Network


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

Hotspot


Dive into the research topics where Susanne I. Wells is active.

Publication


Featured researches published by Susanne I. Wells.


Nature | 2011

Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro

Jason R. Spence; Christopher N. Mayhew; Scott A. Rankin; Matthew Kuhar; Jefferson Vallance; Kathryn Tolle; Elizabeth E. Hoskins; Vladimir V. Kalinichenko; Susanne I. Wells; Aaron M. Zorn; Noah F. Shroyer; James M. Wells

Studies in embryonic development have guided successful efforts to direct the differentiation of human embryonic and induced pluripotent stem cells (PSCs) into specific organ cell types in vitro. For example, human PSCs have been differentiated into monolayer cultures of liver hepatocytes and pancreatic endocrine cells that have therapeutic efficacy in animal models of liver disease and diabetes, respectively. However, the generation of complex three-dimensional organ tissues in vitro remains a major challenge for translational studies. Here we establish a robust and efficient process to direct the differentiation of human PSCs into intestinal tissue in vitro using a temporal series of growth factor manipulations to mimic embryonic intestinal development. This involved activin-induced definitive endoderm formation, FGF/Wnt-induced posterior endoderm pattering, hindgut specification and morphogenesis, and a pro-intestinal culture system to promote intestinal growth, morphogenesis and cytodifferentiation. The resulting three-dimensional intestinal ‘organoids’ consisted of a polarized, columnar epithelium that was patterned into villus-like structures and crypt-like proliferative zones that expressed intestinal stem cell markers. The epithelium contained functional enterocytes, as well as goblet, Paneth and enteroendocrine cells. Using this culture system as a model to study human intestinal development, we identified that the combined activity of WNT3A and FGF4 is required for hindgut specification whereas FGF4 alone is sufficient to promote hindgut morphogenesis. Our data indicate that human intestinal stem cells form de novo during development. We also determined that NEUROG3, a pro-endocrine transcription factor that is mutated in enteric anendocrinosis, is both necessary and sufficient for human enteroendocrine cell development in vitro. PSC-derived human intestinal tissue should allow for unprecedented studies of human intestinal development and disease.


Frontiers in Bioscience | 2008

Papillomavirus E6 and E7 proteins and their cellular targets.

Susanne I. Wells

The mucosal human papillomaviruses (HPVs) infect human genital and oral epithelial cells and cause lesions ranging in severity from benign to malignant. HPV associated malignancies include cervical and other anogenital cancers as well as a subpopulation of head and neck cancers. Viral infection of epidermal stem or transit amplifying cells can result in long term viral persistence, and the development of carcinogenesis over a significant amount of time then requires additional cooperating genetic hits. Only the so-called high risk HPV types mediate human carcinogenesis, whereas the low risk HPVs have been linked to benign epithelial lesions that are not generally life threatening, but nonetheless are a major health burden. Expression of the high risk HPV E6 and E7 oncogenes is sufficient for primary human keratinocyte immortalization and is required for initiation and all subsequent stages of carcinogenic progression. Together with the finding that high levels of E6/E7 are a unifying hallmark of HPV positive cancers, these two genes are presumed to be the relevant virus-derived transformation stimuli in humans. E6 and E7 proteins do not possess intrinsic enzymatic activities, but instead function though a number of direct and indirect interactions with cellular proteins, a number of which are well known cellular tumor suppressors. We will summarize here current insights into E6 and E7 interactions with specific cellular targets that stimulate aspects of the viral life cycle, interfere with cell cycle controls and promote carcinogenic processes.


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 2011

Alteration of microRNA profiles in squamous cell carcinoma of the head and neck cell lines by human papillomavirus.

Abigail I. Wald; Elizabeth E. Hoskins; Susanne I. Wells; Robert L. Ferris; Saleem A. Khan

Human papillomavirus (HPV)‐positive cases of squamous cell carcinoma of the head and neck (SCCHN) have a much better disease outcome compared to SCCHN cases lacking HPV. Differences in microRNA (miRNA) expression may affect their clinical outcomes.


Journal of Virology | 2005

The Human DEK Proto-Oncogene Is a Senescence Inhibitor and an Upregulated Target of High-Risk Human Papillomavirus E7

Hillary V. Allen; Megan N. Thobe; Elizabeth E. Jones; Kristen B. Habash; Karl Münger; Susanne I. Wells

ABSTRACT The human DEK proto-oncogene is a nucleic acid binding protein with suspected roles in human carcinogenesis, autoimmune disease, and viral infection. Intracellular DEK functions, however, are poorly understood. In papillomavirus-positive cervical cancer cells, downregulation of viral E6/E7 oncogene expression results in cellular senescence. We report here the specific repression of DEK message and protein levels in senescing human papillomavirus type 16- (HPV16-) and HPV18-positive cancer cell lines as well as in primary cells undergoing replicative senescence. Cervical cancer cell senescence was partially overcome by DEK overexpression, and DEK overexpression was sufficient for extending the life span of primary keratinocytes, supporting critical roles for this molecule as a senescence regulator. In order to determine whether DEK is a bona fide HPV oncogene target in primary cells, DEK expression was monitored in human keratinocytes transduced with HPV E6 and/or E7. The results identify high-risk HPV E7 as a positive DEK regulator, an activity that is not shared by low-risk HPV E7 protein. Experiments in mouse embryo fibroblasts recapitulated the observed E7-mediated DEK induction and demonstrated that both basal and E7-induced regulation of DEK expression are controlled by the retinoblastoma protein family. Taken together, our results suggest that DEK upregulation may be a common event in human carcinogenesis and may reflect its senescence inhibitory function.


Molecular and Cellular Biology | 2006

Apoptosis Inhibition by the Human DEK Oncoprotein Involves Interference with p53 Functions

Hillary V. Allen; Elizabeth E. Jones; Kristen B. Habash; Hiroshi Matsuo; Susanne I. Wells

ABSTRACT The DEK proto-oncogene has been associated with human carcinogenesis—either as a fusion with the CAN nucleoporin protein or when transcriptionally upregulated. Mechanisms of intracellular DEK functions, however, have remained relatively unexplored. We have recently demonstrated that DEK expression is induced by the high-risk human papillomavirus (HPV) E7 protein in a manner which is dependent upon retinoblastoma protein function and have implicated DEK in the inhibition of cellular senescence. Additionally, overexpression of DEK resulted in significant life span extension of primary human keratinocytes. In order to determine whether DEK expression is required for cellular proliferation and/or survival, we monitored cellular responses to the knockdown of DEK in cancer and primary cells. The results indicate that DEK expression protects both HPV-positive cancer and primary human cells from apoptotic cell death. Cell death in response to DEK depletion was accompanied by increased protein stability and transcriptional activity of the p53 tumor suppressor and consequent upregulation of known p53 target genes such as p21CIP and Bax. Consistent with a possible role for p53 in DEK-mediated cell death inhibition, the p53-negative human osteosarcoma cell line SAOS-2 was resistant to the knockdown of DEK. Finally, expression of a dominant negative p53 miniprotein inhibited DEK RNA interference-induced p53 transcriptional induction, as well as cell death, thus directly implicating p53 activation in the observed apoptotic phenotype. These findings suggest a novel role for DEK in cellular survival, involving the destabilization of p53 in a manner which is likely to contribute to human carcinogenesis.


Cancer Research | 2009

Overexpression of the cellular DEK protein promotes epithelial transformation in vitro and in vivo

Rachael A. Mintz-Cole; Teresa A. Morris; David S. Simpson; Kathryn A. Wikenheiser-Brokamp; Mark A. Currier; Timothy P. Cripe; Gerard Grosveld; Susanne I. Wells

High levels of expression of the human DEK gene have been correlated with numerous human malignancies. Intracellular DEK functions have been described in vitro and include DNA supercoiling, DNA replication, RNA splicing, and transcription. We have shown that DEK also suppresses cellular senescence, apoptosis, and differentiation, thus promoting cell growth and survival in monolayer and organotypic epithelial raft models. Such functions are likely to contribute to cancer, but direct evidence to implicate DEK as an oncogene has remained elusive. Here, we show that in line with an early role in tumorigenesis, murine papilloma formation in a classical chemical carcinogenesis model was reduced in DEK knockout mice. Additionally, human papillomavirus E6/E7, hRas, and DEK cooperated in the transformation of keratinocytes in soft agar and xenograft establishment, thus also implicating DEK in tumor promotion at later stages. Finally, adenoviral DEK depletion via short hairpin RNA expression resulted in cell death in human tumor cells in vitro and in vivo, but did not significantly affect differentiated epithelial cells. Taken together, our data uncover oncogenic DEK activities as postulated from its frequent up-regulation in human malignancies, and suggest that the targeted suppression of DEK may become a strategic approach to the treatment of cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Transcriptome signature of irreversible senescence in human papillomavirus-positive cervical cancer cells

Susanne I. Wells; Bruce J. Aronow; Trisha M. Wise; Sarah S. Williams; Jennifer A. Couget; Peter M. Howley

A frequent characteristic of human papillomavirus (HPV)-positive cervical cancers is the loss of viral E2 gene expression in HPV-infected cervical epithelial cells as a consequence of viral DNA integration into the cellular genome. The expression of E2 in HPV-positive cancer cells results in the repression of the viral E6/E7 oncogenes, activation of the p53 and pRB pathways, and a G1 cell cycle arrest, followed by induction of cellular senescence. The transcriptional consequences of E2-mediated cell cycle arrest that lead to senescence currently are unknown. Using conditional senescence induction in HeLa cells and microarray analysis, we describe here the expression profile of cells irreversibly committed to senescence. Our results provide insight into the molecular anatomy of senescence pathways and its regulation by HPV on-coproteins. These include the induction of the RAB vesicular transport machinery and a general down-regulation of chromatin regulatory molecules. The repression of tumor-specific G antigens during E2 senescence supports a reversal of the tumorigenic phenotype by E2 and the potential approach of tumor-specific G antigen-specific immunotherapy for cervical cancer.


American Journal of Pathology | 2009

DEK Proto-Oncogene Expression Interferes with the Normal Epithelial Differentiation Program

Richard J. Morreale; Teresa A. Morris; Rachael A. Mintz-Cole; Elizabeth E. Hoskins; Scott J. Balsitis; Nader Husseinzadeh; David P. Witte; Kathryn A. Wikenheiser-Brokamp; Paul F. Lambert; Susanne I. Wells

Overexpression of the DEK gene is associated with multiple human cancers, but its specific roles as a putative oncogene are not well defined. DEK transcription was previously shown to be induced by the high-risk human papillomavirus (HPV) E7 oncogene via E2F and Rb pathways. Transient DEK overexpression was able to inhibit both senescence and apoptosis in cultured cells. In at least the latter case, this mechanism involved the destabilization of p53 and the decreased expression of p53 target genes. We show here that DEK overexpression disrupts the normal differentiation program in a manner that is independent of either p53 or cell death. DEK expression was distinctly repressed upon the differentiation of cultured primary human keratinocytes, and stable DEK overexpression caused epidermal thickening in an organotypic raft model system. The observed hyperplasia involved a delay in keratinocyte differentiation toward a more undifferentiated state, and expansion of the basal cell compartment was due to increased proliferation, but not apoptosis. These phenotypes were accompanied by elevated p63 expression in the absence of p53 destabilization. In further support of bona fide oncogenic DEK activities, we report here up-regulated DEK protein levels in both human papilloma virus-positive hyperplastic murine skin and a subset of human squamous cell carcinomas. We suggest that DEK up-regulation may contribute to carcinoma development at least in part through increased proliferation and retardation of differentiation.


Oncogene | 2011

The human DEK oncogene stimulates β-catenin signaling, invasion and mammosphere formation in breast cancer.

Lisa M. Privette Vinnedge; Rebecca J. McClaine; Purnima K. Wagh; Kathryn A. Wikenheiser-Brokamp; Susan E. Waltz; Susanne I. Wells

Breast cancer is a major cause of cancer-related deaths in American women; therefore, the identification of novel breast cancer-related molecules for the discovery of new markers and drug targets remains essential. The human DEK gene, which encodes a chromatin-binding protein and DNA topology regulator, is upregulated in many types of cancer. DEK has been implicated as an oncogene in breast cancer based on mRNA expression studies, but its functional significance in breast cancer growth and progression has not yet been tested directly. We demonstrate that DEK is highly expressed in breast cancer cells compared with normal tissue, and functionally important for cellular growth, invasion and mammosphere formation. DEK overexpression in non-tumorigenic MCF10A cells resulted in increased growth and motility, with a concomitant downregulation of E-cadherin. Conversely, DEK knockdown in MCF7 and MDA-MB-468 breast cancer cells resulted in decreased growth and motility with upregulation of E-cadherin. The use of DEK-proficient and -deficient breast cancer cells in orthotopic xenografts provided further in vivo evidence that DEK contributes to tumor growth. Activation of the β-catenin signaling pathway is important for normal and cancer stem cell character, growth and metastasis. We show that DEK expression stimulated, and DEK knockdown repressed β-catenin nuclear translocation and activity. Importantly, the expression of constitutively active β-catenin rescued breast cancer invasion defects of DEK knockdown cells. Together, our data indicate that DEK expression stimulates the growth, stem cell character and motility of breast cancer cells, and that DEK-dependent cellular invasion occurs at least in part via β-catenin activation.


Oncogene | 2009

Fanconi anemia deficiency stimulates HPV-associated hyperplastic growth in organotypic epithelial raft culture

Elizabeth E. Hoskins; Teresa A. Morris; Jennifer M. Higginbotham; Nicole Spardy; Elliot Cha; Patrick Kelly; David A. Williams; Kathryn A. Wikenheiser-Brokamp; Stefan Duensing; Susanne I. Wells

Fanconi anemia (FA) is a recessive genome instability syndrome characterized by heightened cellular sensitivity to DNA damage, aplastic anemia and cancer susceptibility. Leukemias and squamous cell carcinomas (SCCs) are the most predominant FA-associated cancers, with the latter exhibiting markedly early disease onset and aggressiveness. Although studies of hematopoietic cells derived from FA patients have provided much insight into bone marrow deficiencies and leukemogenesis, molecular transforming events in FA-deficient keratinocytes, which are the cell type of origin for SCC, are poorly understood. We describe here the growth and molecular properties of FANCA-deficient versus FANCA-corrected HPV E6/E7 immortalized keratinocytes in monolayer and organotypic epithelial raft culture. In response to DNA damage, FANCA-deficient patient-derived keratinocyte cultures displayed a G2/M phase arrest, senescence and apoptosis. Organotypic raft cultures exhibited DNA repair-associated defects with more 53BP1 foci and TdT-mediated dNTP nick end labeling-positive cells over their corrected counterparts. Interestingly, together with reduced rates of DNA damage, FA correction resulted in a marked decrease in epithelial thickness and the presence of fewer cell layers. The observed FANCA-mediated suppression of hyperplasia correlated with the detection of fewer cells transiting through the cell cycle in the absence of gross differentiation abnormalities or apoptotic differences. Importantly, the knockdown of either FANCA or FANCD2 in HPV-positive keratinocytes was sufficient for increasing epithelial hyperplasia. Our findings support a new role for FA pathways in the maintenance of differentiation-dependent cell cycle exit, with the implication that FA deficiencies may contribute to the high risk of FA patients for developing HPV-associated SCC.

Collaboration


Dive into the Susanne I. Wells's collaboration.

Top Co-Authors

Avatar

Elizabeth E. Hoskins

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kathryn A. Wikenheiser-Brokamp

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Parinda A. Mehta

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stella M. Davies

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kasiani C. Myers

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

James M. Wells

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lindsey E. Romick-Rosendale

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Melinda Butsch Kovacic

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Paul F. Lambert

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Lisa M. Privette Vinnedge

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge