UB Vice President for Research and Economic Development markup
Satrajit Sinha UB Profile
Biochemistry
Professor
701 Ellicott Street
Buffalo NY 14203
716-881-7994
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Christian Gluck, Akinsola Oyelakin, Alexandra Glathar, Michael Buck, Rose-Anne Romano, Satrajit Sinha
Proceedings-article: Poster Presentations - Proffered Abstracts, 2020
Authors: Sangwon Min, Akinsola Oyelakin, Christian Gluck, Jonathan E. Bard, Eun-Ah Christine Song, Kirsten Smalley, Monika Che, Elsa Flores, Satrajit Sinha, Rose-Anne Romano
Journal-article: iScience, vol. 23, pp. 101524, 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: Snahlata Singh, Sushil Kumar, Ratnesh Kumar Srivastava, Ajeya Nandi, Gatha Thacker, Hemma Murali, Sabrina Kim, Mary Baldeon, John Tobias, Mario Andres Blanco, Rizwan Saffie, M. Raza Zaidi, Satrajit Sinha, Luca Busino, Serge Y. Fuchs, Rumela Chakrabarti
Journal-article: Nature Cell Biology, vol. 22, pp. 591-602, 2020
Authors: Sushil Kumar, Ajeya Nandi, Aakash Mahesh, Satrajit Sinha, Elsa Flores, Rumela Chakrabarti
Journal-article: FEBS Letters, 2019
Authors: A. Oyelakin, E.A.C. Song, S. Min, J.E. Bard, J.V. Kann, E. Horeth, K. Smalley, J.M. Kramer, S. Sinha, R.A. Romano
Journal-article: Journal of Dental Research, vol. 98, pp. 1539-1547, 2019
Authors: Christian Gluck, Alexandra Glathar, Maria Tsompana, Norma Nowak, Lee Ann Garrett-Sinha, Michael J Buck, Satrajit Sinha
PubMedId: 31306413
1: PLoS genetics, vol. 15, 2018
Authors: Maria Tsompana, Christian Gluck, Isha Sethi, Ishita Joshi, Jonathan Bard, Norma J Nowak, Satrajit Sinha, Michael J Buck
PubMedId: 31477832
1: Oncogene, vol. 39, 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: Claudia Russo, Christian Osterburg, Anna Sirico, Dario Antonini, Raffaele Ambrosio, Julia Maren W?rz, J?rg Rinnenthal, Marco Ferniani, Sebastian Kehrloesser, Birgit Sch?fer, Peter G?ntert, Satrajit Sinha, Volker D?tsch, Caterina Missero
PubMedId: 29339502
1: Proceedings of the National Academy of Sciences of the United States of America, vol. 115, 2018
Authors: Akinsola Oyelakin, Christian Gluck, Satrajit Sinha, Rose-Anne Romano
Journal-article: Cancer Research, vol. 78, pp. 3350-3350, 2018
Authors: Sushil Kumar, David W. Wilkes, Nina Samuel, Mario Andres Blanco, Anupma Nayak, Kevin Alicea-Torres, Christian Gluck, Satrajit Sinha, Dmitry Gabrilovich, Rumela Chakrabarti
Journal-article: Journal of Clinical Investigation, vol. 128, pp. 5095-5109, 2018
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: Rose Richardson, Karen Mitchell, Nigel L. Hammond, Maria Rosaria Mollo, Evelyn N. Kouwenhoven, Niki D. Wyatt, Ian J. Donaldson, Leo Zeef, Tim Burgis, Rognvald Blance, Simon J. van Heeringen, Hendrik G. Stunnenberg, Huiqing Zhou, Caterina Missero, Rose Anne Romano, Satrajit Sinha, Michael J. Dixon, Jill Dixon
Abstract: Cleft palate is a common congenital disorder that affects up to 1 in 2500 live births and results in considerable morbidity to affected individuals and their families. The aetiology of cleft palate is complex with both genetic and environmental factors implicated. Mutations in the transcription factor p63 are one of the major individual causes of cleft palate; however, the gene regulatory networks in which p63 functions remain only partially characterized. Our findings demonstrate that p63 functions as an essential regulatory molecule in the spatio-temporal control of palatal epithelial cell fate to ensure appropriate fusion of the palatal shelves. Initially, p63 induces periderm formation and controls its subsequent maintenance to prevent premature adhesion between adhesion-competent, intra-oral epithelia. Subsequently, TGF3-induced down-regulation of p63 in the medial edge epithelia of the palatal shelves is a pre-requisite for palatal fusion by facilitating periderm migration from, and reducing the proliferative potential of, the midline epithelial seam thereby preventing cleft palate.
PubMedId: 28604778
Journal-article: PLOS Genetics, vol. 13, pp. e1006828, 2017
Authors: Christian Gluck, Isha Sethi, Maria Tsompana, Satrajit Sinha
Proceedings-article: Breakthroughs in Imaging Head and Neck Cancers, 2017
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, Maria Tsompana, Christian Gluck, Michael J. Buck, Satrajit Sinha
Journal-article: Cancer Research, vol. 76, pp. B13-B13, 2016
Authors: Isha Sethi, Christian Gluck, Huiqing Zhou, Michael J Buck, Satrajit Sinha
Abstract: Although epidermal keratinocyte development and differentiation proceeds in similar fashion between humans and mice, evolutionary pressures have also wrought significant species-specific physiological differences. These differences between species could arise in part, by the rewiring of regulatory network due to changes in the global targets of lineage-specific transcriptional master regulators such as p63. Here we have performed a systematic and comparative analysis of the p63 target gene network within the integrated framework of the transcriptomic and epigenomic landscape of mouse and human keratinocytes. We determined that there exists a core set of 1600 genomic regions distributed among enhancers and super-enhancers, which are conserved and occupied by p63 in keratinocytes from both species. Notably, these DNA segments are typified by consensus p63 binding motifs under purifying selection and are associated with genes involved in key keratinocyte and skin-centric biological processes. However, the majority of the p63-bound mouse target regions consist of either murine-specific DNA elements that are not alignable to the human genome or exhibit no p63 binding in the orthologous syntenic regions, typifying an occupancy lost subset. Our results suggest that these evolutionarily divergent regions have undergone significant turnover of p63 binding sites and are associated with an underlying inactive and inaccessible chromatin state, indicative of their selective functional activity in the transcriptional regulatory network in mouse but not human. Furthermore, we demonstrate that this selective targeting of genes by p63 correlates with subtle, but measurable transcriptional differences in mouse and human keratinocytes that converges on major metabolic processes, which often exhibit species-specific trends. Collectively our study offers possible molecular explanation for the observable phenotypic differences between the mouse and human skin and broadly informs on the prevailing principles that govern the tug-of-war between evolutionary forces of rigidity and plasticity over transcriptional regulatory programs.
PubMedId: 28505376
Journal-article: Nucleic acids research, vol. 45, 2016
Authors: Christian Gluck, Isha Sethi, Maria Tsompana, Satrajit Sinha
Journal-article: Cancer Research, vol. 76, pp. A50-A50, 2016
Authors: H Si, H Lu, X Yang, A Mattox, M Jang, Y Bian, E Sano, H Viadiu, B Yan, C Yau, S Ng, S K Lee, R-A Romano, S Davis, R L Walker, W Xiao, H Sun, L Wei, S Sinha, C C Benz, J M Stuart, P S Meltzer, C Van Waes, Z Chen
Abstract: The Cancer Genome Atlas (TCGA) network study of 12 cancer types (PanCancer 12) revealed frequent mutation of TP53, and amplification and expression of related TP63 isoform Np63 in squamous cancers. Further, aberrant expression of inflammatory genes and TP53/p63/p73 targets were detected in the PanCancer 12 project, reminiscent of gene programs comodulated by cREL/Np63/TAp73 transcription factors we uncovered in head and neck squamous cell carcinomas (HNSCCs). However, how inflammatory gene signatures and cREL/p63/p73 targets are comodulated genome wide is unclear. Here, we examined how the inflammatory factor tumor necrosis factor- (TNF-) broadly modulates redistribution of cREL with Np63/TAp73 complexes and signatures genome wide in the HNSCC model UM-SCC46 using chromatin immunoprecipitation sequencing (ChIP-seq). TNF- enhanced genome-wide co-occupancy of cREL with Np63 on TP53/p63 sites, while unexpectedly promoting redistribution of TAp73 from TP53 to activator protein-1 (AP-1) sites. cREL, Np63 and TAp73 binding and oligomerization on NF-B-, TP53- or AP-1-specific sequences were independently validated by ChIP-qPCR (quantitative PCR), oligonucleotide-binding assays and analytical ultracentrifugation. Function of the binding activity was confirmed using TP53-, AP-1- and NF-B-specific REs or p21, SERPINE1 and IL-6 promoter luciferase reporter activities. Concurrently, TNF- regulated a broad gene network with cobinding activities for cREL, Np63 and TAp73 observed upon array profiling and reverse transcription-PCR. Overlapping target gene signatures were observed in squamous cancer subsets and in inflamed skin of transgenic mice overexpressing Np63. Furthermore, multiple target genes identified in this study were linked to TP63 and TP73 activity and increased gene expression in large squamous cancer samples from PanCancer 12 TCGA by CircleMap. PARADIGM inferred pathway analysis revealed the network connection of TP63 and NF-B complexes through an AP-1 hub, further supporting our findings. Thus, inflammatory cytokine TNF- mediates genome-wide redistribution of the cREL/p63/p73, and AP-1 interactome, to diminish TAp73 tumor suppressor function and reciprocally activate NF-B and AP-1 gene programs implicated in malignancy.
PubMedId: 27132513
Journal-article: Oncogene, vol. 35, pp. 5781-5794, 2016
Authors: Rumela Chakrabarti, Satrajit Sinha, Yibin Kang
Journal-article: Molecular Cancer Research, vol. 14, pp. B21-B21, 2016
Authors: F. Rahimi, H. Li, S. Sinha, G. Bitan
Book-chapter: Developing Therapeutics for Alzheimer's Disease, pp. 97-191, 2016
Authors: J M Rizzo, A Oyelakin, S Min, K Smalley, J Bard, W Luo, J Nyquist, E Guttman-Yassky, T Yoshida, A De Benedetto, L A Beck, S Sinha, R-A Romano
Abstract: Atopic dermatitis (AD) is the most common inflammatory skin disease with no well-delineated cause or effective cure. Here we show that the p53 family member p63, specifically the Np63, isoform has a key role in driving keratinocyte activation in AD. We find that overexpression of Np63 in transgenic mouse epidermis results in a severe skin phenotype that shares many of the key clinical, histological and molecular features associated with human AD. This includes pruritus, epidermal hyperplasia, aberrant keratinocyte differentiation, enhanced expression of selected cytokines and chemokines and the infiltration of large numbers of inflammatory cells including type 2 T-helper cells - features that are highly representative of AD dermatopathology. We further demonstrate several of these mediators to be direct transcriptional targets of Np63 in keratinocytes. Of particular significance are two p63 target genes, IL-31 and IL-33, both of which are key players in the signaling pathways implicated in AD. Importantly, we find these observations to be in good agreement with elevated levels of Np63 in skin lesions of human patients with AD. Our studies reveal an important role for Np63 in the pathogenesis of AD and offer new insights into its etiology and possible therapeutic targets.
PubMedId: 26768665
Journal-article: Cell Death & Differentiation, vol. 23, pp. 1073-1085, 2016
Authors: Samir S El-Dahr, Yuwen Li, Jiao Liu, Elleny Gutierrez, Kathleen S Hering-Smith, Sabina Signoretti, Jean-Christophe Pignon, Satrajit Sinha, Zubaida Saifudeen
PubMedId: 28469077
1: JCI insight, vol. 2, 2016
Authors: Justin Grassmeyer, Malini Mukherjee, Jennifer deRiso, Casey Hettinger, Monica Bailey, Satrajit Sinha, Jane E Visvader, Haotian Zhao, Eric Fogarty, Kameswaran Surendran
PubMedId: 28215940
1: Developmental biology, vol. 424, 2016
Authors: Heather R Conti, Vincent M Bruno, Erin E Childs, Sean Daugherty, Joseph P Hunter, Bemnet G Mengesha, Danielle L Saevig, Matthew R Hendricks, Bianca M Coleman, Lucas Brane, Norma Solis, J Agustin Cruz, Akash H Verma, Abhishek V Garg, Amy G Hise, Jonathan P Richardson, Julian R Naglik, Scott G Filler, Jay K Kolls, Satrajit Sinha, Sarah L Gaffen
PubMedId: 27923704
1: Cell host & microbe, vol. 20, 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: Jason M. Rizzo, Satrajit Sinha
Abstract: Eukaryotic DNA is wrapped around histone octamers, known as nucleosomes, in an orderly fashion that provides the primary structure of chromatin organization. The compaction of DNA into nucleosomal repeats not only allows the tight packaging of the large eukaryotic genomes into the nucleus, it also dictates the accessibility of genetic information. Thus, in order to understand how nucleosomes can affect the dynamics of DNA-protein interactions, such as those associated with transcriptional regulatory mechanisms, it is important to define nucleosomal positioning and occupancy along genomic DNA. Here we describe a method that relies on the enzymatic activity of micrococcal nuclease (MNase) to determine nucleosomal footprints and boundaries. By pairing this technique with next generation sequencing techniques (i.e., MNase-seq), it is possible to generate a genome-wide detailed map of chromatin architecture.
PubMedId: 24676786
Book-chapter: Methods in Molecular Biology, pp. 49-59, 2014
Authors: Isha Sethi, Satrajit Sinha, Michael J Buck
Abstract: The Transcription Factor (TF) p63 is a master regulator of epidermal development and differentiation as evident from the remarkable skin phenotype of p63 mouse knockouts. Furthermore, ectopic expression of p63 alone is sufficient to convert simple epithelium into stratified epithelial tissues in vivo and p63 is required for efficient transdifferentiation of fibroblasts into keratinocytes. However, little is known about the molecular mechanisms of p63 function, in particular how it selects its target sites in the genome. p63, which acts both as an activator and repressor of transcription, recognizes a canonical binding motif that occurs over 1 million times in the human genome. But, in human keratinocytes less than 12,000 of these sites are bound in vivo suggesting that underlying chromatin architecture and cooperating TFs mediate p63-genome interactions. We find that the chromatin architecture at p63-bound targets possess distinctive features and can be used to categorize p63 targets into proximal promoters (1%), enhancers (59%) and repressed or inactive (40%) regulatory elements. Our analysis shows that the chromatin modifications H3K4me1, H3K27me3, along with overall chromatin accessibility status can accurately predict bonafide p63-bound sites without a priori DNA sequence information. Interestingly, however there exists a qualitative correlation between the p63 binding motif and accessibility and H3K4me1 levels. Furthermore, we use a comprehensive in silico approach that leverages ENCODE data to identify several known TFs such as AP1, AP2 and novel TFs (RFX5 for e.g.) that can potentially cooperate with p63 to modulate its myriad biological functions in keratinocytes. Our analysis shows that p63 bound genomic locations in keratinocytes are accessible, marked by active histone modifications, and co-targeted by other developmentally important transcriptional regulators. Collectively, our results suggest that p63 might actively remodel and/or influence chromatin dynamics at its target sites and in the process dictate its own DNA binding and possibly that of adjacent TFs.
PubMedId: 25433490
Journal-article: BMC Genomics, vol. 15, pp. 1042, 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: 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: Monica M. Laronda, Kenji Unno, Kazutomo Ishi, Vanida A. Serna, Lindsey M. Butler, Alea A. Mills, Grant D. Orvis, Richard R. Behringer, Chuxia Deng, Satrajit Sinha, Takeshi Kurita
Journal-article: Developmental Biology, vol. 381, pp. 5-16, 2013
Authors: S-Y Kim, M H Cordeiro, V A Serna, K Ebbert, L M Butler, S Sinha, A A Mills, T K Woodruff, T Kurita
Journal-article: Cell Death & Differentiation, vol. 20, pp. 987-997, 2013
Authors: E M Alexandrova, O Petrenko, A Nemajerova, R-A Romano, S Sinha, U M Moll
Abstract: Somatic cells can be converted into induced pluripotent stem cells (iPSCs) by forced expression of various combinations of transcription factors, but the molecular mechanisms of reprogramming are poorly understood. Specifically, evidence that the reprogramming process can take many distinct routes only begins to emerge. It is definitively established that p53 deficiency greatly enhances reprogramming, revealing p53's barrier function for induced pluripotency, but the role of its homologs p63 and p73 are unknown. Here we report that in stark contrast to p53, p73 has no role in reprogramming. However, p63 is an enabling (rather than a barrier) factor for Oct4, Sox2 and Klf4 (OSK) and Oct4 and Sox2 (OS), but not for Oct4 and Klf4 (OK) reprogramming of mouse embryonic fibroblasts. Specifically, p63 is essential during reprogramming for maximum efficiency, albeit not for the ability to reprogram per se, and is dispensable for maintaining stability and pluripotency of established iPSC colonies. Np63, but not TAp63, is the principal isoform involved. Loss of p63 can affect reprogramming via several mechanisms such as reduced expression of mesenchymal-epithelial transition and pluripotency genes, hypoproliferation and loss of the most reprogrammable cell populations. During OSK and OS reprogramming, different mechanisms seem to be critical, such as regulation of epithelial and pluripotency genes in OSK reprogramming versus regulation of proliferation in OS reprogramming. Finally, our data reveal three different routes of reprogramming by OSK, OS or OK, based on their differential p63 requirements for iPSC efficiency and pluripotency marker expression. This supports the concept that many distinct routes of reprogramming exist.
PubMedId: 24013722
Journal-article: Cell Death & Differentiation, vol. 20, pp. 1698-1708, 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: S. S. Chin, R.-A. Romano, P. Nagarajan, S. Sinha, L. A. Garrett-Sinha
Keywords: Differentiation Epidermis Notch Transcription Transgenic mice
Abstract: The transcription factor Ets1 is expressed at low levels in epidermal keratinocytes under physiological conditions, but is over-expressed in cutaneous squamous cell carcinoma (SCC). We previously showed that over-expression of Ets1 in differentiated keratinocytes of the skin leads to significant pro-tumorigenic alterations. Here, we further extend these studies by testing the effects of over-expressing Ets1 in the proliferative basal keratinocytes of the skin, which includes the putative epidermal stem cells. We show that induction of the Ets1 transgene in the basal layer of skin during embryogenesis results in epidermal hyperplasia and impaired differentiation accompanied by attenuated expression of spinous and granular layer markers. A similar hyper-proliferative skin phenotype was observed when the transgene was induced in the basal layer of the skin of adult mice leading to hair loss and open sores. The Ets1-mediated phenotype is accompanied by a variety of changes in gene expression including alterations in Notch signaling, a crucial mediator of normal skin differentiation. Finally, we show that Ets1 disrupts Notch signaling in part via its ability to upregulate Np63, an established transcriptional repressor of several of the Notch receptors. Given the established tumor suppressive role for Notch signaling in skin tumorigenesis, the demonstrated ability of Ets1 to interfere with this signaling pathway may be important in mediating its pro-tumorigenic activities.
PubMedId: 24337118
Journal-article: Biology Open, vol. 2, pp. 1336-1345, 2013
Authors: Ari Jon Arason, Hulda R Jonsdottir, Skarphedinn Halldorsson, Berglind Eva Benediktsdottir, Jon Thor Bergthorsson, Saevar Ingthorsson, Olafur Baldursson, Satrajit Sinha, Thorarinn Gudjonsson, Magnus K Magnusson
PubMedId: 24533135
1: PloS one, vol. 9, 2013
Authors: Briana Lee, Alvaro Villarreal-Ponce, Magid Fallahi, Jeremy Ovadia, Peng Sun, Qian-Chun Yu, Seiji Ito, Satrajit Sinha, Qing Nie, Xing Dai
PubMedId: 24735878
1: Developmental cell, vol. 29, 2013
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: 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: Rose-Anne Romano, Kirsten Smalley, Caitlin Magraw, Vanida Ann Serna, Takeshi Kurita, Srikala Raghavan, Satrajit Sinha
Journal-article: Journal of Cell Science, vol. 125, pp. e1.1-e1, 2012
Authors: R.-A. Romano, K. Smalley, C. Magraw, V. A. Serna, T. Kurita, S. Raghavan, S. Sinha
Journal-article: Development, vol. 139, pp. 772-782, 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: 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: Monica M. Laronda, Kenji Unno, Lindsey M. Butler, Vanida A. Serna, Alea A. Mills, Richard R. Behringer, Chuxia Deng, Satrajit Sinha, Takeshi Kurita
Journal-article: Biology of Reproduction, vol. 87, pp. 147-147, 2012
Authors: H. Lu, X. Yang, P. Duggal, C. T. Allen, B. Yan, J. Cohen, L. Nottingham, R.-A. Romano, S. Sinha, K. E. King, W. C. Weinberg, Z. Chen, C. Van Waes
Journal-article: Cancer Research, vol. 71, pp. 6867-6877, 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: X. Yang, H. Lu, B. Yan, R.-A. Romano, Y. Bian, J. Friedman, P. Duggal, C. Allen, R. Chuang, R. Ehsanian, H. Si, S. Sinha, C. Van Waes, Z. Chen
Abstract: Head and neck squamous cell carcinoma (HNSCC) and many other epithelial malignancies exhibit increased proliferation, invasion, and inflammation, concomitant with aberrant nuclear activation of TP53 and NF-B family members Np63, cRel, and RelA. However, the mechanisms of cross-talk by which these transcription factors coordinate gene expression and the malignant phenotype remain elusive. In this study, we showed that Np63 regulates a cohort of genes involved in cell growth, survival, adhesion, and inflammation, which substantially overlaps with the NF-B transcriptome. Np63 with cRel and/or RelA are recruited to form novel binding complexes on p63 or NF-B/Rel sites of multitarget gene promoters. Overexpressed Np63- or TNF--induced NF-B and inflammatory cytokine interleukin-8 (IL-8) reporter activation depended on RelA/cRel regulatory binding sites. Depletion of RelA or Np63 by small interfering RNA (siRNA) significantly inhibited NF-B-specific, or TNF--induced IL-8 reporter activation. Np63 siRNA significantly inhibited proliferation, survival, and migration by HNSCC cells in vitro. Consistent with these data, an increase in nuclear Np63, accompanied by increased proliferation (Ki-67) and adhesion (4 integrin) markers, and induced inflammatory cell infiltration was observed throughout HNSCC specimens, when compared with the basilar pattern of protein expression and minimal inflammation seen in nonmalignant mucosa. Furthermore, overexpression of Np63 in squamous epithelial cells in transgenic mice leads to increased suprabasilar cRel, Ki-67, and cytokine expression, together with epidermal hyperplasia and diffuse inflammation, similar to HNSCC. Our study reveals Np63 as a master transcription factor that, in coordination with NF-B/Rels, orchestrates a broad gene program promoting epidermal hyperplasia, inflammation, and the malignant phenotype of HNSCC.
PubMedId: 21576089
Journal-article: Cancer Research, vol. 71, pp. 3688-3700, 2011
Authors: Priyadharsini Nagarajan, Shu Shien Chin, Dan Wang, Song Liu, Satrajit Sinha, Lee Ann Garrett-Sinha
Abstract: The transcription factor Ets1 is normally expressed in the proliferative layer of stratified epithelium, but expression of Ets1 is significantly upregulated in squamous cell carcinomas. How elevated levels of Ets1 impact tumor initiation and progression is not well understood. To determine the biological consequences of overexpression of Ets1, we developed a transgenic mouse model that allows induction of Ets1 expression in keratinocytes of stratified epithelium in a regulatable fashion. Induction of Ets1 during embryonic development results in a dramatic alteration in epidermal structure and function by suppressing the expression of multiple stratum corneum constituents, while at the same time inducing expression of EGF ligands, AP1 transcription factors and matrix metalloproteases. Interestingly, expression of certain immune-related genes, including defensins, chemokines and cytokines was increased as well, suggesting a possible role for immune dysregulation in the promotion of squamous dysplasia. Experiments using cultured mouse keratinocytes indicate that Ets1 can induce expression of some of these mediators in a cell-intrinsic fashion. Collectively, our data reveal that elevated expression of Ets1 has a much broader array of pro-tumorigenic effects on epithelial cells than previously appreciated.
PubMedId: 20930145
Journal-article: Journal of cell science, vol. 123, 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, 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: R. A. Romano, K. Smalley, S. Liu, S. Sinha
Journal-article: Development, vol. 137, pp. 1775-1775, 2010
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: Kori Ortt, Satrajit Sinha
Abstract: Protein-DNA interactions, such as those that are necessary for transcription, are critical in regulating cellular function and behavior. The identification of DNA sequences that interact with transcriptional regulatory proteins is an important step necessary to better understand the molecular mechanisms regulating gene expression. Chromatin immunoprecipitation (ChIP) is one such procedure that provides a snapshot of which transcription factors are occupying specific DNA sequences. This method allows one not only to determine whether a particular genomic region is occupied by transcription factors but also to identify specific regulatory sequences that potentially control expression of their target genes. Recently, ChIP has been combined with both microarray analysis and a new generation of sequencing allowing a true genome-wide examination of transcription factor binding. Identifying the exact DNA sequence that a transcriptional regulatory protein binds, the precise timing of this association, and what other factors are involved in these interactions are important steps that will shed light on the transcriptional control mechanisms that dictate the biology of all cells, including keratinocytes.
PubMedId: 19908003
Journal-article: Methods in molecular biology (Clifton, N.J.), vol. 585, 2009
Authors: L W Solomon, P C Stark, L Winter, V Kumar, S Sinha
Abstract: To develop a novel test for chronic ulcerative stomatitis (CUS), a chronic immunologically mediated condition that produces oral ulcerations. Current diagnostic methods require expensive and technically demanding in situ immunofluorescence (IF) studies. An Enzyme-Linked ImmunoSorbent Assay (ELISA) was prepared and tested with serum samples from patients with CUS and negative controls. The N-terminal portion of the CUS autoantigen, DeltaNp63alpha, was produced as a purified recombinant protein and used to coat ELISA plates. Sera from 25 patients with CUS and 16 negative controls were analyzed for reactive antibodies. The optimal cut-offs for positive and negative samples were determined. The optimal cut-off of 0.236 resulted in a sensitivity and specificity of the ELISA of 0.80 and 0.75, respectively (exact 95% confidence intervals, P-value of <0.001). The ELISA developed in this study provides a novel and reliable diagnostic assessment to distinguish CUS from other oral ulcerative diseases. Immunoassay will allow the true incidence and prevalence of CUS to be determined in future studies. When combined with clinical correlations, the ELISA results will facilitate the evaluation of the prognostic utility of antibody titers and allow correlation with treatment responses in individual CUS cases.
PubMedId: 19682320
Journal-article: Oral diseases, vol. 16, 2009
Authors: Silvia Pozzi, Michael Boergesen, Satrajit Sinha, Susanne Mandrup, Roberto Mantovani
Abstract: p63 is a master switch in the complex network of signaling pathways controlling the establishment and maintenance of stratified epithelia. We provide evidence that peroxisome proliferator-activated receptor-alpha (PPARalpha), a ligand-activated nuclear receptor that participates in the skin wound healing process, is a target of p63 in human keratinocytes. Silencing of p63 by RNA interference and transient transfections showed that p63 represses PPARalpha through a functional region of promoter B. Chromatin immunoprecipitation analyses indicate that p63 is bound to this region, in the absence of a recognizable p63-binding motif, suggesting that it acts through interactions with other transcription factors (TFs). Distinct PPARalpha transcripts are differentially regulated by p63, indicating a bimodal action in promoter and/or transcription start specification. PPARalpha repression is consistent with lack of expression in the interfollicular epidermis under physiological conditions. Furthermore, we show that PPARalpha is a negative regulator of DeltaNp63alpha levels and that it also binds to a functional region of the DeltaNp63 promoter that lacks PPRE motifs. Therefore, the reciprocal regulation is exerted either through binding to non-consensus sites or through interactions with other DNA-bound TFs. In conclusion, our data establish a link between two TFs intimately involved in the maintenance of skin homeostatic conditions.
PubMedId: 19458633
Journal-article: The Journal of investigative dermatology, vol. 129, 2009
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: Narasimharao V. Marella, Brandon Seifert, Priyadharsini Nagarajan, Satrajit Sinha, Ronald Berezney
Journal-article: Journal of Cellular Physiology, vol. 221, pp. 139-146, 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: Priyadharsini Nagarajan, Neha Parikh, Lee Ann Garrett-Sinha, Satrajit Sinha
Abstract: Ets1 is an oncogene that functions as a transcription factor and regulates the activity of many genes potentially important for tumor initiation and progression. Interestingly, the Ets1 oncogene is over-expressed in many human squamous cell cancers and over-expression is highly correlated with invasion and metastasis. Thus, Ets1 is believed to mainly play a role in later stages of the oncogenic process, but not early events. To better define the role of Ets1 in squamous cell carcinogenesis, we generated a transgenic mouse model in which expression of the Ets1 oncogene could be temporally and spatially regulated. Upon Ets1 induction in differentiating cells of stratified squamous epithelium, these mice exhibited dramatic changes in epithelial organization including increased proliferation and blocked terminal differentiation. The phenotype was completely reversed when Ets1 expression was suppressed. In mice where Ets1 expression was re-induced at a later age, the phenotype was more localized and the lesions that developed were more invasive. Many potential Ets1 targets were upregulated in the skin of these mice with the most dramatic being the metalloprotease MMP13, which we demonstrate to be a direct transcriptional target of Ets1. Collectively, our data reveal that upregulation of Ets1 can be an early event that promotes pre-neoplastic changes in epidermal tissues via its regulation of key genes driving growth and invasion. Thus, the Ets1 oncogene may be important for oncogenic processes in both early and late stages of tumor development.
PubMedId: 19142229
Journal-article: PloS one, vol. 4, 2008
Authors: Kori Ortt, Eli Raveh, Uri Gat, Satrajit Sinha
Journal-article: Journal of Cellular Biochemistry, vol. 104, pp. 1204-1219, 2008
Authors: Xiaohua Su, Maryline Paris, Young Jin Gi, Kenneth Y Tsai, Min Soon Cho, Yu-Li Lin, Jeffrey A Biernaskie, Satrajit Sinha, Carol Prives, Larysa H Pevny, Freda D Miller, Elsa R Flores
Abstract: The cellular mechanisms that regulate the maintenance of adult tissue stem cells are still largely unknown. We show here that the p53 family member, TAp63, is essential for maintenance of epidermal and dermal precursors and that, in its absence, these precursors senesce and skin ages prematurely. Specifically, we have developed a TAp63 conditional knockout mouse and used it to ablate TAp63 in the germline (TAp63(-/-)) or in K14-expressing cells in the basal layer of the epidermis (TAp63(fl/fl);K14cre+). TAp63(-/-) mice age prematurely and develop blisters, skin ulcerations, senescence of hair follicle-associated dermal and epidermal cells, and decreased hair morphogenesis. These phenotypes are likely due to loss of TAp63 in dermal and epidermal precursors since both cell types show defective proliferation, early senescence, and genomic instability. These data indicate that TAp63 serves to maintain adult skin stem cells by regulating cellular senescence and genomic stability, thereby preventing premature tissue aging.
PubMedId: 19570515
Journal-article: Cell stem cell, vol. 5, 2008
Authors: Yeon Sook Choi, Rumela Chakrabarti, Rosalba Escamilla-Hernandez, Satrajit Sinha
Abstract: The transcription factor Elf5 plays an important role in mammary gland development. However, because of the embryonic lethality of Elf5 straight knockout mice, prior studies have been limited to experiments with Elf5 haploinsufficient animals, overexpression systems or transplants. Here, we have utilized K14-Cre to generate mammary-gland specific Elf5 conditional knockout mice. During pregnancy, Elf5-null mammary epithelium completely failed to initiate alveologenesis, and a characteristic of virgin ductal epithelial cells persisted postpartum. We demonstrate that the loss of Elf5 leads to the absence of alveolar secretory markers confirming previous published data. Interestingly, the developmental block due to a lack of Elf5 could not be restored by multiple gestations. Elf5-null mammary epithelial cells also display disorganized cell structures as evident by altered cell polarities, which might be the cause for collapsed lumina. We observe reduced levels of Stat5 and attenuated Stat5 activity as measured by p-Stat5 levels both in Elf5-null mammary glands as well as cultured mammary epithelial cells. This data suggests that the failure of alveolar and lactogenic differentiation due to the loss of Elf5 is mediated in part due to impaired Stat5 activity. In support of this hypothesis, we show by ChIP experiments that Stat5a promoter contains a conserved Elf5-binding site that is occupied by Elf5 in mammary glands. Mammary epithelia lacking Elf5 exhibited downregulation of several other critical genes involved in alveologenesis, suggesting Elf5 as a master regulator in alveolar development. We propose a model for Elf5-mediated alveolar development, in which Elf5 regulates the expression of key mediators of the PrlR/Jak2/Stat5 signaling pathway.
PubMedId: 19269284
Journal-article: Developmental biology, vol. 329, 2008
Authors: Neha Parikh, Priyadharsini Nagarajan, Matsui Sei-ichi, Satrajit Sinha, Lee Ann Garrett-Sinha
Abstract: To establish an oral epithelial cell line of mouse origin for molecular and biochemical assays. Epithelial cells were isolated from the oral cavity of adult mice and established as a spontaneously immortalized cell line in culture, designated immortalized oral keratinocyte cells (IMOK cells). The cells were then characterized for growth characteristics, differentiation potential, karyotype, transfectability, susceptibility to viral infection and responses to siRNA. The IMOK cells exhibit robust growth in both serum-containing and serum-free medium for at least 100 population doublings. IMOK cells have a near diploid karyotype, express keratinocyte marker proteins and can be induced to undergo differentiation by the addition of high levels of calcium to the medium. The differentiation process is characterized by morphological changes and by the induction of oral epithelium specific differentiation marker proteins such as K4 and K13. Transient transfection analyses reveal that IMOK cells are highly transfectable and that several promoters of epithelial cells are active in these cells. Moreover, upon differentiation with calcium, there is an up-regulation of differentiation-specific K4 and Elf5 promoter activity. Finally, we show that the oral keratinocytes are also amenable to infection with retroviruses and to siRNA-based knockdown of gene expression. Our study is the first to establish an immortalized oral keratinocyte cell line of murine origin that can recapitulate the oral epithelium differentiation program and thus could serve as a useful tool for toxicological and molecular analyses of the oral tissue.
PubMedId: 18721915
Journal-article: Archives of oral biology, vol. 53, 2008
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, Satrajit Sinha
Abstract: The tetracycline (Tet) responsive system is a valuable tool that is routinely used in a wide variety of mammalian cells for regulatable expression of gene products. However, technical difficulties such as harsh selection conditions and extensive screening processes to identify suitably responsive clones limit the generation of stable cell lines. Hence, application of this system in mammalian cells with relatively slow growth rates and/or the capacity to undergo terminal differentiation such as primary mouse keratinocytes is particularly challenging. To our knowledge, no Tet-responsive stable cell lines have been generated from mouse keratinocytes, presumably due to their sensitivity to selection conditions. Our goal was to utilize a modified and robust Tet-expression system to generate a stable primary mouse keratinocyte cell line. These cells could be then utilized for conditional expression of potentially toxic proteins in an inducible fashion. We utilized a eukaryotic promoter instead of a viral promoter to express a modified reverse tetracycline transactivator in mouse keratinocytes and optimized the selection process for generating stable cell lines. Here, we report the generation of a stable mouse keratinocyte cell line for Tet-regulated gene expression with minimal leakiness and high degree of Tet responsivity. This mouse keratinocyte cell line was further engineered for generation of a double stable cell line, which expresses the transcription factor AP-2alpha in an inducible manner. Importantly, the selected cells retain their inherent keratinocyte morphology, respond to differentiation signals and exhibit a persistent and highly tunable Tet-inducibility upon continuous culturing. We have generated a tetracycline inducible gene expression model system in mouse epidermal keratinocytes. Such inducible cell lines will serve as valuable in vitro models for future gain-of-function and loss-of-function studies.
PubMedId: 17964120
Journal-article: Journal of Dermatological Science, vol. 49, pp. 73-84, 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: Regina Landesberg, Matthew Cozin, Serge Cremers, Victoria Woo, Stavroula Kousteni, Satrajit Sinha, LeeAnn Garrett-Sinha, Srikala Raghavan
Abstract: Bisphosphonates (BPs) are a widely used class of drugs that are effective in the treatment and prevention of osteoporosis, hypercalcemia of malignancy, and bone metastases associated with multiple myeloma, breast cancer, and other solid tumors. In the past several years there have been numerous reports describing the occurrence of osteonecrosis of the jaws (ONJ) associated with these drugs. Whether the ONJ lesion initiates in the oral mucosa or derives from the underlying bone is not well understood. In this report we describe the effect of pamidronate, a second-generation BP, on oral mucosal cells. Murine oral keratinocytes were isolated and exposed to pamidronate at a range of clinically relevant doses. Cellular proliferation was measured using a MTS/PMS reagent-based kit and wound healing was examined with a scratch assay. To determine whether oral keratinocytes undergo apoptosis following exposure to pamidronate, TUNEL, caspase-3, and DAPI apoptosis assays were performed. We show that BP pretreatment of oral mucosal cells inhibits proliferation and wound healing at clinically relevant doses, and that this inhibition is not due to cellular apoptosis. To our knowledge this is the first report investigating the effect of nitrogen-containing BPs on oral mucosal cells. This study suggests that BPs inhibit oral keratinocyte wound healing which may play a significant role in the initiation of ONJ.
PubMedId: 18423269
Journal-article: Journal of oral and maxillofacial surgery : official journal of the American Association of Oral and Maxillofacial Surgeons, vol. 66, 2007
Authors: Barbara Birkaya, Kori Ortt, Satrajit Sinha
Abstract: p63 is a transcription factor that plays an important role in skin epidermal development and differentiation. The p63 gene encodes for two major protein isoforms, those containing an amino-terminal trans-activation domain (TAp63) and those lacking this domain (DeltaNp63). Both the TA and DeltaN transcripts are also alternatively spliced at the 3' end producing proteins with unique C-termini that are designated as alpha, beta and gamma isoforms. Recent research has suggested that DeltaNp63 is the predominant isoform expressed and active in keratinocytes. To better elucidate the biological role of p63 in regulating gene expression in keratinocytes we performed chromatin immunoprecipitation (ChIP) experiments with DeltaNp63-specific antibodies. We included an additional step in the ChIP procedure to enrich for DeltaNp63 targets by screening the library of immunoprecipitated DNA for its ability to bind recombinant GST-DeltaNp63. Cloning of DeltaNp63-ChIP-derived DNA fragments identified more than 60 potential DeltaNp63 target loci that were located close to or embedded within known or predicted genes. Identity of these target genes suggests that they may participate in a myriad of cellular processes including transcriptional regulation, signaling and metabolism. Here we confirm the binding of DeltaNp63 to several of these genomic loci both by EMSA and replicate ChIP assays. Finally we show that the expression of many of these target genes is altered when DeltaNp63 levels in keratinocytes are reduced by siRNA, further confirming that these are bona fide targets. This unbiased genomic approach has allowed us to uncover functional targets of DeltaNp63 and serves as the initial step in further analysis of the transcriptional regulatory mechanisms that are governed by p63 in keratinocytes.
PubMedId: 17521434
Journal-article: BMC Molecular Biology, vol. 8, pp. 43, 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: Yeon Sook Choi, Jun Cheng, Julie Segre, Satrajit Sinha
Journal-article: Histochemistry and Cell Biology, vol. 129, pp. 85-94, 2007
Authors: Yeon??Sook Choi, Satrajit Sinha
Abstract: The ESE (epithelium-specific Ets) subfamily of Ets transcription factors plays an important role in regulating gene expression in a variety of epithelial cell types. Although ESE proteins have been shown to bind to regulatory elements of some epithelial genes, the optimal DNA-binding sequence has not been experimentally ascertained for any member of the ESE subfamily of transcription factors. This has made the identification and validation of their targets difficult. We are studying ESE-2 (Elf5), which is highly expressed in epithelial cells of many tissues including skin keratinocytes. Here, we identify the preferred DNA-binding site of ESE-2 by performing CASTing (cyclic amplification and selection of targets) experiments. Our analysis shows that the optimal ESE-2 consensus motif consists of a GGA core and an AT-rich 5'- and 3'-flanking sequences. Mutational and competition experiments demonstrate that the flanking sequences that confer high DNA-binding affinity for ESE-2 show considerable differences from the known consensus DNA-binding sites of other Ets proteins, thus reinforcing the idea that the flanking sequences may impart recognition specificity for Ets proteins. In addition, we have identified a novel isoform of murine ESE-2, ESE-2L, that is generated by use of a hitherto unreported new exon and an alternate promoter. Interestingly, transient transfection assays with an optimal ESE-2 responsive reporter show that both ESE-2 and ESE-2L are weak transactivators. However, similar studies utilizing GAL4 chimaeras of ESE-2 demonstrate that while the DNA-binding ETS (E twenty-six) domain functions as a repressor, the PNT (pointed domain) of ESE-2 can act as a potent transcriptional activation domain. This novel transactivating property of PNT is also shared by ESE-3, another ESE family member. Identification of the ESE-2 consensus site and characterization of the transcriptional activation properties of ESE-2 shed new light on its potential as a regulator of target genes.
PubMedId: 16704374
Journal-article: Biochemical Journal, vol. 398, pp. 497-507, 2006
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: Ramakumar Tummala, Satrajit Sinha
Abstract: Epithelium specific Ets-2 (ESE-2), an epithelium-specific ETS-domain transcription factor, is highly expressed in differentiated keratinocytes. To understand the molecular mechanisms that govern the cell-type and differentiation-specific expression of ESE-2 in keratinocytes, we have focused our studies on the identification and characterization of its cis-regulatory elements. We first performed DNase I hypersensitive site mapping and demonstrated that the promoter region of ESE-2 is in open chromatin conformation in differentiated keratinocytes. Next, we performed transient transfection assays with several 5' serially deleted constructs containing segments of the ESE-2 promoter. These experiments have led to the identification of a short fragment that shows remarkable sequence conservation between several species and harbors most of the transcriptional activity. Interestingly, a high level of transcriptional activity was only observed when the transfected keratinocytes were induced to differentiate by increasing the calcium concentration in the cell-culture medium. To identify the factors that mediate the transcriptional activity, we analyzed this segment by mutational and electrophoretic mobility shift assays (EMSA) experiments. Our studies have identified a critical stretch of nucleotides that is important for both basal as well as calcium responsive reporter activity and that binds to a nuclear factor, keratinocyte restricted factor (KRF). KRF is a novel transcription factor that is restricted to nuclear extracts isolated from keratinocytes and that binds to unique DNA sequences, which do not resemble any known consensus binding motif for transcription factors. Our preliminary experiments shed light on the biochemical nature of KRF and set the stage for future studies in identification of KRF and testing its role in governing ESE-2 gene expression in vivo.
PubMedId: 16229011
Journal-article: Journal of Cellular Biochemistry, vol. 97, pp. 766-781, 2006
Authors: Kori Ortt, Satrajit Sinha
Abstract: p63 is a member of the p53 family of proteins and plays an important role in epithelial development and differentiation. Although some p63 binding sites in the regulatory elements of epithelial genes have been identified, the optimal DNA-binding sequence has not been ascertained for this transcription factor. Here, we identify the preferred DNA-binding site of p63 by performing in vitro DNA selection experiments. Our analysis shows that the optimal p63 DNA-binding consensus motif consists of a CA(T)TG core and an AT-rich 5' and 3' flanking sequence. Gel shift and competition experiments demonstrate that there are specific sequence requirements that confer high DNA-binding affinity for p63 and that significant deviation from the consensus sequences result in poor or no binding. This pattern of DNA-binding is similar for both recombinant p63 and the endogenous protein present in keratinocyte nuclear extracts. Furthermore, we show that the consensus sequence is distinctly different from that of p53, particularly in the flanking sequences. Identification of the p63 consensus DNA-binding sequence will facilitate the validation of in vivo p63-responsive elements that mediate transcriptional regulation of a wide variety of target genes.
PubMedId: 16870177
Journal-article: FEBS Letters, vol. 580, pp. 4544-4550, 2006
Authors: Satrajit Sinha
PubMedId: 15646622
1: Methods in cell biology, vol. 78, 2005
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: C. K. Kaufman, S. Sinha, D. Bolotin, J. Fan, E. Fuchs
Journal-article: Molecular and Cellular Biology, vol. 22, pp. 4293-4308, 2002
Authors: S. Sinha, E. Fuchs
Journal-article: Proceedings of the National Academy of Sciences, vol. 98, pp. 2455-2460, 2001
Authors: S. Sinha, L. Degenstein, C. Copenhaver, E. Fuchs
Journal-article: Molecular and Cellular Biology, vol. 20, pp. 2543-2555, 2000
Authors: I S Kim, S Sinha, B de Crombrugghe, S N Maity
Journal-article: Molecular and Cellular Biology, vol. 16, pp. 4003-4013, 1996
Authors: S Sinha, I S Kim, K Y Sohn, B de Crombrugghe, S N Maity
Journal-article: Molecular and Cellular Biology, vol. 16, pp. 328-337, 1996
Authors: S. Sinha, S. N. Maity, J. Lu, B. de Crombrugghe
Journal-article: Proceedings of the National Academy of Sciences, vol. 92, pp. 1624-1628, 1995
Presented by CUBS © 2024