dino Amadori, Nagarajan Kannan, Connie J. Eaves, Satrajit Sinha, Yibin Kang
Abstract: Emerging evidence suggests that cancer is populated and maintained by tumour-initiating cells (TICs) with stem-like properties similar to those of adult tissue stem cells. Despite recent advances, the molecular regulatory mechanisms that may be shared between normal and malignant stem cells remain poorly understood. Here we show that the Np63 isoform of the Trp63 transcription factor promotes normal mammary stem cell (MaSC) activity by increasing the expression of the Wnt receptor Fzd7, thereby enhancing Wnt signalling. Importantly, Fzd7-dependent enhancement of Wnt signalling by Np63 also governs tumour-initiating activity of the basal subtype of breast cancer. These findings establish Np63 as a key regulator of stem cells in both normal and malignant mammary tissues and provide direct evidence that breast cancer TICs and normal MaSCs share common regulatory mechanisms.
PubMedId: 25241036
Journal-article: Nature Cell Biology, vol. 16, pp. 1004-1015, 2014
Authors: Rose-Anne Romano, Satrajit Sinha
Keywords: genomics keratinocytes p53 family squamous cell carcinoma
Abstract: The p53 family (p53, p63 and p73) is intimately linked with an overwhelming number of cellular processes during normal physiological as well as pathological conditions including cancer. The fact that these proteins are expressed in myriad isoforms, each with unique biochemical properties and distinct effects on tumorigenesis, complicates their study. A case in point is Squamous Cell Carcinoma (SCC) where p53 is often mutated and the Np63 isoform is overexpressed. Given that p53 and p63 can hetero-dimerize, bind to quite similar DNA elements and share common co-factors, any alterations in their individual expression levels, activity and/or mutation can severely disrupt the family equilibrium. The burgeoning genomics data sets and new additions to the experimental toolbox are offering crucial insights into the complex role of the p53 family in SCC, but more mechanistic studies are needed.
PubMedId: 24690037
Journal-article: Experimental Dermatology, vol. 23, pp. 238-239, 2014
Authors: Jihui Du, Rose-Anne Romano, Han Si, Austin Mattox, Yansong Bian, Xinping Yang, Satrajit Sinha, Carter Van Waes, Zhong Chen
Journal-article: The Journal of Pathology, vol. 232, pp. 356-368, 2014
Authors: N S Chari, R A Romano, M I Koster, V Jaks, D Roop, E R Flores, S Teglund, S Sinha, W Gruber, F Aberger, L J Medeiros, R Toftgard, T J McDonnell
Abstract: Deregulation of the hedgehog (HH) pathway results in overexpression of the GLI target BCL2 and is an initiating event in specific tumor types including basal cell carcinoma of the skin. Regulation of the HH pathway during keratinocyte differentiation is not well understood. We measured HH pathway activity in response to differentiation stimuli in keratinocytes. An upregulation of suppressor of fused (SUFU), a negative regulator of the HH pathway, lowered HH pathway activity and was accompanied by loss of BCL2 expression associated with keratinocyte differentiation. We used in vitro and in vivo models to demonstrate that Np63, a crucial regulator of epidermal development, activates SUFU transcription in keratinocytes. Increasing SUFU protein levels inhibited GLI-mediated gene activation in suprabasal keratinocytes and promoted differentiation. Loss of SUFU expression caused deregulation of keratinocyte differentiation and BCL2 overexpression. Using in vivo murine models, we also provide evidence of GLI-mediated regulation of the TP63 pathway. p63 expression appears essential to establish an optimally functioning HH pathway. These observations present a regulatory mechanism by which SUFU acts as an interacting node between the HH and TP63 pathways to mediate differentiation and maintain epidermal homeostasis. Disruption of this regulatory node can be an important contributor to multistep carcinogenesis.
PubMedId: 23686138
Journal-article: Cell Death & Differentiation, vol. 20, pp. 1080-1088, 2013
Authors: Robert M Weiss, Songshan Guo, Alan Shan, Hongmei Shi, Rose-Anne Romano, Satrajit Sinha, Lloyd G Cantley, Jian-Kan Guo
PubMedId: 23449535
Journal Article: Journal of the American Society of Nephrology : JASN, vol. 24, 2013
Authors: Rumela Chakrabarti, Julie Hwang, Mario Andres Blanco, Yong Wei, Martin Luka??i??in, Rose-Anne Romano, Kirsten Smalley, Song Liu, Qifeng Yang, Toni Ibrahim, Laura Mercatali, Dino Amadori, Bruce G. Haffty, Satrajit Sinha, Yibin Kang
Abstract: The epithelial-mesenchymal transition (EMT) is a complex process that occurs during organogenesis and in cancer metastasis. Despite recent progress, the molecular pathways connecting the physiological and pathological functions of EMT need to be better defined. Here we show that the transcription factor Elf5, a key regulator of mammary gland alveologenesis, controls EMT in both mammary gland development and metastasis. We uncovered this role for Elf5 through analyses of Elf5 conditional knockout animals, various in vitro and in vivo models of EMT and metastasis, an MMTV-neu transgenic model of mammary tumour progression and clinical breast cancer samples. Furthermore, we demonstrate that Elf5 suppresses EMT by directly repressing the transcription of Snail2, a master regulator of mammary stem cells and a known inducer of EMT. These findings establish Elf5 not only as a key cell lineage regulator during normal mammary gland development, but also as a suppressor of EMT and metastasis in breast cancer.
PubMedId: 23086238
Journal-article: Nature Cell Biology, vol. 14, pp. 1212-1222, 2012
Authors: Rumela Chakrabarti, Yong Wei, Rose-Anne Romano, Christina DeCoste, Yibin Kang, Satrajit Sinha
Abstract: The transcription factor E74-like factor 5 (Elf5) functions downstream of the prolactin receptor signaling pathway and plays an important role in mammary gland development. Using conditional mouse knockouts, we have previously shown that Elf5-null mammary glands exhibit a complete failure of alveologenesis during pregnancy. The Elf5-null developmental phenotype is mediated through alteration in the expression of several critical genes involved in alveologenesis, particularly those belonging to the JAK/STAT pathway. Here, we demonstrate that in addition to regulating terminal differentiation of alveolar cells, Elf5 also plays a critical role in determining cell fate and in regulating the stem/progenitor function of the mammary epithelium. Targeted deletion of Elf5 in the mammary glands leads to accumulation of cell types with dual luminal/basal properties such as coexpression of K8 and K14 and an increase in CD61(+) luminal progenitor population during pregnancy. Further interrogation suggests that the abnormal increase in K14(+) K8(+) cells may represent the CD61(+) luminal progenitors blocked in differentiation. Remarkably, Elf5 deficiency in mammary epithelium also triggers an increase of adult mammary stem activity as evidenced by the accumulation of mammary stem cell (MaSC)-enriched cell population in both pregnant and virgin mice and further confirmed by mammosphere and transplantation assays. Additional support for this phenotype comes from the enriched MaSC gene signature based on transcriptomic analysis of the Elf5-null mammary gland. Finally, our biochemical studies suggest that Elf5 loss leads to hyperactivation of the Notch signaling pathway, which might constitute in part, the underlying molecular mechanism for the altered cell lineage decisions in Elf5-null mammary epithelial cells.
PubMedId: 22523003
Journal-article: STEM CELLS, vol. 30, pp. 1496-1508, 2012
Authors: Karen Mitchell, James O'Sullivan, Caterina Missero, Ed Blair, Rose Richardson, Beverley Anderson, Dario Antonini, Jeffrey??C. Murray, Alan??L. Shanske, Brian??C. Schutte, Rose-Anne Romano, Satrajit Sinha, Sanjeev??S. Bhaskar, Graeme??C.M. Black, Jill Dixon, Michael??J. Dixon
Journal-article: The American Journal of Human Genetics, vol. 90, pp. 69-75, 2012
Authors: Jihui Du, Rose-Anne Romano, Xinping Yang, Han Si, Yansong Bian, Satrajit Sinha, Carter Van Waes, Zhong Chen
Journal-article: Cancer Research, vol. 72, pp. 3549-3549, 2012
Authors: Rose-Anne Romano, Satrajit Sinha
PubMedId: 22013738
Journal Article: Indian journal of experimental biology, vol. 49, 2011
Authors: Adam Packard, Nikolai Schnittke, Rose-Anne Romano, Satrajit Sinha, James E Schwob
Abstract: The ability of the olfactory epithelium (OE) to regenerate after injury is mediated by at least two populations of presumed stem cells-globose basal cells (GBCs) and horizontal basal cells (HBCs). Of the two, GBCs are molecularly and phenotypically analogous to the olfactory progenitors of the embryonic placode (OPPs). In contrast, HBCs are a reserve stem cell population that appears later in development and requires activation by severe epithelial damage before contributing to epithelial reconstitution. Neither HBC emergence nor the mechanism of activation after injury is understood. Here we show that the transcription factor p63 (Trp63), which is expressed selectively by adult HBCs, is required for HBC differentiation. The first evidence of HBC differentiation is the expression of p63 by cells that closely resemble embryonic OPPs and adult GBCs by morphology and expression of the transcription factors Sox2, Ascl1, and Hes1. HBC formation is delayed in Ascl1 knock-out OE and is completely abrogated in p63-null mice. Strikingly, other cell types of the OE form normally in the p63 knock-out OE. The role of p63 in HBC differentiation appears to be conserved in the regenerating rat OE, where HBCs disappear and then reappear after tissue lesion. Finally, p63 protein is downregulated in HBCs activated by lesion to become multipotent progenitor cells. Together, our data identify a novel mechanism for the generation of a reserve stem cell population and suggest that a p63-dependent molecular switch is responsible for activating reserve stem cells when they are needed.
PubMedId: 21677159
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 31, 2011
Authors: R A Romano, L W Solomon, S Sinha
Abstract: The Tp63 gene encodes for multiple isoforms of the p63 transcription factor, a member of the p53 family of proteins. Much like its more famous sibling, the biological role of p63 is quite complex, with wide-ranging effects on development, differentiation, and cellular lineage choices. The crucial function of p63 is epitomized by the striking phenotype of p63 knockout mice. These animals have a profound block in the development of stratified epithelia and aplasia of multiple ectodermal appendages, as well as orofacial clefting and limb defects. Remarkably, a similar spectrum of phenotypic alterations is observed in human syndromes resulting from Tp63 gene mutations. p63 is an important hub in the transcriptional and signaling networks of epithelial cells; thus, it is not surprising that dysregulation of this transcription factor is associated with squamous cell carcinoma. Finally, as a testament to the growing repertoire of p63-associated diseases, autoantibodies to p63 are associated with chronic ulcerative stomatitis, an oral immunologically mediated disease. Over the past decade, our understanding of the broad biologic and pathophysiological roles of p63 has grown significantly. In this review, we discuss the molecular attributes of Tp63 and the clinical consequences of Tp63 dysregulation, particularly as it pertains to oral tissues.
PubMedId: 21646640
Journal-article: Journal of dental research, vol. 91, 2011
Authors: Rose-Anne Romano, Kirsten Smalley, Song Liu, Satrajit Sinha
Abstract: The transcription factor p63 plays an essential role in epidermal morphogenesis. Animals lacking p63 fail to form many ectodermal organs, including the skin and hair follicles. Although the indispensable role of p63 in stratified epithelial skin development is well established, relatively little is known about this transcriptional regulator in directing hair follicle morphogenesis. Here, using specific antibodies, we have established the expression pattern of DeltaNp63 in hair follicle development and cycling. DeltaNp63 is expressed in the developing hair placode, whereas in mature hair its expression is restricted to the outer root sheath (ORS), matrix cells and to the stem cells of the hair follicle bulge. To investigate the role of DeltaNp63 in hair follicle morphogenesis and cycling, we have utilized a Tet-inducible mouse model system with targeted expression of this isoform to the ORS of the hair follicle. DeltaNp63 transgenic animals display dramatic defects in hair follicle development and cycling, eventually leading to severe hair loss. Strikingly, expression of DeltaNp63 leads to a switch in cell fate of hair follicle keratinocytes, causing them to adopt an interfollicular epidermal (IFE) cell identity. Moreover, DeltaNp63 transgenic animals exhibit a depleted hair follicle stem-cell niche, which further contributes to the overall cycling defects observed in the mutant animals. Finally, global transcriptome analysis of transgenic skin identified altered expression levels of crucial mediators of hair morphogenesis, including key members of the Wnt/beta-catenin signaling pathway, which, in part, account for these effects. Our data provide evidence supporting a role for DeltaNp63alpha in actively suppressing hair follicle differentiation and directing IFE cell lineage commitment.
PubMedId: 20335364
Journal-article: Development (Cambridge, England), vol. 137, 2010
Authors: Rosalba Escamilla-Hernandez, Rumela Chakrabarti, Rose-Anne Romano, Kirsten Smalley, Qianqian Zhu, William Lai, Marc S Halfon, Michael J Buck, Satrajit Sinha
Abstract: The ETS transcription factor Elf5 (also known as ESE-2) is highly expressed in the mammary gland and plays an important role in its development and differentiation. Indeed studies in mice have illustrated an essential role for Elf5 in directing alveologenesis during pregnancy. Although the molecular mechanisms that underlie the developmental block in Elf5 null mammary glands are beginning to be unraveled, this investigation has been hampered by limited information about the identity of Elf5-target genes. To address this shortcoming, in this study we have performed ChIP-cloning experiments to identify the specific genomic segments that are occupied by Elf5 in pregnant mouse mammary glands. Sequencing and genomic localization of cis-regulatory regions bound by Elf5 in vivo has identified several potential target genes covering broad functional categories. A subset of these target genes demonstrates higher expression levels in Elf5-null mammary glands suggesting a repressive functional role for this transcription factor. Here we focus on one putative target of Elf5, the Ccnd2 gene that appeared in our screen. We identify a novel Elf5-binding segment upstream of the Ccnd2 gene and demonstrate that Elf5 can transcriptionally repress Ccnd2 by directly binding to the proximal promoter region. Finally, using Elf5-null mammary epithelial cells and mammary glands, we show that loss of Elf5 in vivo leads to up regulation of Ccnd2 and an altered expression pattern in luminal cells. Identification of Elf5-targets is an essential first step in elucidating the transcriptional landscape that is shaped by this important regulator. Our studies offer new toolbox in examining the biological role of Elf5 in mammary gland development and differentiation.
PubMedId: 20831799
Journal-article: BMC molecular biology, vol. 11, 2010
Authors: Rose-Anne Romano, Kori Ortt, Barbara Birkaya, Kirsten Smalley, Satrajit Sinha
Abstract: One major defining characteristic of the basal keratinocytes of the stratified epithelium is the expression of the keratin genes K5 and K14. The temporal and spatial expression of these two genes is usually tightly and coordinately regulated at the transcriptional level. This ensures the obligate pairing of K5 and K14 proteins to generate an intermediate filament (IF) network that is essential for the structure and function of the proliferative keratinocytes. Our previous studies have shown that the basal-keratinocyte restricted transcription factor p63 is a direct regulator of K14 gene. Here we provide evidence that p63, specifically the DeltaN isoform also regulates the expression of the K5 gene by binding to a conserved enhancer within the 5' upstream region. By using specific antibodies against DeltaNp63, we show a concordance in the expression between basal keratins and DeltaNp63 proteins but not the TAp63 isoforms during early embryonic skin development. We demonstrate, that contrary to a previous report, transgenic mice expressing DeltaNp63 in lung epithelium exhibit squamous metaplasia with de novo induction of K5 and K14 as well as transdifferentiation to the epidermal cell lineage. Interestingly, the in vivo epidermal inductive properties of DeltaNp63 do not require the C-terminal SAM domain. Finally, we show that DeltaNp63 alone can restore the expression of the basal keratins and reinitiate the failed epidermal differentiation program in the skin of p63 null animals. DeltaNp63 is a critical mediator of keratinocyte stratification program and directly regulates the basal keratin genes.
PubMedId: 19461998
Journal-article: PloS one, vol. 4, 2009
Authors: R. Kommagani, M. K. Leonard, S. Lewis, R.-A. Romano, S. Sinha, M. P. Kadakia
Abstract: The p63 transcription factor has a pivotal role in epithelial morphogenesis. Multiple transcripts of the TP63 gene are generated because of alternative promoter usage and splicing. DeltaNp63alpha is the predominant isoform of p63 observed during epithelial morphogenesis and in human cancers. Loss of DeltaNp63alpha expression has been shown to promote invasiveness in a subset of human cancer cell lines. Here, we studied whether the regulation of VDR by DeltaNp63alpha controls the invasiveness of an epidermoid cancer cell line. We demonstrate that VDR expression is induced by all p63 isoforms, including DeltaNp63alpha. Endogenous DeltaNp63alpha protein was observed to bind to the VDR promoter, and silencing of endogenous DeltaNp63alpha resulted in diminished VDR expression. Although silencing of p63 inhibits VDR expression leading to an increase in cell migration, overexpression of p63 or VDR results in reduced cell migration as a result of increased VDR expression. Therefore, it is conceivable that p63 inhibits cell invasion by regulating VDR expression. Finally, we observed that expression of p63 and VDR overlaps in the wild-type mouse skin, but a reduced or complete absence of VDR expression was observed in skin from p63-null mice and in p63-null mouse embryonic fibroblasts. In conclusion, we demonstrate a direct transcriptional regulation of VDR by DeltaNp63alpha. Our results highlight a crucial role for VDR in p63-mediated biological functions.
PubMedId: 19622632
Journal-article: Journal of Cell Science, vol. 122, pp. 2828-2835, 2009
Authors: Rose-Anne Romano, Satrajit Sinha
Abstract: The ability to specifically manipulate gene expression in vivo using mouse models has been one of the most important advances in understanding gene function over the last few decades. Methods used to control gene activities in the mouse include gene targeting and transgenic approaches. While gene targeting methods have proven to be powerful genetic tools designed to eliminate gene function by creating a "knockout" or "null mutant," transgenic studies offer gain-of-function capabilities, expression of dominant negative or knockdown of specific target genes and thus have often served a complementary and useful role. This chapter provides an overview for the generation of transgenic mouse models to study important questions in skin biology by taking advantage of the tetracycline-inducible gene expression system.
PubMedId: 19908011
Journal-article: Methods in molecular biology (Clifton, N.J.), vol. 585, 2009
Authors: Yiping Huang, Alice Y Chuang, Rose-Anne Romano, Nanette J Li?geois, Satrajit Sinha, Barry Trink, Edward Ratovitski, David Sidransky
PubMedId: 20023394
Journal Article: Cell cycle (Georgetown, Tex.), vol. 9, 2009
Authors: Rose-Anne Romano, Satrajit Sinha, Priyadharsini Nagarajan
Journal-article: Critical Reviews??? in Eukaryotic Gene Expression, vol. 18, pp. 57-79, 2008
Authors: Priyadharsini Nagarajan, Rose-Anne Romano, Satrajit Sinha
PubMedId: 18197786
Journal Article: Critical reviews in eukaryotic gene expression, vol. 18, 2008
Authors: Rose-Anne Romano, Barbara Birkaya, Satrajit Sinha
Abstract: Keratin14 (K14) is a prototypic marker of dividing basal keratinocytes where its gene is transcribed at high levels. Transcriptional regulation of K14 is governed by an evolutionarily conserved functional enhancer marked by DNase 1 hypersensitive sites present upstream of the gene. This enhancer is sufficient to confer epidermal-specific gene expression, which is mediated in part by binding of members of activator protein-2 (AP)-2, AP-1, Ets, and Sp1 families of transcription factors. Here we provide evidence that a keratinocyte-specific nuclear protein identified as deltaNp63 binds to a conserved motif within this enhancer. Interestingly, the selective expression profile of deltaNp63 in various cell lines correlates with both the nuclear complex and the expression of K14. Biochemical studies reveal that deltaNp63 can bind to a specific DNA sequence present in the K14 enhancer and this binding leads to transactivation. In addition, chromatin immunoprecipitation experiments with deltaNp63-specific antibodies demonstrate that the enhancer is occupied by deltaNp63 in cultured keratinocytes and in mouse skin epidermal cells in vivo. Finally, we show that ectopic expression of either p63 isoform (deltaN or TA) can induce de novo expression of K14. These studies provide a potential mechanism by which deltaNp63 directly governs the expression of K14 in a keratinocyte-specific manner.
PubMedId: 17159913
Journal-article: Journal of Investigative Dermatology, vol. 127, pp. 1175-1186, 2007
Authors: Serena Borrelli, Barbara Testoni, Maurizio Callari, Daniela Alotto, Carlotta Castagnoli, Rose-Anne Romano, Satrajit Sinha, Alessandra M Vigan??, Roberto Mantovani
PubMedId: 17903252
Journal-article: BMC Molecular Biology, vol. 8, pp. 85, 2007
Authors: Rose-Anne Romano, Barbara Birkaya, Satrajit Sinha
Abstract: p63, a homolog of the tumor suppressor p53, plays an important role in the formation of stratified epithelium such as those in the epidermis of the skin. The p63 gene gives rise to multiple functionally distinct protein isoforms, including the DeltaNp63 class of isoforms, which lacks the N-terminal transactivation domain and is synthesized from an internal promoter. DeltaNp63 proteins are the predominant isoforms expressed in keratinocytes and are thought to be important for maintenance of the proliferative capacity of these cells. Here, we have examined the transcriptional control mechanisms that govern the expression DeltaNp63 in keratinocytes. We first performed DNase I hypersensitive site mapping and demonstrated that the promoter region of DeltaNp63 is in open chromatin state in keratinocytes. To identify the cis-elements that regulate DeltaNp63, we have performed transient transfection assays in keratinocytes with several DeltaNp63 promoter constructs. This identified a short evolutionarily conserved fragment that harbors most of the transcriptional activity of the DeltaNp63 promoter. Biochemical studies of this element have revealed critical roles for CCAAT-box-binding factor (CBF/NF-Y) and Sp1/Sp3 family of proteins. In addition, our data suggest that DeltaNp63 is recruited to and can activate its own promoter, possibly through protein-protein interactions, thus providing an auto-regulatory loop of self-regulation. These studies support the notion that unique and distinct pathways control the expression of individual p53 family members and their various isoforms.
PubMedId: 16645595
Journal-article: The Journal of investigative dermatology, vol. 126, 2006
Authors: Rose-Anne Romano, Hongxiu Li, Ramakumar Tummala, Robert Maul, Satrajit Sinha
Abstract: We used a bioinformatics approach to identify Basonuclin2, the second member of the Basonuclin zinc-finger family of transcription factors. The mouse Basonuclin2 protein consists of 1049 amino acids and contains three pairs of zinc fingers in the C-terminus that show a high level of amino acid sequence similarity with Basonuclin1. In addition, other characteristic domains of Basonuclin1, such as the serine strip and a nuclear localization signal, are also present in Basonuclin2. We used genomic and in silico database analysis to identify the human and rat homologs of basonuclin2. A search of the mouse genome showed that the basonuclin2 gene maps to chromosome 4 and consists of six exons spanning approximately 300 kb. Northern blot analysis revealed multiple transcripts of basonuclin2 in tissues of the reproductive system (ovary and testis) and also in kidney and skin. We demonstrate that, as expected from sequence conservation, recombinant Basonuclin2 can bind to a sequence in the promoter of a rRNA gene previously characterized as a Basonuclin-binding site. Full-length Basonuclin2 exclusively localizes to the nucleus, indicating that it likely plays an important role in nuclear function, probably in gene regulation. Our study establishes Basonuclin2 as a novel member of the Basonuclin family. Moreover, the structural and functional similarities with Basonuclin1 suggest that Basonuclin2 may play an analogous function in germ cells and skin keratinocytes.
PubMedId: 15081112
Journal-article: Genomics, vol. 83, pp. 821-833, 2004
Authors: Ramakumar Tummala, Rose-Anne Romano, Elaine Fuchs, Satrajit Sinha
Journal-article: Gene, vol. 321, pp. 93-102, 2003
Authors: Ramakumar Tummala, Rose-Anne Romano, Elaine Fuchs, Satrajit Sinha
PubMedId: 14636996
Journal Article: Gene, vol. 321, 2003
Presented by CUBS © 2024