Network


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

Hotspot


Dive into the research topics where Philip A. Seymour is active.

Publication


Featured researches published by Philip A. Seymour.


Proceedings of the National Academy of Sciences of the United States of America | 2007

SOX9 is required for maintenance of the pancreatic progenitor cell pool

Philip A. Seymour; Kristine K. Freude; Man N. Tran; Erin Mayes; Jan Jensen; Ralf Kist; Gerd Scherer; Maike Sander

The factors necessary to maintain organ-specific progenitor cells are poorly understood and yet of extreme clinical importance. Here, we identify the transcription factor SOX9 as the first specific marker and maintenance factor of multipotential progenitors during pancreas organogenesis. In the developing pancreas, SOX9 expression is restricted to a mitotically active, Notch-responsive subset of PDX1+ pluripotent progenitors and is absent from committed endocrine precursors or differentiated cells. Similar to Notch mutations, organ-specific Sox9 inactivation in mice causes severe pancreatic hypoplasia resulting from depletion of the progenitor cell pool. We show that Sox9 maintains pancreatic progenitors by stimulating their proliferation, survival, and persistence in an undifferentiated state. Our finding that SOX9 regulates the Notch-effector HES1 suggests a Notch-dependent mechanism and establishes a possible genetic link between SOX factors and Notch. These findings will be of major significance for the development of in vitro protocols for cell replacement therapies.


Development | 2011

Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas.

Janel L. Kopp; Claire L. Dubois; Ashleigh E. Schaffer; Ergeng Hao; Hung Ping Shih; Philip A. Seymour; Jenny Ma; Maike Sander

One major unresolved question in the field of pancreas biology is whether ductal cells have the ability to generate insulin-producing β-cells. Conclusive examination of this question has been limited by the lack of appropriate tools to efficiently and specifically label ductal cells in vivo. We generated Sox9CreERT2 mice, which, during adulthood, allow for labeling of an average of 70% of pancreatic ductal cells, including terminal duct/centroacinar cells. Fate-mapping studies of the Sox9+ domain revealed endocrine and acinar cell neogenesis from Sox9+ cells throughout embryogenesis. Very small numbers of non-β endocrine cells continue to arise from Sox9+ cells in early postnatal life, but no endocrine or acinar cell neogenesis from Sox9+ cells occurs during adulthood. In the adult pancreas, pancreatic injury by partial duct ligation (PDL) has been suggested to induce β-cell regeneration from a transient Ngn3+ endocrine progenitor cell population. Here, we identify ductal cells as a cell of origin for PDL-induced Ngn3+ cells, but fail to observe β-cell neogenesis from duct-derived cells. Therefore, although PDL leads to activation of Ngn3 expression in ducts, PDL does not induce appropriate cues to allow for completion of the entire β-cell neogenesis program. In conclusion, although endocrine cells arise from the Sox9+ ductal domain throughout embryogenesis and the early postnatal period, Sox9+ ductal cells of the adult pancreas no longer give rise to endocrine cells under both normal conditions and in response to PDL.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Sustained Neurog3 expression in hormone-expressing islet cells is required for endocrine maturation and function

Sui Wang; Jan Jensen; Philip A. Seymour; Wei Hsu; Yuval Dor; Maike Sander; Mark A. Magnuson; Palle Serup; Guoqiang Gu

Neurog3 (Neurogenin 3 or Ngn3) is both necessary and sufficient to induce endocrine islet cell differentiation from embryonic pancreatic progenitors. Since robust Neurog3 expression has not been detected in hormone-expressing cells, Neurog3 is used as an endocrine progenitor marker and regarded as dispensable for the function of differentiated islet cells. Here we used 3 independent lines of Neurog3 knock-in reporter mice and mRNA/protein-based assays to examine Neurog3 expression in hormone-expressing islet cells. Neurog3 mRNA and protein are detected in hormone-producing cells at both embryonic and adult stages. Significantly, inactivating Neurog3 in insulin-expressing β cells at embryonic stages or in Pdx1-expressing islet cells in adults impairs endocrine function, a phenotype that is accompanied by reduced expression of several Neurog3 target genes that are essential for islet cell differentiation, maturation, and function. These findings demonstrate that Neurog3 is required not only for initiating endocrine cell differentiation, but also for promoting islet cell maturation and maintaining islet function.


Development | 2012

A Notch-dependent molecular circuitry initiates pancreatic endocrine and ductal cell differentiation

Hung Ping Shih; Janel L. Kopp; Manbir Sandhu; Claire L. Dubois; Philip A. Seymour; Anne Grapin-Botton; Maike Sander

In the pancreas, Notch signaling is thought to prevent cell differentiation, thereby maintaining progenitors in an undifferentiated state. Here, we show that Notch renders progenitors competent to differentiate into ductal and endocrine cells by inducing activators of cell differentiation. Notch signaling promotes the expression of Sox9, which cell-autonomously activates the pro-endocrine gene Ngn3. However, at high Notch activity endocrine differentiation is blocked, as Notch also induces expression of the Ngn3 repressor Hes1. At the transition from high to intermediate Notch activity, only Sox9, but not Hes1, is maintained, thus de-repressing Ngn3 and initiating endocrine differentiation. In the absence of Sox9 activity, endocrine and ductal cells fail to differentiate, resulting in polycystic ducts devoid of primary cilia. Although Sox9 is required for Ngn3 induction, endocrine differentiation necessitates subsequent Sox9 downregulation and evasion from Notch activity via cell-autonomous repression of Sox9 by Ngn3. If high Notch levels are maintained, endocrine progenitors retain Sox9 and undergo ductal fate conversion. Taken together, our findings establish a novel role for Notch in initiating both ductal and endocrine development and reveal that Notch does not function in an on-off mode, but that a gradient of Notch activity produces distinct cellular states during pancreas development.


Developmental Biology | 2008

A Dosage-Dependent Requirement for Sox9 in Pancreatic Endocrine Cell Formation

Philip A. Seymour; Kristine K. Freude; Claire L. Dubois; Hung-Ping Shih; Nisha A. Patel; Maike Sander

We have previously shown the transcription factor SOX9 to be required for the maintenance of multipotential pancreatic progenitor cells in the early embryonic pancreas. However, the association of pancreatic endocrine defects with the Sox9-haploinsufficiency syndrome campomelic dysplasia (CD) implies additional later roles for Sox9 in endocrine development. Using short-term lineage tracing in mice, we demonstrate here that SOX9 marks a pool of multipotential pancreatic progenitors throughout the window of major cell differentiation. During mid-pancreogenesis, both endocrine and exocrine cells simultaneously arise from the SOX9(+) epithelial cords. Our analysis of mice with 50%-reduced Sox9 gene dosage in pancreatic progenitors reveals endocrine-specific defects phenocopying CD. By birth, these mice display a specific reduction in endocrine cell mass, while their exocrine compartment and total organ size is normal. The decrease in endocrine cells is caused by reduced generation of endocrine progenitors from the SOX9(+) epithelium. Conversely, formation of exocrine progenitors is insensitive to reduced Sox9 gene dosage, thus explaining the normal organ size at birth. Our results show that not only is SOX9 required for the maintenance of early pancreatic progenitors, but also governs their adoption of an endocrine fate. Our findings therefore suggest that defective endocrine specification might underlie the pancreatic phenotype of individuals with CD.


Diabetes | 2011

Historical Perspective: Beginnings of the β-Cell: Current Perspectives in β-Cell Development

Philip A. Seymour; Maike Sander

Over the course of the last half-century, we have gleaned much about the developmental biology of the pancreas and in particular the insulin-producing β-cell, the autoimmune destruction of which results in type 1 diabetes. Deciphering the mechanisms driving β-cell neogenesis in vivo holds great allure, in large part because of the potential therapeutic applications that stand to be gained. The field of pancreas development was arguably established by such scientists as Rutter, Grobstein, Wessells, and Cohen in the 1960s. Their seminal studies were the first to demonstrate the significance of the mesenchyme in supporting development of the pancreas or indeed any organ. Their work was further extended by Teitelman and colleagues who did much to characterize pancreatic differentiation via immunohistochemistry. Over the last two decades, the emergence of increasingly elaborate knockout and transgenic mouse technologies has exponentially expanded our insight into the signaling pathways and transcriptional nexus governing pancreas development. Thus, it is fitting that this review will mainly focus on the development of the pancreas and β-cells in the mouse. Through recapitulating endogenous signaling pathways governing β-cell neogenesis in the embryo, it has recently proven possible to generate insulin-producing cells in vitro from human embryonic stem cells (hESCs) (1). Although this milestone accentuates the great therapeutic potential of studying β-cell neogenesis in vivo, the currently insufficient functionality of hESC-derived insulin cells argues the case for further examination of β-cell development in order to understand how and why such engineered cells differ from their endogenous counterparts. It is likely that resolving these differences will lie in better characterizing the relationships between the many signaling pathways and key factors already known to govern the pancreatic program in regards to spatial and temporal pancreatic expression and their impact on pancreatic differentiation. It is envisaged that such incremental knowledge gains will be applied …


Development | 2012

A Sox9/Fgf feed-forward loop maintains pancreatic organ identity

Philip A. Seymour; Hung Ping Shih; Nisha A. Patel; Kristine K. Freude; Ruiyu Xie; Christopher J. Lim; Maike Sander

All mature pancreatic cell types arise from organ-specific multipotent progenitor cells. Although previous studies have identified cell-intrinsic and -extrinsic cues for progenitor cell expansion, it is unclear how these cues are integrated within the niche of the developing organ. Here, we present genetic evidence in mice that the transcription factor Sox9 forms the centerpiece of a gene regulatory network that is crucial for proper organ growth and maintenance of organ identity. We show that pancreatic progenitor-specific ablation of Sox9 during early pancreas development causes pancreas-to-liver cell fate conversion. Sox9 deficiency results in cell-autonomous loss of the fibroblast growth factor receptor (Fgfr) 2b, which is required for transducing mesenchymal Fgf10 signals. Likewise, Fgf10 is required to maintain expression of Sox9 and Fgfr2 in epithelial progenitors, showing that Sox9, Fgfr2 and Fgf10 form a feed-forward expression loop in the early pancreatic organ niche. Mirroring Sox9 deficiency, perturbation of Fgfr signaling in pancreatic explants or genetic inactivation of Fgf10 also result in hepatic cell fate conversion. Combined with previous findings that Fgfr2b or Fgf10 are necessary for pancreatic progenitor cell proliferation, our results demonstrate that organ fate commitment and progenitor cell expansion are coordinately controlled by the activity of a Sox9/Fgf10/Fgfr2b feed-forward loop in the pancreatic niche. This self-promoting Sox9/Fgf10/Fgfr2b loop may regulate cell identity and organ size in a broad spectrum of developmental and regenerative contexts.


Development | 2015

A conserved role for Notch signaling in priming the cellular response to Shh through ciliary localisation of the key Shh transducer Smo

Magdalena Stasiulewicz; Shona D. Gray; Ioanna Mastromina; Joana Clara Silva; Mia Björklund; Philip A. Seymour; David Booth; Calum Thompson; Richard J. Green; Emma A. Hall; Palle Serup; J. Kim Dale

Notochord-derived Sonic Hedgehog (Shh) is essential for dorsoventral patterning of the overlying neural tube. Increasing concentration and duration of Shh signal induces progenitors to acquire progressively more ventral fates. We show that Notch signalling augments the response of neuroepithelial cells to Shh, leading to the induction of higher expression levels of the Shh target gene Ptch1 and subsequently induction of more ventral cell fates. Furthermore, we demonstrate that activated Notch1 leads to pronounced accumulation of Smoothened (Smo) within primary cilia and elevated levels of full-length Gli3. Finally, we show that Notch activity promotes longer primary cilia both in vitro and in vivo. Strikingly, these Notch-regulated effects are Shh independent. These data identify Notch signalling as a novel modulator of Shh signalling that acts mechanistically via regulation of ciliary localisation of key components of its transduction machinery. Highlighted article: Shh signalling controls dorso-ventral cell fate in the neural tube. Notch regulates ciliary architecture and localisation of key Shh pathway components, thus sensitising cells to Shh.


Cell Reports | 2015

A Gene Regulatory Network Cooperatively Controlled by Pdx1 and Sox9 Governs Lineage Allocation of Foregut Progenitor Cells.

Hung Ping Shih; Philip A. Seymour; Nisha A. Patel; Ruiyu Xie; Allen Wang; Patrick P. Liu; Gene W. Yeo; Mark A. Magnuson; Maike Sander

The generation of pancreas, liver, and intestine from a common pool of progenitors in the foregut endoderm requires the establishment of organ boundaries. How dorsal foregut progenitors activate pancreatic genes and evade the intestinal lineage choice remains unclear. Here, we identify Pdx1 and Sox9 as cooperative inducers of a gene regulatory network that distinguishes the pancreatic from the intestinal lineage. Genetic studies demonstrate dual and cooperative functions for Pdx1 and Sox9 in pancreatic lineage induction and repression of the intestinal lineage choice. Pdx1 and Sox9 bind to regulatory sequences near pancreatic and intestinal differentiation genes and jointly regulate their expression, revealing direct cooperative roles for Pdx1 and Sox9 in gene activation and repression. Our study identifies Pdx1 and Sox9 as important regulators of a transcription factor network that initiates pancreatic fate and sheds light on the gene regulatory circuitry that governs the development of distinct organs from multi-lineage-competent foregut progenitors.


PLOS ONE | 2011

Sox9-Haploinsufficiency Causes Glucose Intolerance in Mice

Claire L. Dubois; Hung Ping Shih; Philip A. Seymour; Nisha A. Patel; James M. Behrmann; Victoria Ngo; Maike Sander

The HMG box transcription factor Sox9 plays a critical role in progenitor cell expansion during pancreas organogenesis and is required for proper endocrine cell development in the embryo. Based on in vitro studies it has been suggested that Sox9 controls expression of a network of important developmental regulators, including Tcf2/MODY5, Hnf6, and Foxa2, in pancreatic progenitor cells. Here, we sought to: 1) determine whether Sox9 regulates this transcriptional network in vivo and 2) investigate whether reduced Sox9 gene dosage leads to impaired glucose homeostasis in adult mice. Employing two genetic models of temporally-controlled Sox9 inactivation in pancreatic progenitor cells, we demonstrate that contrary to in vitro findings, Sox9 is not required for Tcf2, Hnf6, or Foxa2 expression in vivo. Moreover, our analysis revealed a novel role for Sox9 in maintaining the expression of Pdx1/MODY4, which is an important transcriptional regulator of beta-cell development. We further show that reduced beta-cell mass in Sox9-haploinsufficient mice leads to glucose intolerance during adulthood. Sox9-haploinsufficient mice displayed 50% reduced beta-cell mass at birth, which recovered partially via a compensatory increase in beta-cell proliferation early postnatally. Endocrine islets from mice with reduced Sox9 gene dosage exhibited normal glucose stimulated insulin secretion. Our findings show Sox9 plays an important role in endocrine development by maintaining Ngn3 and Pdx1 expression. Glucose intolerance in Sox9-haploinsufficient mice suggests that mutations in Sox9 could play a role in diabetes in humans.

Collaboration


Dive into the Philip A. Seymour's collaboration.

Top Co-Authors

Avatar

Maike Sander

University of California

View shared research outputs
Top Co-Authors

Avatar

Palle Serup

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Hung Ping Shih

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nisha A. Patel

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Jensen

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge