26 results on '"Tye G. Deering"'
Search Results
2. Sphingolipid metabolism determines the therapeutic efficacy of nanoliposomal ceramide in acute myeloid leukemia
- Author
-
Brian M. Barth, Weiyuan Wang, Paul T. Toran, Todd E. Fox, Charyguly Annageldiyev, Regina M. Ondrasik, Nicole R. Keasey, Timothy J. Brown, Viola G. Devine, Emily C. Sullivan, Andrea L. Cote, Vasiliki Papakotsi, Su-Fern Tan, Sriram S. Shanmugavelandy, Tye G. Deering, David B. Needle, Stephan T. Stern, Junjia Zhu, Jason Liao, Aaron D. Viny, David J. Feith, Ross L. Levine, Hong-Gang Wang, Thomas P. Loughran, Jr, Arati Sharma, Mark Kester, and David F. Claxton
- Subjects
Specialties of internal medicine ,RC581-951 - Published
- 2019
- Full Text
- View/download PDF
3. Ceramide-tamoxifen regimen targets bioenergetic elements in acute myelogenous leukemia1
- Author
-
Samy A.F. Morad, Terence E. Ryan, P. Darrell Neufer, Tonya N. Zeczycki, Traci S. Davis, Matthew R. MacDougall, Todd E. Fox, Su-Fern Tan, David J. Feith, Thomas P. Loughran, Jr., Mark Kester, David F. Claxton, Brian M. Barth, Tye G. Deering, and Myles C. Cabot
- Subjects
ceramides ,sphingolipids ,leukemia ,mitochondria ,Biochemistry ,QD415-436 - Abstract
The objective of our study was to determine the mechanism of action of the short-chain ceramide analog, C6-ceramide, and the breast cancer drug, tamoxifen, which we show coactively depress viability and induce apoptosis in human acute myelogenous leukemia cells. Exposure to the C6-ceramide-tamoxifen combination elicited decreases in mitochondrial membrane potential and complex I respiration, increases in reactive oxygen species (ROS), and release of mitochondrial proapoptotic proteins. Decreases in ATP levels, reduced glycolytic capacity, and reduced expression of inhibitors of apoptosis proteins also resulted. Cytotoxicity of the drug combination was mitigated by exposure to antioxidant. Cells metabolized C6-ceramide by glycosylation and hydrolysis, the latter leading to increases in long-chain ceramides. Tamoxifen potently blocked glycosylation of C6-ceramide and long-chain ceramides. N-desmethyltamoxifen, a poor antiestrogen and the major tamoxifen metabolite in humans, was also effective with C6-ceramide, indicating that traditional antiestrogen pathways are not involved in cellular responses. We conclude that cell death is driven by mitochondrial targeting and ROS generation and that tamoxifen enhances the ceramide effect by blocking its metabolism. As depletion of ATP and targeting the “Warburg effect” represent dynamic metabolic insult, this ceramide-containing combination may be of utility in the treatment of leukemia and other cancers.
- Published
- 2016
- Full Text
- View/download PDF
4. Ceramide nanoliposomes augment the efficacy of venetoclax and cytarabine in models of acute myeloid leukemia
- Author
-
Andrei V. Khokhlatchev, Arati Sharma, Tye G. Deering, Jeremy J. P. Shaw, Pedro Costa‐Pinheiro, Upendarrao Golla, Charyguly Annageldiyev, Myles C. Cabot, Mark R. Conaway, Su‐Fern Tan, Johnson Ung, David J. Feith, Thomas P. Loughran, David F. Claxton, Todd E. Fox, and Mark Kester
- Subjects
Leukemia, Myeloid, Acute ,Sulfonamides ,Genetics ,Cytarabine ,Animals ,Antineoplastic Agents ,Bridged Bicyclo Compounds, Heterocyclic ,Ceramides ,Molecular Biology ,Biochemistry ,Biotechnology - Abstract
Despite several new therapeutic options for acute myeloid leukemia (AML), disease relapse remains a significant challenge. We have previously demonstrated that augmenting ceramides can counter various drug-resistance mechanisms, leading to enhanced cell death in cancer cells and extended survival in animal models. Using a nanoscale delivery system for ceramide (ceramide nanoliposomes, CNL), we investigated the effect of CNL within a standard of care venetoclax/cytarabine (Ara-C) regimen. We demonstrate that CNL augmented the efficacy of venetoclax/cytarabine in in vitro, ex vivo, and in vivo models of AML. CNL treatment induced non-apoptotic cytotoxicity, and augmented cell death induced by Ara-C and venetoclax. Mechanistically, CNL reduced both venetoclax (Mcl-1) and cytarabine (Chk1) drug-resistant signaling pathways. Moreover, venetoclax and Ara-C augmented the generation of endogenous pro-death ceramide species, which was intensified with CNL. Taken together, CNL has the potential to be utilized as an adjuvant therapy to improve outcomes, potentially extending survival, in patients with AML.
- Published
- 2022
5. Sphingolipid metabolism determines the therapeutic efficacy of nanoliposomal ceramide in acute myeloid leukemia
- Author
-
Charyguly Annageldiyev, Nicole R. Keasey, Emily Sullivan, Paul T. Toran, Hong Gang Wang, Regina M. Ondrasik, Su Fern Tan, Andrea L. Cote, Tye G. Deering, Todd E. Fox, David J. Feith, Brian M. Barth, Timothy J Brown, Stephan T. Stern, Mark Kester, David F. Claxton, Thomas P. Loughran, Vasiliki Papakotsi, Arati Sharma, Ross L. Levine, David B. Needle, Junjia Zhu, Sriram S. Shanmugavelandy, Aaron D. Viny, Viola Devine, Weiyuan Wang, and Jason Liao
- Subjects
0301 basic medicine ,Myeloid ,Treatment outcome ,Ceramides ,Vinblastine ,03 medical and health sciences ,Drug Delivery Systems ,0302 clinical medicine ,hemic and lymphatic diseases ,Humans ,Medicine ,neoplasms ,Drug Carriers ,Sphingolipids ,Nanoliposomal Ceramide ,business.industry ,Extramural ,Myeloid leukemia ,Hematology ,medicine.disease ,Stimulus Report ,Antineoplastic Agents, Phytogenic ,Nanostructures ,carbohydrates (lipids) ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Liposomes ,Sphingolipid metabolism ,Cancer research ,lipids (amino acids, peptides, and proteins) ,business ,medicine.drug - Abstract
Key Points Distinct sphingolipid metabolism of AML with MDS-related changes defines unique sensitivity to nanoliposomal C6-ceramide. Vinblastine alters sphingolipid metabolism to enhance the sensitivity of AML to nanoliposomal C6-ceramide.
- Published
- 2019
6. MIST1 and PTF1 Collaborate in Feed-Forward Regulatory Loops That Maintain the Pancreatic Acinar Phenotype in Adult Mice
- Author
-
Ana C. Azevedo-Pouly, Mei Jiang, Galvin H. Swift, Raymond J. MacDonald, David A. Hess, Chinh Q. Hoang, Daniel DiRenzo, Tye G. Deering, and Stephen F. Konieczny
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Endoplasmic reticulum ,Transcription factor complex ,Articles ,Cell Biology ,Biology ,Cell biology ,Chromatin ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Secretory protein ,Transcription (biology) ,Internal medicine ,Acinar cell ,medicine ,Enhancer ,Molecular Biology ,Transcription factor ,030217 neurology & neurosurgery - Abstract
Much remains unknown regarding the regulatory networks formed by transcription factors in mature, differentiated mammalian cells in vivo, despite many studies of individual DNA-binding transcription factors. We report a constellation of feed-forward loops formed by the pancreatic transcription factors MIST1 and PTF1 that govern the differentiated phenotype of the adult pancreatic acinar cell. PTF1 is an atypical basic helix-loop-helix transcription factor complex of pancreatic acinar cells and is critical to acinar cell fate specification and differentiation. MIST1, also a basic helix-loop-helix transcription factor, enhances the formation and maintenance of the specialized phenotype of professional secretory cells. The MIST1 and PTF1 collaboration controls a wide range of specialized cellular processes, including secretory protein synthesis and processing, exocytosis, and homeostasis of the endoplasmic reticulum. PTF1 drives Mist1 transcription, and MIST1 and PTF1 bind and drive the transcription of over 100 downstream acinar genes. PTF1 binds two canonical bipartite sites within a 0.7-kb transcriptional enhancer upstream of Mist1 that are essential for the activity of the enhancer in vivo. MIST1 and PTF1 coregulate target genes synergistically or additively, depending on the target transcriptional enhancer. The frequent close binding proximity of PTF1 and MIST1 in pancreatic acinar cell chromatin implies extensive collaboration although the collaboration is not dependent on a stable physical interaction.
- Published
- 2016
7. Ceramide-tamoxifen regimen targets bioenergetic elements in acute myelogenous leukemia
- Author
-
Thomas P. Loughran, Brian M. Barth, Mark Kester, Matthew R. MacDougall, Su-Fern Tan, Tonya N. Zeczycki, Todd E. Fox, Myles C. Cabot, David F. Claxton, P. Darrell Neufer, Traci S. Davis, Terence E. Ryan, David J. Feith, Tye G. Deering, and Samy A.F. Morad
- Subjects
0301 basic medicine ,Ceramide ,Programmed cell death ,Chemistry ,Cell Biology ,Mitochondrion ,Antiestrogen ,Biochemistry ,Warburg effect ,Sphingolipid ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Endocrinology ,Apoptosis ,medicine ,Cancer research ,Tamoxifen ,medicine.drug - Abstract
The objective of our study was to determine the mechanism of action of the short-chain ceramide analog, C6-ceramide, and the breast cancer drug, tamoxifen, which we show coactively depress viability and induce apoptosis in human acute myelogenous leukemia cells. Exposure to the C6-ceramide-tamoxifen combination elicited decreases in mitochondrial membrane potential and complex I respiration, increases in reactive oxygen species (ROS), and release of mitochondrial proapoptotic proteins. Decreases in ATP levels, reduced glycolytic capacity, and reduced expression of inhibitors of apoptosis proteins also resulted. Cytotoxicity of the drug combination was mitigated by exposure to antioxidant. Cells metabolized C6-ceramide by glycosylation and hydrolysis, the latter leading to increases in long-chain ceramides. Tamoxifen potently blocked glycosylation of C6-ceramide and long-chain ceramides. N-desmethyltamoxifen, a poor antiestrogen and the major tamoxifen metabolite in humans, was also effective with C6-ceramide, indicating that traditional antiestrogen pathways are not involved in cellular responses. We conclude that cell death is driven by mitochondrial targeting and ROS generation and that tamoxifen enhances the ceramide effect by blocking its metabolism. As depletion of ATP and targeting the "Warburg effect" represent dynamic metabolic insult, this ceramide-containing combination may be of utility in the treatment of leukemia and other cancers.
- Published
- 2016
8. Ceramide nanoliposomes as a MLKL-dependent, necroptosis-inducing, chemotherapeutic reagent in ovarian cancer
- Author
-
Masumi Ishibashi, Masafumi Toyoshima, Mark Kester, Tye G. Deering, Nobuo Yaegashi, Toshinori Usui, Todd E. Fox, Mahy Egiz, Junko Minato, Shogo Shigeta, Kazuyuki Kitatani, and Xuewei Zhang
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Ceramide ,Necroptosis ,Cell ,Apoptosis ,Biology ,Ceramides ,Transfection ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Necrosis ,medicine ,Humans ,Ovarian Neoplasms ,Kinase ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,Cancer cell ,Female ,Ovarian cancer ,Protein Kinases - Abstract
Ceramides are bioactive lipids that mediate cell death in cancer cells, and ceramide-based therapy is now being tested in dose-escalating phase I clinical trials as a cancer treatment. Multiple nanoscale delivery systems for ceramide have been proposed to overcome the inherent toxicities, poor pharmacokinetics, and difficult biophysics associated with ceramide. Using the ceramide nanoliposomes (CNL), we now investigate the therapeutic efficacy and signaling mechanisms of this nanoscale delivery platform in refractory ovarian cancer. Treatment of ovarian cancer cells with CNL decreased the number of living cells through necroptosis but not apoptosis. Mechanistically, dying SKOV3 ovarian cancer cells exhibit activation of pseudokinase mixed lineage kinase domain-like (MLKL) as evidenced by oligomerization and relocalization to the blebbing membranes, showing necroptotic characteristics. Knockdown of MLKL, but not its upstream protein kinases such as receptor-interacting protein kinases, with siRNA significantly abolished CNL-induced cell death. Monomeric MLKL protein expression inversely correlated with the IC50 values of CNL in distinct ovarian cancer cell lines, suggesting MLKL as a possible determinant for CNL-induced cell death. Finally, systemic CNL administration suppressed metastatic growth in an ovarian cancer cell xenograft model. Taken together, these results suggest that MLKL is a novel pronecroptotic target for ceramide in ovarian cancer models. Mol Cancer Ther; 17(1); 50–59. ©2017 AACR.
- Published
- 2017
9. The nuclear hormone receptor family member NR5A2 controls aspects of multipotent progenitor cell formation and acinar differentiation during pancreatic organogenesis
- Author
-
Jumin Xue, Galvin H. Swift, Raymond J. MacDonald, Tye G. Deering, Michael A. Hale, Toshi Masui, Youn Kyoung Lee, and Chinh Q. Hoang
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Cellular differentiation ,Population ,Receptors, Cytoplasmic and Nuclear ,Mice, Transgenic ,Enteroendocrine cell ,Acinar Cells ,Biology ,Mice ,Internal medicine ,medicine ,Acinar cell ,Animals ,Cell Lineage ,Transgenes ,Progenitor cell ,education ,Pancreas ,Molecular Biology ,Cell Proliferation ,education.field_of_study ,Base Sequence ,Stem Cells ,Liver receptor homolog-1 ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Stem Cells and Regeneration ,Cell biology ,Phenotype ,medicine.anatomical_structure ,Endocrinology ,Mutation ,Stem cell ,Developmental Biology - Abstract
The orphan nuclear receptor NR5A2 is necessary for the stem-like properties of the epiblast of the pre-gastrulation embryo and for cellular and physiological homeostasis of endoderm-derived organs postnatally. Using conditional gene inactivation, we show that Nr5a2 also plays crucial regulatory roles during organogenesis. During the formation of the pancreas, Nr5a2 is necessary for the expansion of the nascent pancreatic epithelium, for the subsequent formation of the multipotent progenitor cell (MPC) population that gives rise to pre-acinar cells and bipotent cells with ductal and islet endocrine potential, and for the formation and differentiation of acinar cells. At birth, the NR5A2-deficient pancreas has defects in all three epithelial tissues: a partial loss of endocrine cells, a disrupted ductal tree and a >90% deficit of acini. The acinar defects are due to a combination of fewer MPCs, deficient allocation of those MPCs to pre-acinar fate, disruption of acinar morphogenesis and incomplete acinar cell differentiation. NR5A2 controls these developmental processes directly as well as through regulatory interactions with other pancreatic transcriptional regulators, including PTF1A, MYC, GATA4, FOXA2, RBPJL and MIST1 (BHLHA15). In particular, Nr5a2 and Ptf1a establish mutually reinforcing regulatory interactions and collaborate to control developmentally regulated pancreatic genes by binding to shared transcriptional regulatory regions. At the final stage of acinar cell development, the absence of NR5A2 affects the expression of Ptf1a and its acinar specific partner Rbpjl, so that the few acinar cells that form do not complete differentiation. Nr5a2 controls several temporally distinct stages of pancreatic development that involve regulatory mechanisms relevant to pancreatic oncogenesis and the maintenance of the exocrine phenotype.
- Published
- 2014
10. The Role of Ceramide Structure in Regulating the Stability of Membrane Domains
- Author
-
Mark Kester, John Katsaras, Tye G. Deering, Mitchell DiPasquale, Drew Marquardt, and Frederick A. Heberle
- Subjects
Ceramide ,chemistry.chemical_compound ,Membrane ,Chemistry ,Biophysics - Published
- 2019
11. Ceramide nanoliposomes as a MLKL-dependent, necroptosis-inducing, chemotherapeutic reagent in cancer
- Author
-
Takeshi Nabe, Kazuyuki Kitatani, Masaya Matsuda, Mark Kester, Xuewei Zhang, Nobuo Yaegashi, Tye G. Deering, and Yudai Yamamoto
- Subjects
Ceramide ,chemistry.chemical_compound ,chemistry ,Applied Mathematics ,General Mathematics ,Reagent ,Necroptosis ,medicine ,Cancer research ,Cancer ,medicine.disease - Published
- 2019
12. Hemodynamic flow improves rat hepatocyte morphology, function, and metabolic activity in vitro
- Author
-
Brian R. Wamhoff, Ryan E. Feaver, Nicole E. Hastings, Edward L. LeCluyse, Ajit Dash, Timothy L. Pruett, Diana J Berry, Michael B. Simmers, Tye G. Deering, and Brett R. Blackman
- Subjects
Male ,Drug ,medicine.medical_specialty ,Physiology ,media_common.quotation_subject ,Blotting, Western ,Cmax ,Hemodynamics ,Biology ,Cytochrome P-450 Enzyme System ,Microscopy, Electron, Transmission ,In vivo ,Internal medicine ,medicine ,Animals ,media_common ,Reverse Transcriptase Polymerase Chain Reaction ,Articles ,Cell Biology ,Metabolism ,Immunohistochemistry ,Rats, Inbred F344 ,In vitro ,Rats ,medicine.anatomical_structure ,Endocrinology ,Liver ,Hepatocyte ,Toxicity ,Hepatocytes ,Liver Circulation - Abstract
In vitro primary hepatocyte systems typically elicit drug induction and toxicity responses at concentrations much higher than corresponding in vivo or clinical plasma Cmax levels, contributing to poor in vitro-in vivo correlations. This may be partly due to the absence of physiological parameters that maintain metabolic phenotype in vivo. We hypothesized that restoring hemodynamics and media transport would improve hepatocyte architecture and metabolic function in vitro compared with nonflow cultures. Rat hepatocytes were cultured for 2 wk either in nonflow collagen gel sandwiches with 48-h media changes or under controlled hemodynamics mimicking sinusoidal circulation within a perfused Transwell device. Phenotypic, functional, and metabolic parameters were assessed at multiple times. Hepatocytes in the devices exhibited polarized morphology, retention of differentiation markers [E-cadherin and hepatocyte nuclear factor-4α (HNF-4α)], the canalicular transporter [multidrug-resistant protein-2 (Mrp-2)], and significantly higher levels of liver function compared with nonflow cultures over 2 wk (albumin ∼4-fold and urea ∼5-fold). Gene expression of cytochrome P450 (CYP) enzymes was significantly higher (fold increase over nonflow: CYP1A1: 53.5 ± 10.3; CYP1A2: 64.0 ± 15.1; CYP2B1: 15.2 ± 2.9; CYP2B2: 2.7 ± 0.8; CYP3A2: 4.0 ± 1.4) and translated to significantly higher basal enzyme activity (device vs. nonflow: CYP1A: 6.26 ± 2.41 vs. 0.42 ± 0.015; CYP1B: 3.47 ± 1.66 vs. 0.4 ± 0.09; CYP3A: 11.65 ± 4.70 vs. 2.43 ± 0.56) while retaining inducibility by 3-methylcholanthrene and dexamethasone (fold increase over DMSO: CYP1A = 27.33 and CYP3A = 4.94). These responses were observed at concentrations closer to plasma levels documented in vivo in rats. The retention of in vivo-like hepatocyte phenotype and metabolic function coupled with drug response at more physiological concentrations emphasizes the importance of restoring in vivo physiological transport parameters in vitro.
- Published
- 2013
13. Transcriptional Maintenance of Pancreatic Acinar Identity, Differentiation, and Homeostasis by PTF1A
- Author
-
Fong Cheng Pan, Ana C. Azevedo-Pouly, Spencer G. Willet, Hans Peter Elsässer, Christopher V.E. Wright, Chinh Q. Hoang, Raymond J. MacDonald, Michael A. Hale, Mark A. Magnuson, Galvin H. Swift, and Tye G. Deering
- Subjects
0301 basic medicine ,Cell type ,Transcription, Genetic ,Cellular differentiation ,Acinar Cells ,Biology ,03 medical and health sciences ,Gene Knockout Techniques ,Mice ,medicine ,Acinar cell ,Animals ,Homeostasis ,Molecular Biology ,Transcription factor ,Protein Unfolding ,Regulation of gene expression ,Sequence Analysis, RNA ,Gene Expression Profiling ,Cell Differentiation ,Cell Biology ,Articles ,Pancreas, Exocrine ,Cell biology ,Gene expression profiling ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Unfolded protein response ,Unfolded Protein Response ,Pancreas ,Transcription Factors - Abstract
Maintenance of cell type identity is crucial for health, yet little is known of the regulation that sustains the long-term stability of differentiated phenotypes. To investigate the roles that key transcriptional regulators play in adult differentiated cells, we examined the effects of depletion of the developmental master regulator PTF1A on the specialized phenotype of the adult pancreatic acinar cell in vivo. Transcriptome sequencing and chromatin immunoprecipitation sequencing results showed that PTF1A maintains the expression of genes for all cellular processes dedicated to the production of the secretory digestive enzymes, a highly attuned surveillance of unfolded proteins, and a heightened unfolded protein response (UPR). Control by PTF1A is direct on target genes and indirect through a ten-member transcription factor network. Depletion of PTF1A causes an imbalance that overwhelms the UPR, induces cellular injury, and provokes acinar metaplasia. Compromised cellular identity occurs by derepression of characteristic stomach genes, some of which are also associated with pancreatic ductal cells. The loss of acinar cell homeostasis, differentiation, and identity is directly relevant to the pathologies of pancreatitis and pancreatic adenocarcinoma.
- Published
- 2016
14. MIST1 Links Secretion and Stress as both Target and Regulator of the Unfolded Protein Response
- Author
-
Ann-Hwee Lee, Tye G. Deering, David A. Hess, Ana C. Azevedo-Pouly, Stephen F. Konieczny, Anju Karki, Raymond J. MacDonald, Chinh Q. Hoang, Katherine M. Strelau, and Mei Jiang
- Subjects
0301 basic medicine ,XBP1 ,Endoplasmic reticulum ,Cellular differentiation ,Regulator ,Cell Biology ,Articles ,Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Gene expression ,Unfolded protein response ,Transcriptional regulation ,Molecular Biology ,Transcription factor - Abstract
Transcriptional networks that govern secretory cell specialization, including instructing cells to develop a unique cytoarchitecture, amass extensive protein synthesis machinery, and be embodied to respond to endoplasmic reticulum (ER) stress, remain largely uncharacterized. In this study, we discovered that the secretory cell transcription factor MIST1 (Bhlha15), previously shown to be essential for cytoskeletal organization and secretory activity, also functions as a potent ER stress-inducible transcriptional regulator. Genome-wide DNA binding studies, coupled with genetic mouse models, revealed MIST1 gene targets that function along the entire breadth of the protein synthesis, processing, transport, and exocytosis networks. Additionally, key MIST1 targets are essential for alleviating ER stress in these highly specialized cells. Indeed, MIST1 functions as a coregulator of the unfolded protein response (UPR) master transcription factor XBP1 for a portion of target genes that contain adjacent MIST1 and XBP1 binding sites. Interestingly, Mist1 gene expression is induced during ER stress by XBP1, but as ER stress subsides, MIST1 serves as a feedback inhibitor, directly binding the Xbp1 promoter and repressing Xbp1 transcript production. Together, our findings provide a new paradigm for XBP1-dependent UPR regulation and position MIST1 as a potential biotherapeutic for numerous human diseases.
- Published
- 2016
15. Ptf1a-mediated control of Dll1 reveals an alternative to the lateral inhibition mechanism
- Author
-
Jan N. Jensen, Palle Serup, Rasmus Klinck, Mette C. Jørgensen, Raymond J. MacDonald, Jonas Ahnfelt-Rønne, Tye G. Deering, Christopher V.E. Wright, Ole D. Madsen, and Ernst-Martin Füchtbauer
- Subjects
Chromatin Immunoprecipitation ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system ,Pancreatic Polypeptide-Secreting Cells ,Indoles ,Pancreas morphogenesis ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Mice ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Progenitor cell ,HES1 ,Pancreas ,Molecular Biology ,Progenitor ,Homeodomain Proteins ,Regulation of gene expression ,Stem Cells ,Calcium-Binding Proteins ,Galactosides ,Development and Stem Cells ,Immunohistochemistry ,Molecular biology ,Cell biology ,medicine.anatomical_structure ,Bromodeoxyuridine ,Gene Expression Regulation ,embryonic structures ,Intercellular Signaling Peptides and Proteins ,Transcription Factor HES-1 ,Endoderm ,Transcription Factors ,Developmental Biology - Abstract
Neurog3-induced Dll1 expression in pancreatic endocrine progenitors ostensibly activates Hes1 expression via Notch and thereby represses Neurog3 and endocrine differentiation in neighboring cells by lateral inhibition. Here we show in mouse that Dll1 and Hes1 expression deviate during regionalization of early endoderm, and later during early pancreas morphogenesis. At that time, Ptf1a activates Dll1 in multipotent pancreatic progenitor cells (MPCs), and Hes1 expression becomes Dll1 dependent over a brief time window. Moreover, Dll1, Hes1 and Dll1/Hes1 mutant phenotypes diverge during organ regionalization, become congruent at early bud stages, and then diverge again at late bud stages. Persistent pancreatic hypoplasia in Dll1 mutants after eliminating Neurog3 expression and endocrine development, together with reduced proliferation of MPCs in both Dll1 and Hes1 mutants, reveals that the hypoplasia is caused by a growth defect rather than by progenitor depletion. Unexpectedly, we find that Hes1 is required to sustain Ptf1a expression, and in turn Dll1 expression in early MPCs. Our results show that Ptf1a-induced Dll1 expression stimulates MPC proliferation and pancreatic growth by maintaining Hes1 expression and Ptf1a protein levels.
- Published
- 2012
16. LRH-1 and PTF1-L coregulate an exocrine pancreas-specific transcriptional network for digestive function
- Author
-
Steven A. Kliewer, Franciscus J Poelwijk, Raymond J. MacDonald, Galvin H. Swift, Tye G. Deering, Sam R. Holmstrom, and David J. Mangelsdorf
- Subjects
Male ,Chromatin Immunoprecipitation ,Antineoplastic Agents, Hormonal ,genetic processes ,Blotting, Western ,Molecular Sequence Data ,Down-Regulation ,Receptors, Cytoplasmic and Nuclear ,Mice, Transgenic ,Biology ,Research Communication ,Mice ,Gene expression ,Genetics ,medicine ,Animals ,Humans ,natural sciences ,Gene Regulatory Networks ,Secretion ,Transcription factor ,Cholecystokinin ,Mice, Knockout ,Base Sequence ,Sequence Homology, Amino Acid ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Lipase ,Molecular biology ,Pancreas, Exocrine ,Tamoxifen ,medicine.anatomical_structure ,Nuclear receptor ,Cistrome ,Female ,Pancreas ,Chromatin immunoprecipitation ,Peptide Hydrolases ,Transcription Factors ,Developmental Biology - Abstract
We have determined the cistrome and transcriptome for the nuclear receptor liver receptor homolog-1 (LRH-1) in exocrine pancreas. Chromatin immunoprecipitation (ChIP)-seq and RNA-seq analyses reveal that LRH-1 directly induces expression of genes encoding digestive enzymes and secretory and mitochondrial proteins. LRH-1 cooperates with the pancreas transcription factor 1-L complex (PTF1-L) in regulating exocrine pancreas-specific gene expression. Elimination of LRH-1 in adult mice reduced the concentration of several lipases and proteases in pancreatic fluid and impaired pancreatic fluid secretion in response to cholecystokinin. Thus, LRH-1 is a key regulator of the exocrine pancreas-specific transcriptional network required for the production and secretion of pancreatic fluid.
- Published
- 2011
17. Abstract 5832: Sphingolipid metabolism determines the efficacy of nanoliposomal ceramide in acute myeloid leukemia
- Author
-
Todd E. Fox, Charyguly Annageldiyev, Nicole R. Keasey, Ross L. Levine, Megan M. Young, Arati Sharma, David F. Claxton, Tye G. Deering, Hong-Gang Wan Wang, Stephan T. Stern, Junjia Zhu, Mark Kester, Jason Liao, Brian M. Barth, Thomas P. Loughran, Aaron D. Viny, and Viola Devine
- Subjects
Cancer Research ,Ceramide ,business.industry ,Autophagy ,CD34 ,Myeloid leukemia ,Cancer ,medicine.disease ,Sphingolipid ,Vinblastine ,chemistry.chemical_compound ,Leukemia ,Oncology ,chemistry ,hemic and lymphatic diseases ,Cancer research ,Medicine ,business ,medicine.drug - Abstract
Therapeutic advances for the treatment of acute myeloid leukemia (AML) have been limited in part due to the heterogeneity and complexity of the disease and a poor understanding of its underlying biology. The leukemia stem cell (LSC) arguably resists current therapy resulting in relapses for most initially treatment sensitive patients. AML with myelodysplastic syndrome related changes (AML-MRC) highlights this challenge, representing a very poor outlook subset. The present study sought to understand the underlying sphingolipid biology in and AML, and to evaluate the efficacy of nanoliposomal ceramide (Lip-C6). Sphingolipids play essential roles in cell survival and proliferation, as well as stress and death. Lip-C6, which delivers a short-chain analog of the pro-apoptotic sphingolipid ceramide, has been in development as an anticancer therapeutic. The efficacy of Lip-C6 therapy was evaluated in both in vitro and in vivo models using primary AML cells and AML cell lines. Evaluation and characterization of the effect of treatment with Lip-C6 was done through lipidomic, short term assays such as apoptosis, autophagy and colony formation assays. Efficacy of Lip-C6 and vinblastine was tested in patient derived xenograft models and mouse - human cell line xenograft MV-411. NOD SCID gamma (NSG) mice were injected with luciferase/YFP labeled cells and monitored by bioluminescence imaging (BLI) for the leukemia progression and efficacy. Sphingolipid metabolism was observed to be elevated in patient samples with De Novo AML but not those with AML-MRC. Apoptosis induced by Lip-C6 in CD34+ve/CD38-ve “LSCs” was robust in AML-MRC, but limited in De Novo AMLs. Similarly, AML colonies forming cells were more sensitive to Lip-C6 in AML- MRC than in De Novo cases. It was hypothesized that elevated sphingolipid metabolism and the upregulation of pro-survival pathways such as autophagy contributed to Lip-C6 resistance in De Novo AML. Vinblastine, when combined with Lip-C6, focused sphingolipid metabolism towards pro-apoptotic metabolites and blocked autophagy. In-vivo combination of Lip-C6 and vinblastine uniquely yielded long term control of leukemia progression without systemic toxicity, translating in to prolonged overall leukemia free survival compared to single agents. Altogether, this study shows fundamental biological differences in sphingolipid metabolism between De Novo AML and AML-MRC. The combination of Vinblastine and Lip-C6 targets the LSC and yields apparent cure of lethal human xenograft AML. Citation Format: Charyguly Annageldiyev, Arati Sharma, Brian M. Barth, Todd E. Fox, Tye Deering, Viola Devine, Nicole R. Keasey, Stephan T. Stern, Megan M. Young, Hong-Gang Wan Wang, Jason Liao, Junjia Zhu, Aaron D. Viny, Ross L. Levine, Thomas P. Loughran, Mark Kester, David F. Claxton. Sphingolipid metabolism determines the efficacy of nanoliposomal ceramide in acute myeloid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5832.
- Published
- 2018
18. Nanoliposome C6-Ceramide Increases the Anti-tumor Immune Response and Slows Growth of Liver Tumors in Mice
- Author
-
Dai Liu, Todd D. Schell, Kevin F. Staveley-O’Carroll, Guangfu Li, Eric T. Kimchi, Diego M. Avella, Mark Kester, Yariswamy Manjunath, Xinjian Liu, Tye G. Deering, Don C. Rockey, Todd E. Fox, Jussuf T. Kaifi, and Xiaoqiang Qi
- Subjects
Male ,0301 basic medicine ,Time Factors ,Antigens, Polyomavirus Transforming ,medicine.medical_treatment ,HCC Model ,Apoptosis ,CD8-Positive T-Lymphocytes ,Immunotherapy, Adoptive ,Immune Regulation ,0302 clinical medicine ,Neoplasms ,Tumor Microenvironment ,Nanotechnology ,Promoter Regions, Genetic ,Cell Line, Transformed ,Chemistry ,Liver Neoplasms ,Gastroenterology ,Magnetic Resonance Imaging ,Tumor Burden ,3. Good health ,Cytokine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cytokines ,Signal Transduction ,Liver Cancer ,Macrophage colony-stimulating factor ,Carcinoma, Hepatocellular ,Regulatory T cell ,Antineoplastic Agents ,Mice, Transgenic ,Tumor-associated macrophage ,Ceramides ,Article ,03 medical and health sciences ,Immune system ,medicine ,Humans ,Animals ,Cell Proliferation ,Tumor microenvironment ,Hepatology ,Macrophages ,Proteins ,Mice, Inbred C57BL ,030104 developmental biology ,Liposomes ,Cancer cell ,Cancer research ,Myeloid-derived Suppressor Cell ,Nanoparticles ,Tumor Escape ,Reactive Oxygen Species ,Proto-Oncogene Proteins c-akt - Abstract
BACKGROUND & AIMS Ceramide, a sphingolipid metabolite, affects T-cell signaling, induces apoptosis of cancer cells, and slows tumor growth in mice. However, it has not been used as a chemotherapeutic agent because of its cell impermeability and precipitation in aqueous solution. We developed a nanoliposome-loaded C6-ceremide (LipC6) to overcome this limitation and investigated its effects in mice with liver tumors. METHODS Immune competent C57BL/6 mice received intraperitoneal injections of carbon tetrachlo-ride and intra-splenic injections of oncogenic hepatocytes. As a result, tumors resembling human hepatocellular carcinomas developed in a fibrotic liver setting. After tumors formed, mice were given an injection of LipC6 or vehicle via tail vein every other day for 2 weeks. This was followed by administration, also via tail vein, of tumor antigen-specific (TAS) CD8+ T cells isolated from the spleens of line 416 mice, and subsequent immunization by intraperitoneal injection of tumor antigen-expressing B6/WT-19 cells. Tumor growth was monitored with magnetic resonance imaging. Tumor apoptosis, proliferation, and AKT expression were analyzed using immunohistochemistry and immunoblots. Cytokine production, phenotype, and function of TAS CD8+ T cells and tumor-associated macrophages (TAMs) were studied with flow cytometry, real-time polymerase chain reaction (PCR), and ELISA. Reactive oxygen species (ROS) in TAMs and bone marrow-derived macrophages, induced by colony stimulating factor 2 (GMCSF or CSF2) or colony stimulating factor 1 (MCSF or CSF1), were detected using a luminescent assay. RESULTS Injection of LipC6 slowed tumor growth by reducing tumor cell proliferation and phosphorylation of AKT, and increasing tumor cell apoptosis, compared with vehicle. Tumors grew more slowly in mice given the combination of LipC6 injection and TAS CD8+ T cells followed by immunization compared with mice given vehicle, LipC6, the T cells, or immunization alone. LipC6 injection also reduced numbers of TAMs and their production of ROS. LipC6 induced TAMs to differentiate into an M1 phenotype, which reduced immune suppression and increased activity of CD8+ T cells. These results were validated by experiments with bone marrow-derived macrophages induced by GMCSF or MCSF. CONCLUSIONS In mice with liver tumors, injection of LipC6 reduces the number of TAMs and the ability of TAMs to suppress the anti-tumor immune response. LipC6 also increases the anti-tumor effects of TAS CD8+ T cells. LipC6 might therefore increase the efficacy of immune therapy in patients with hepatocellular carcinoma., Graphical Abstract
- Published
- 2018
19. Peroxisome Proliferator-Activated Receptor γ Activation Restores Islet Function in Diabetic Mice through Reduction of Endoplasmic Reticulum Stress and Maintenance of Euchromatin Structure
- Author
-
Carmella Evans-Molina, Raghavendra G. Mirmira, James C. Garmey, Craig S. Nunemaker, Reiesha D. Robbins, Tatsuyoshi Kono, Sarah A. Tersey, George L. Vestermark, Susanna R. Keller, Bernhard Maier, and Tye G. Deering
- Subjects
Blood Glucose ,Agonist ,endocrine system ,medicine.medical_specialty ,SERCA ,endocrine system diseases ,medicine.drug_class ,medicine.medical_treatment ,Peroxisome proliferator-activated receptor ,Biology ,Endoplasmic Reticulum ,Sarcoplasmic Reticulum Calcium-Transporting ATPases ,Euchromatin ,Islets of Langerhans ,Mice ,Downregulation and upregulation ,Mice, Inbred NOD ,Internal medicine ,medicine ,Animals ,Insulin ,Molecular Biology ,Glucose Transporter Type 2 ,Homeodomain Proteins ,chemistry.chemical_classification ,geography ,geography.geographical_feature_category ,Endoplasmic reticulum ,Articles ,Cell Biology ,Islet ,Up-Regulation ,IRS1 ,PPAR gamma ,Endocrinology ,chemistry ,Insulin Receptor Substrate Proteins ,Trans-Activators - Abstract
The nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-gamma) is an important target in diabetes therapy, but its direct role, if any, in the restoration of islet function has remained controversial. To identify potential molecular mechanisms of PPAR-gamma in the islet, we treated diabetic or glucose-intolerant mice with the PPAR-gamma agonist pioglitazone or with a control. Treated mice exhibited significantly improved glycemic control, corresponding to increased serum insulin and enhanced glucose-stimulated insulin release and Ca(2+) responses from isolated islets in vitro. This improved islet function was at least partially attributed to significant upregulation of the islet genes Irs1, SERCA, Ins1/2, and Glut2 in treated animals. The restoration of the Ins1/2 and Glut2 genes corresponded to a two- to threefold increase in the euchromatin marker histone H3 dimethyl-Lys4 at their respective promoters and was coincident with increased nuclear occupancy of the islet methyltransferase Set7/9. Analysis of diabetic islets in vitro suggested that these effects resulting from the presence of the PPAR-gamma agonist may be secondary to improvements in endoplasmic reticulum stress. Consistent with this possibility, incubation of thapsigargin-treated INS-1 beta cells with the PPAR-gamma agonist resulted in the reduction of endoplasmic reticulum stress and restoration of Pdx1 protein levels and Set7/9 nuclear occupancy. We conclude that PPAR-gamma agonists exert a direct effect in diabetic islets to reduce endoplasmic reticulum stress and enhance Pdx1 levels, leading to favorable alterations of the islet gene chromatin architecture.
- Published
- 2009
20. Methyltransferase Set7/9 Maintains Transcription and Euchromatin Structure at Islet-Enriched Genes
- Author
-
Raghavendra G. Mirmira, Bernhard Maier, Takeshi Ogihara, Anthony P. Trace, and Tye G. Deering
- Subjects
Chromatin Immunoprecipitation ,endocrine system ,Transcription, Genetic ,Euchromatin ,Endocrinology, Diabetes and Metabolism ,Immunoblotting ,030209 endocrinology & metabolism ,RNA polymerase II ,Islets of Langerhans ,Mice ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,Transcription (biology) ,Insulin-Secreting Cells ,Commentaries ,Internal Medicine ,Animals ,Protein Methyltransferases ,RNA, Small Interfering ,Promoter Regions, Genetic ,Enhancer ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Promoter ,Histone-Lysine N-Methyltransferase ,Immunohistochemistry ,Molecular biology ,Mice, Inbred C57BL ,Glucose ,Gene Expression Regulation ,DNA methylation ,Histone Methyltransferases ,NIH 3T3 Cells ,biology.protein - Abstract
OBJECTIVE—The activation of β-cell genes, particularly of those encoding preproinsulin, requires an appropriate euchromatin (or “open”) DNA template characterized by hypermethylation of Lys4 of histone H3. We hypothesized that this modification is maintained in islet β-cells by the action of the histone methyltransferase Set7/9. RESEARCH DESIGN AND METHODS—To identify the role of Set7/9, we characterized its expression pattern and gene regulation and studied its function using RNA interference in both cell lines and primary mouse islets. RESULTS—Within the pancreas, Set7/9 protein shows striking specificity for islet cells, including α- and β-cells, as well as occasional cells within ducts. Consistent with these findings, the Set7/9 gene promoter contained an islet-specific enhancer located between −5,768 and −6,030 base pairs (relative to the transcriptional start site) that exhibited Pdx1-responsive activation in β-cells. To study Set7/9 function, we depleted insulinoma cells and primary mouse islets of Set7/9 protein using siRNA. Following siRNA treatment, we observed striking repression of genes involved in glucose-stimulated insulin secretion, including Ins1/2, Glut2, and MafA. These changes in transcription were accompanied by loss of dimethylated H3 Lys4 and RNA polymerase II recruitment, particularly at the Ins1/2 and Glut2 genes. Consistent with these data, depletion of Set7/9 in islets led to defects in glucose-stimulated Ca2+ mobilization and insulin secretion. CONCLUSIONS—We conclude that Set7/9 is required for normal β-cell function, likely through the maintenance of euchromatin structure at genes necessary for glucose-stimulated insulin secretion.
- Published
- 2009
21. A feat of metabolic proportions: Pdx1 orchestrates islet development and function in the maintenance of glucose homeostasis
- Author
-
Raghavendra G. Mirmira, Tye G. Deering, and Daniella A. Babu
- Subjects
endocrine system ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Cellular differentiation ,Biochemistry ,Article ,Neogenesis ,Maturity onset diabetes of the young ,Islets of Langerhans ,Endocrinology ,Internal medicine ,Genetics ,medicine ,Animals ,Homeostasis ,Humans ,Glucose homeostasis ,Molecular Biology ,Homeodomain Proteins ,geography ,geography.geographical_feature_category ,biology ,Pancreatic islets ,biology.organism_classification ,Islet ,medicine.disease ,Glucose ,medicine.anatomical_structure ,Trans-Activators ,PDX1 ,Pancreas - Abstract
Emerging evidence over the past decade indicates a central role for transcription factors in the embryonic development of pancreatic islets and the consequent maintenance of normal glucose homeostasis. Pancreatic and duodenal homeobox 1 (Pdx1) is the best studied and perhaps most important of these factors. Whereas deletion or inactivating mutations of the Pdx1 gene causes whole pancreas agenesis in both mice and humans, even haploinsufficiency of the gene or alterations in its expression in mature islet cells causes substantial impairments in glucose tolerance and the development of a late-onset form of diabetes known as maturity onset diabetes of the young. The study of Pdx1 has revealed crucial phenotypic interrelationships of the varied cell types within the pancreas, particularly as these impinge upon cellular differentiation in the embryo and neogenesis and regeneration in the adult. In this review, we describe the actions of Pdx1 in the developing and mature pancreas and attempt to unify these actions with its known roles in modulating transcriptional complex formation and chromatin structure at the molecular genetic level.
- Published
- 2007
22. Sphingoproteomics: Proteomic Strategies to Examine Sphingolipid Biology
- Author
-
Bruce A. Stanley, Todd E. Fox, and Tye G. Deering
- Subjects
Ceramide ,chemistry.chemical_compound ,chemistry ,Proteome ,Sphingolipid metabolism ,Sphingosine kinase ,Posttranslational modification ,lipids (amino acids, peptides, and proteins) ,Computational biology ,Biology ,Proteomics ,Sphingolipid ,Cell biology - Abstract
Interest in sphingolipids has increased in the past couple of decades as the number of biological activities identified has greatly expanded. These include roles in inflammation, proliferation, survival, and metastasis. Sphingolipids can exert these effects through an increasing number of identified interacting cellular targets. To facilitate the understanding of the intrinsic biology of sphingolipids and the development of sphingolipid-based therapeutics, further knowledge is needed. Various analytical protocols assist this endeavor, with mass spectrometry-based techniques seeing increasing usage, especially for measuring steady-state lipid levels. The area of mass spectrometry-based proteomics is also seeing increased usage in the study of lipid biology. This chapter provides an introduction to hypothesis-generating and hypothesis-testing protein-based analytical approaches to investigate sphingolipids and sphingolipid-metabolizing enzymes. These tools can serve to identify how sphingolipids regulate the proteome, to define how post-translational modifications control enzymatic activity, to identify protein–protein and protein–lipid interactions as well as to facilitate inhibitor development, among other concepts. These approaches can help delineate the roles and consequences of perturbations of sphingolipid metabolism in cancer.
- Published
- 2015
23. Abstract 3083A: C6-ceramide nanoliposomes sensitize paclitaxel resistant ovarian cancer cells to chemotherapy treatment
- Author
-
Samantha G. Sherwood, Mark Kester, Tye G. Deering, Danielle C. Llaneza, John Robert Cornelison, and Charles N. Landen
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,C6-ceramide ,chemistry.chemical_compound ,Paclitaxel ,chemistry ,Internal medicine ,medicine ,Ovarian cancer cells ,business - Abstract
Ovarian cancer is the fifth leading cause of cancer deaths among women, and the majority of women are diagnosed at advanced stages resulting in poorer outcomes and survival rates. One of the driving forces behind these outcomes is the development of chemoresistance, and subsequent proliferation and invasion of tumor cells. In a patient-derived xenograft model of ovarian cancer, we identified the sphingosine pathway as significantly enriched in tumors after treatment with conventional chemotherapy. In order to target the sphingosine pathway, we investigated the potential effects of combining paclitaxel with C6-ceramide nanoliposomes in paclitaxel-resistant ovarian cancer cell lines, HeyA8-MDR and SKOV3-TR. Ceramide has been shown to be a key player in several processes important for tumor suppression including apoptosis, autophagy, and cell cycle arrest. We administered paclitaxel and C6-ceramide to HeyA8-MDR and SKOV3-TR cell lines in vitro and measured cell viability and expression of apoptotic and autophagic proteins. Results showed that in both cell lines, C6-ceramide significantly decreases the IC50 dose of paclitaxel, compared to control ghost nanoliposome treated cells. The IC50 of paclitaxel decreased in HeyA8-MDR cells from 400nM to 150nM, and in SKOV3-TR cells from 1000nM to 450nM. Western blot analyses indicate that combination treatment increases expression of apoptotic (cleaved caspase-3) factors in HeyA8-MDR cells, and autophagic (LC3) factors in SKOV3-TR cells, over single agent treatment or control. Paclitaxel is known to increase expression of apoptotic factors while decreasing expression of autophagic factors. Ceramide may increase both apoptotic and autophagic factors. In an orthotopic mouse model with the HeyA8-MDR line, combination treatment of paclitaxel and C6-ceramide resulted in significantly decreased tumor burden (1.38g +/- 0.74), compared to control ghost (p = 0.022, 2.81g +/- 1.57), C6-ceramide alone (p = 0.078, 2.50g +/- 1.58), and paclitaxel alone (p = 0.005, 2.48g +/- 0.75) groups. The combination of C6-ceramide and paclitaxel may prove useful in elucidating and targeting underlying chemoresistance mechanisms in ovarian cancer. Citation Format: Danielle C. Llaneza, Tye G. Deering, Samantha G. Sherwood, John R. Cornelison, Mark Kester, Charles N. Landen. C6-ceramide nanoliposomes sensitize paclitaxel resistant ovarian cancer cells to chemotherapy treatment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3083A. doi:10.1158/1538-7445.AM2017-3083A
- Published
- 2017
24. Program Specificity for Ptf1a in Pancreas versus Neural Tube Development Correlates with Distinct Collaborating Cofactors and Chromatin Accessibility
- Author
-
Trisha K. Savage, Galvin H. Swift, Raymond J. MacDonald, David Meredith, Chengcheng Shen, Bradford Casey, Manonmani Kumar, Kuang-Chi Tung, Paul R. Mayer, Mark D. Borromeo, Jane E. Johnson, Chinh Q. Hoang, and Tye G. Deering
- Subjects
Neural Tube ,Mice, Transgenic ,Biology ,FOX proteins ,Cell Line ,Mice ,Gene expression ,Consensus Sequence ,Animals ,Humans ,Molecular Biology ,Gene ,Transcription factor ,Pancreas ,ChIA-PET ,Regulation of gene expression ,Genetics ,Base Sequence ,SOXB1 Transcription Factors ,Gene Expression Regulation, Developmental ,Cell Biology ,Articles ,DNA ,Chromatin ,Cell biology ,Immunoglobulin J Recombination Signal Sequence-Binding Protein ,Hepatocyte Nuclear Factor 3-beta ,FOXA2 ,Protein Binding ,Transcription Factors - Abstract
The lineage-specific basic helix-loop-helix transcription factor Ptf1a is a critical driver for development of both the pancreas and nervous system. How one transcription factor controls diverse programs of gene expression is a fundamental question in developmental biology. To uncover molecular strategies for the program-specific functions of Ptf1a, we identified bound genomic regions in vivo during development of both tissues. Most regions bound by Ptf1a are specific to each tissue, lie near genes needed for proper formation of each tissue, and coincide with regions of open chromatin. The specificity of Ptf1a binding is encoded in the DNA surrounding the Ptf1a-bound sites, because these regions are sufficient to direct tissue-restricted reporter expression in transgenic mice. Fox and Sox factors were identified as potential lineage-specific modifiers of Ptf1a binding, since binding motifs for these factors are enriched in Ptf1a-bound regions in pancreas and neural tube, respectively. Of the Fox factors expressed during pancreatic development, Foxa2 plays a major role. Indeed, Ptf1a and Foxa2 colocalize in embryonic pancreatic chromatin and can act synergistically in cell transfection assays. Together, these findings indicate that lineage-specific chromatin landscapes likely constrain the DNA binding of Ptf1a, and they identify Fox and Sox gene families as part of this process.
- Published
- 2013
25. Induced Mist1 expression promotes remodeling of mouse pancreatic acinar cells
- Author
-
Daniel DiRenzo, Barbara Damsz, Chirayu P. Goswami, David A. Hess, J. E. Hallett, Tye G. Deering, Raymond J. MacDonald, Brett Marshall, Yunlong Liu, and Stephen F. Konieczny
- Subjects
Acinar Cells ,Cell fate determination ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,Mice ,Gene expression ,medicine ,Acinar cell ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Secretion ,Progenitor cell ,Transcription factor ,Mice, Knockout ,Hepatology ,Gastroenterology ,Zymogen granule ,Molecular biology ,Pancreas, Exocrine ,Mice, Inbred C57BL ,Microscopy, Electron ,medicine.anatomical_structure ,Pancreas ,Biomarkers ,Signal Transduction - Abstract
Background & Aims Early embryogenesis involves cell fate decisions that define the body axes and establish pools of progenitor cells. Development does not stop once lineages are specified; cells continue to undergo specific maturation events, and changes in gene expression patterns lead to their unique physiological functions. Secretory pancreatic acinar cells mature postnatally to synthesize large amounts of protein, polarize, and communicate with other cells. The transcription factor MIST1 is expressed by only secretory cells and regulates maturation events. MIST1-deficient acinar cells in mice do not establish apical-basal polarity, properly position zymogen granules, or communicate with adjacent cells, disrupting pancreatic function. We investigated whether MIST1 directly induces and maintains the mature phenotype of acinar cells. Methods We analyzed the effects of Cre-mediated expression of Mist1 in adult Mist1 –deficient ( Mist1 KO ) mice. Pancreatic tissues were collected and analyzed by light and electron microscopy, immunohistochemistry, real-time polymerase chain reaction analysis, and chromatin immunoprecipitation. Primary acini were isolated from mice and analyzed in amylase secretion assays. Results Induced expression of Mist1 in adult Mist1 KO mice restored wild-type gene expression patterns in acinar cells. The acinar cells changed phenotypes, establishing apical-basal polarity, increasing the size of zymogen granules, reorganizing the cytoskeletal network, communicating intercellularly (by synthesizing gap junctions), and undergoing exocytosis. Conclusions The exocrine pancreas of adult mice can be remodeled by re-expression of the transcription factor MIST1. MIST1 regulates acinar cell maturation and might be used to repair damaged pancreata in patients with pancreatic disorders.
- Published
- 2011
26. Ptf1a control of Dll1 reveals an alternative to the lateral inhibition mechanism
- Author
-
Mette C. Jørgensen, Christopher V.E. Wright, Rasmus Klinck, Jonas Ahnfelt-Rønne, Raymond J. MacDonald, Ernst-Martin Füchtbauer, Ole D. Madsen, Jan N. Jensen, Tye G. Deering, and Palle Serup
- Subjects
Combinatorics ,Lateral inhibition ,Mistake ,Biology ,Control (linguistics) ,Molecular Biology ,Mechanism (sociology) ,Developmental Biology - Abstract
There was an error published in Development 139 , [33-45][1]. On p. 36, Fig. 1 was incorrectly cited several times in place of Fig. 2. The correct paragraph appears below. The authors apologise to readers for this mistake. To determine if endodermal Notch activation and Hes1 expression depends on
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.