Network


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

Hotspot


Dive into the research topics where Dennis Liang Fei is active.

Publication


Featured researches published by Dennis Liang Fei.


Science Signaling | 2012

The Hedgehog Signal Transduction Network

David J. Robbins; Dennis Liang Fei; Natalia A. Riobo

The Hedgehog network includes canonical and noncanonical signaling modules important for development, homeostasis, and disease. Members of the conserved Hedgehog (Hh) family of secreted proteins play fundamental roles during embryonic development and homeostasis of adult tissues. Hh signal transduction uses an unusual derepression mechanism, in which Hh binding to its receptor component Patched (Ptc) inhibits the capacity of Ptc to repress the heterotrimeric guanine nucleotide–binding protein–coupled receptor Smoothened (Smo). Activation of Smo is accomplished through phosphorylation and subcellular translocation from intracellular vesicles to the plasma membrane and, in vertebrates, also to the primary cilium. This Review focuses primarily on the signaling modules that are set in motion by binding of Hh to Ptc: the “canonical” pathway through which Smo promotes activation of the Gli family of transcription factors, the noncanonical type I pathway in which Ptc modulates cell proliferation and survival independently of Smo, and the noncanonical type II pathway by which Smo regulates the actin cytoskeleton through G inhibitory proteins and small guanosine triphosphatases. Hedgehog (Hh) proteins regulate the development of a wide range of metazoan embryonic and adult structures, and disruption of Hh signaling pathways results in various human diseases. Here, we provide a comprehensive review of the signaling pathways regulated by Hh, consolidating data from a diverse array of organisms in a variety of scientific disciplines. Similar to the elucidation of many other signaling pathways, our knowledge of Hh signaling developed in a sequential manner centered on its earliest discoveries. Thus, our knowledge of Hh signaling has for the most part focused on elucidating the mechanism by which Hh regulates the Gli family of transcription factors, the so-called “canonical” Hh signaling pathway. However, in the past few years, numerous studies have shown that Hh proteins can also signal through Gli-independent mechanisms collectively referred to as “noncanonical” signaling pathways. Noncanonical Hh signaling is itself subdivided into two distinct signaling modules: (i) those not requiring Smoothened (Smo) and (ii) those downstream of Smo that do not require Gli transcription factors. Thus, Hh signaling is now proposed to occur through a variety of distinct context-dependent signaling modules that have the ability to crosstalk with one another to form an interacting, dynamic Hh signaling network.


Nature | 2008

G protein Gαi functions immediately downstream of Smoothened in Hedgehog signalling

Stacey K. Ogden; Dennis Liang Fei; Neal S. Schilling; Yashi Ahmed; John Hwa; David J. Robbins

The hedgehog (Hh) signalling pathway has an evolutionarily conserved role in patterning fields of cells during metazoan development, and is inappropriately activated in cancer. Hh pathway activity is absolutely dependent on signalling by the seven-transmembrane protein smoothened (Smo), which is regulated by the Hh receptor patched (Ptc). Smo signals to an intracellular multi-protein complex containing the Kinesin related protein Costal2 (Cos2), the protein kinase Fused (Fu) and the transcription factor Cubitus interruptus (Ci). In the absence of Hh, this complex regulates the cleavage of full-length Ci to a truncated repressor protein, Ci75, in a process that is dependent on the proteasome and priming phosphorylations by Protein kinase A (PKA). Binding of Hh to Ptc blocks Ptc-mediated Smo inhibition, allowing Smo to signal to the intracellular components to attenuate Ci cleavage. Because of its homology with the Frizzled family of G-protein-coupled receptors (GPCR), a likely candidate for an immediate Smo effector would be a heterotrimeric G protein. However, the role that G proteins may have in Hh signal transduction is unclear and quite controversial, which has led to widespread speculation that Smo signals through a variety of novel G-protein-independent mechanisms. Here we present in vitro and in vivo evidence in Drosophila that Smo activates a G protein to modulate intracellular cyclic AMP levels in response to Hh. Our results demonstrate that Smo functions as a canonical GPCR, which signals through Gαi to regulate Hh pathway activation.


Cancer Research | 2010

Activation of Hedgehog Signaling by the Environmental Toxicant Arsenic May Contribute to the Etiology of Arsenic-Induced Tumors

Dennis Liang Fei; Hua Li; Courtney D. Kozul; Kendall E. Black; Samer Singh; Julie A. Gosse; James DiRenzo; Kathleen A. Martin; Baolin Wang; Joshua W. Hamilton; Margaret R. Karagas; David J. Robbins

Exposure to the environmental toxicant arsenic, through both contaminated water and food, contributes to significant health problems worldwide. In particular, arsenic exposure is thought to function as a carcinogen for lung, skin, and bladder cancer via mechanisms that remain largely unknown. More recently, the Hedgehog signaling pathway has also been implicated in the progression and maintenance of these same cancers. Based on these similarities, we tested the hypothesis that arsenic may act in part through activating Hedgehog signaling. Here, we show that arsenic is able to activate Hedgehog signaling in several primary and established tissue culture cells as well as in vivo. Arsenic activates Hedgehog signaling by decreasing the stability of the repressor form of GLI3, one of the transcription factors that ultimately regulate Hedgehog activity. We also show, using tumor samples from a cohort of bladder cancer patients, that high levels of arsenic exposure are associated with high levels of Hedgehog activity. Given the important role Hedgehog signaling plays in the maintenance and progression of a variety of tumors, including bladder cancer, these results suggest that arsenic exposure may in part promote cancer through the activation of Hedgehog signaling. Thus, we provide an important insight into the etiology of arsenic-induced human carcinogenesis, which may be relevant to millions of people exposed to high levels of arsenic worldwide.


Cancer Research | 2011

Hedgehog-Producing Cancer Cells Respond to and Require Autocrine Hedgehog Activity

Samer Singh; Zhiqiang Wang; Dennis Liang Fei; Kendall E. Black; John A. Goetz; Robert Tokhunts; Camilla Giambelli; Jezabel Rodriguez-Blanco; Jun Long; Ethan Lee; Karoline J. Briegel; Pablo A. Bejarano; Ethan Dmitrovsky; Anthony J. Capobianco; David J. Robbins

A number of Smoothened (SMO) pathway antagonists are currently undergoing clinical trials as anticancer agents. These drugs are proposed to attenuate tumor growth solely through inhibition of Hedgehog (HH), which is produced in tumor cells but acts on tumor stromal cells. The pivotal argument underlying this model is that the growth-inhibitory properties of SMO antagonists on HH-producing cancer cells are due to their off-target effects. Here, we show that the tumorigenic properties of such lung cancer cells depend on their intrinsic level of HH activity. Notably, reducing HH signaling in these tumor cells decreases HH target gene expression. Taken together, these results question the dogma that autocrine HH signaling plays no role in HH-dependent cancers, and does so without using SMO antagonists.


Clinical Immunology | 2014

In utero arsenic exposure and fetal immune repertoire in a US pregnancy cohort.

Kari C. Nadeau; Zhigang Li; Shohreh F. Farzan; Devin C. Koestler; David J. Robbins; Dennis Liang Fei; Meena Malipatlolla; Holden T. Maecker; Richard I. Enelow; Susan A. Korrick; Margaret R. Karagas

Arsenic has wide-ranging effects on human health and there is evidence that it alters the immune response by influencing CD4+/CD8+ T cell ratios, IL-2 cytokine levels, and the expression of immune-response genes. We investigated the impact of in utero environmental arsenic exposure on immune development and function in newborns participating in a pregnancy cohort in New Hampshire, U.S., where arsenic levels have exceeded the current EPA maximum contaminant level of 10 μg/L. Our results showed that maternal urinary arsenic concentrations were inversely related to absolute total CD45RA+ CD4+ cord blood CD69+ T cell counts (N=116, p=0.04) and positively associated with CD45RA+ CD69- CD294+ cell counts (p=0.01). In placental samples (N=70), higher in utero urinary arsenic concentrations were positively associated with the expression of IL1β (p=0.03). These data provide evidence that relatively low-level arsenic exposure in utero may alter the fetal immune system and lead to immune dysregulation.


Cancer Research | 2014

Notch Signaling Drives Stemness and Tumorigenicity of Esophageal Adenocarcinoma

Zhiqiang Wang; Thiago G. da Silva; Ke Jin; Xiaoqin Han; Prathibha Ranganathan; Xiaoxia Zhu; Avencia Sanchez-Mejias; Feng Bai; Bin Li; Dennis Liang Fei; Kelly L. Weaver; Rodrigo Vasquez-Del Carpio; Anna Elizabeth Moscowitz; Vadim P. Koshenkov; Lilly Sanchez; L. Sparling; Xin-Hai Pei; Dido Franceschi; Afonso Ribeiro; David J. Robbins; Alan S. Livingstone; Anthony J. Capobianco

Esophageal adenocarcinoma ranks sixth in cancer mortality in the world and its incidence has risen dramatically in the Western population over the last decades. Data presented herein strongly suggest that Notch signaling is critical for esophageal adenocarcinoma and underlies resistance to chemotherapy. We present evidence that Notch signaling drives a cancer stem cell phenotype by regulating genes that establish stemness. Using patient-derived xenograft models, we demonstrate that inhibition of Notch by gamma-secretase inhibitors (GSI) is efficacious in downsizing tumor growth. Moreover, we demonstrate that Notch activity in a patients ultrasound-assisted endoscopic-derived biopsy might predict outcome to chemotherapy. Therefore, this study provides a proof of concept that inhibition of Notch activity will have efficacy in treating esophageal adenocarcinoma, offering a rationale to lay the foundation for a clinical trial to evaluate the efficacy of GSI in esophageal adenocarcinoma treatment.


Cancer Research | 2012

Hedgehog Signaling Regulates Bladder Cancer Growth and Tumorigenicity

Dennis Liang Fei; Avencia Sanchez-Mejias; Zhiqiang Wang; Colin A. Flaveny; Jun Long; Samer Singh; Jezabel Rodriguez-Blanco; Robert Tokhunts; Camilla Giambelli; Karoline J. Briegel; Wolfgang A. Schulz; A. Jay Gandolfi; Margaret R. Karagas; Teresa A. Zimmers; Merce Jorda; Pablo A. Bejarano; Anthony J. Capobianco; David J. Robbins

The role of Hedgehog (HH) signaling in bladder cancer remains controversial. The gene encoding the HH receptor and negative regulator PATCHED1 (PTCH1) resides on a region of chromosome 9q, one copy of which is frequently lost in bladder cancer. Inconsistent with PTCH1 functioning as a classic tumor suppressor gene, loss-of-function mutations in the remaining copy of PTCH1 are not commonly found. Here, we provide direct evidence for a critical role of HH signaling in bladder carcinogenesis. We show that transformed human urothelial cells and many urothelial carcinoma cell lines exhibit constitutive HH signaling, which is required for their growth and tumorigenic properties. Surprisingly, rather than originating from loss of PTCH1, the constitutive HH activity observed in urothelial carcinoma cell lines was HH ligand dependent. Consistent with this finding, increased levels of HH and the HH target gene product GLI1 were found in resected human primary bladder tumors. Furthermore, on the basis of the difference in intrinsic HH dependence of urothelial carcinoma cell lines, a gene expression signature was identified that correlated with bladder cancer progression. Our findings therefore indicate that therapeutic targeting of the HH signaling pathway may be beneficial in the clinical management of bladder cancer.


Cancer Research | 2014

Pyrvinium Attenuates Hedgehog Signaling Downstream of Smoothened

Bin Li; Dennis Liang Fei; Colin A. Flaveny; Nadia Dahmane; Val erie Baubet; Zhiqiang Wang; Feng Bai; Xin-Hai Pei; Jezabel Rodriguez-Blanco; Brian I. Hang; Darren Orton; Lu Han; Baolin Wang; Anthony J. Capobianco; Ethan Lee; David J. Robbins

The Hedgehog (HH) signaling pathway represents an important class of emerging developmental signaling pathways that play critical roles in the genesis of a large number of human cancers. The pharmaceutical industry is currently focused on developing small molecules targeting Smoothened (Smo), a key signaling effector of the HH pathway that regulates the levels and activity of the Gli family of transcription factors. Although one of these compounds, vismodegib, is now FDA-approved for patients with advanced basal cell carcinoma, acquired mutations in Smo can result in rapid relapse. Furthermore, many cancers also exhibit a Smo-independent activation of Gli proteins, an observation that may underlie the limited efficacy of Smo inhibitors in clinical trials against other types of cancer. Thus, there remains a critical need for HH inhibitors with different mechanisms of action, particularly those that act downstream of Smo. Recently, we identified the FDA-approved anti-pinworm compound pyrvinium as a novel, potent (IC50, 10 nmol/L) casein kinase-1α (CK1α) agonist. We show here that pyrvinium is a potent inhibitor of HH signaling, which acts by reducing the stability of the Gli family of transcription factors. Consistent with CK1α agonists acting on these most distal components of the HH signaling pathway, pyrvinium is able to inhibit the activity of a clinically relevant, vismodegib -resistant Smo mutant, as well as the Gli activity resulting from loss of the negative regulator suppressor of fused. We go on to demonstrate the utility of this small molecule in vivo, against the HH-dependent cancer medulloblastoma, attenuating its growth and reducing the expression of HH biomarkers.


PLOS Genetics | 2016

Wild-Type U2AF1 Antagonizes the Splicing Program Characteristic of U2AF1-Mutant Tumors and Is Required for Cell Survival

Dennis Liang Fei; Hayley Motowski; Rakesh Chatrikhi; Sameer Prasad; Jovian Yu; Shaojian Gao; Clara L. Kielkopf; Robert K. Bradley; Harold Varmus

We have asked how the common S34F mutation in the splicing factor U2AF1 regulates alternative splicing in lung cancer, and why wild-type U2AF1 is retained in cancers with this mutation. A human lung epithelial cell line was genetically modified so that U2AF1S34F is expressed from one of the two endogenous U2AF1 loci. By altering levels of mutant or wild-type U2AF1 in this cell line and by analyzing published data on human lung adenocarcinomas, we show that S34F-associated changes in alternative splicing are proportional to the ratio of S34F:wild-type gene products and not to absolute levels of either the mutant or wild-type factor. Preferential recognition of specific 3′ splice sites in S34F-expressing cells is largely explained by differential in vitro RNA-binding affinities of mutant versus wild-type U2AF1 for those same 3′ splice sites. Finally, we show that lung adenocarcinoma cell lines bearing U2AF1 mutations do not require the mutant protein for growth in vitro or in vivo. In contrast, wild-type U2AF1 is required for survival, regardless of whether cells carry the U2AF1S34F allele. Our results provide mechanistic explanations of the magnitude of splicing changes observed in U2AF1-mutant cells and why tumors harboring U2AF1 mutations always retain an expressed copy of the wild-type allele.


PLOS ONE | 2014

Repurposing the FDA-Approved Pinworm Drug Pyrvinium as a Novel Chemotherapeutic Agent for Intestinal Polyposis

Bin Li; Colin A. Flaveny; Camilla Giambelli; Dennis Liang Fei; Lu Han; Brian I. Hang; Feng Bai; Xin Hai Pei; Vania Nose; Oname Burlingame; Anthony J. Capobianco; Darren Orton; Ethan Lee; David J. Robbins

Mutations in the WNT-pathway regulator ADENOMATOUS POLYPOSIS COLI (APC) promote aberrant activation of the WNT pathway that is responsible for APC-associated diseases such as Familial Adenomatous Polyposis (FAP) and 85% of spontaneous colorectal cancers (CRC). FAP is characterized by multiple intestinal adenomas, which inexorably result in CRC. Surprisingly, given their common occurrence, there are few effective chemotherapeutic drugs for FAP. Here we show that the FDA-approved, anti-helminthic drug Pyrvinium attenuates the growth of WNT-dependent CRC cells and does so via activation of CK1α. Furthermore, we show that Pyrvinium can function as an in vivo inhibitor of WNT-signaling and polyposis in a mouse model of FAP: APCmin mice. Oral administration of Pyrvinium, a CK1α agonist, attenuated the levels of WNT-driven biomarkers and inhibited adenoma formation in APCmin mice. Considering its well-documented safe use for treating enterobiasis in humans, our findings suggest that Pyrvinium could be repurposed for the clinical treatment of APC-associated polyposes.

Collaboration


Dive into the Dennis Liang Fei's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bin Li

University of Miami

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ethan Lee

Vanderbilt University

View shared research outputs
Top Co-Authors

Avatar

Robert K. Bradley

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge