27 results on '"Yao, Li-Chin"'
Search Results
2. Neutrophil Dependence of Vascular Remodeling after Mycoplasma Infection of Mouse Airways
- Author
-
Baluk, Peter, Phillips, Keeley, Yao, Li-Chin, Adams, Alicia, Nitschké, Maximilian, and McDonald, Donald M.
- Published
- 2014
- Full Text
- View/download PDF
3. Simultaneous evaluation of treatment efficacy and toxicity for bispecific T‐cell engager therapeutics in a humanized mouse model.
- Author
-
Yang, Jiwon, Jiao, Jing, Draheim, Kyle M., Yang, Guoxiang, Yang, Hongyuan, Yao, Li‐Chin, Shultz, Leonard D., Greiner, Dale L., Rajagopal, Deepa, Vessillier, Sandrine, Maier, Curtis C., Mohanan, Sunish, Cai, Danying, Cheng, Mingshan, Brehm, Michael A., and Keck, James G.
- Published
- 2023
- Full Text
- View/download PDF
4. Plasticity of Button-Like Junctions in the Endothelium of Airway Lymphatics in Development and Inflammation
- Author
-
Yao, Li-Chin, Baluk, Peter, Srinivasan, R. Sathish, Oliver, Guillermo, and McDonald, Donald M.
- Published
- 2012
- Full Text
- View/download PDF
5. Steroid-Resistant Lymphatic Remodeling in Chronically Inflamed Mouse Airways
- Author
-
Yao, Li-Chin, Baluk, Peter, Feng, Jennifer, and McDonald, Donald M.
- Published
- 2010
- Full Text
- View/download PDF
6. α5β1 Integrin Blockade Inhibits Lymphangiogenesis in Airway Inflammation
- Author
-
Okazaki, Tatsuma, Ni, Amy, Ayeni, Oluwasheyi A., Baluk, Peter, Yao, Li-Chin, Vossmeyer, Doerte, Zischinsky, Gunther, Zahn, Grit, Knolle, Jochen, Christner, Claudia, and McDonald, Donald M.
- Published
- 2009
- Full Text
- View/download PDF
7. Development of a Novel Solid Tumor-Bearing Humanized Animal Model for Simultaneously Evaluating the Toxicity and Efficacy of Bispecific T Cell Engager
- Author
-
Yang, Guoxiang, Yao, Li-Chin, Gustafson, Heather, Keck, James G, and Cheng, Mingshan
- Published
- 2023
- Full Text
- View/download PDF
8. TNF-α drives remodeling of blood vessels and lymphatics in sustained airway inflammation in mice
- Author
-
Baluk, Peter, Yao, Li-Chin, Feng, Jennifer, Romano, Talia, Jung, Sonia S., Schreiter, Jessica L., Yan, Li, Shealy, David J., and McDonald, Donald M.
- Subjects
Lymphatics -- Properties -- Models ,Tumor necrosis factor -- Properties -- Models ,Airway obstruction (Medicine) -- Diagnosis -- Care and treatment -- Models ,Health care industry - Abstract
Inflammation is associated with blood vessel and lymphatic vessel proliferation and remodeling. The microvasculature of the mouse trachea provides an ideal opportunity to study this process, as Mycoplasma pulmonis infection of mouse airways induces widespread and sustained vessel remodeling, including enlargement of capillaries into venules and lymphangiogenesis. Although the mediators responsible for these vascular changes in mice have not been identified, VEGF-A is known not to be involved. Here, we sought to determine whether TNF-α drives the changes in blood vessels and lymphatics in M. pulmonis-infected mice. The endothelial cells, but not pericytes, of blood vessels, but not lymphatics, were immunoreactive for TNF receptor 1 (TNF-R1) and lymphotoxin B receptors. Most TNF-R2 immunoreactivity was on leukocytes. Infection resulted in a large and sustained increase in TNF-α expression, as measured by real-time quantitative RT-PCR, and smaller increases in lymphotoxins and TNF receptors that preceded vessel remodeling. Substantially less vessel remodeling and lymphangiogenesis occurred when TNF-α signaling was inhibited by a blocking antibody or was silenced in [Tnfr1.sup.[-/-]] mice. When administered after infection was established, the TNF-α-specific antibody slowed but did not reverse blood vessel remodeling and lymphangiogenesis. The action of TNF-α on blood vessels is probably mediated through direct effects on endothelial cells, but its effects on lymphangiogenesis may require inflammatory mediators from recruited leukocytes. We conclude that TNF-α is a strong candidate for a mediator that drives blood vessel remodeling and lymphangiogenesis in inflammation., Introduction A wide spectrum of changes in blood vessels occurs in inflammation. Acute inflammation is accompanied by reversible vasodilatation, increased blood flow, plasma extravasation, and leukocyte adhesion and transmigration. In [...]
- Published
- 2009
- Full Text
- View/download PDF
9. Enhanced development of functional human NK cells in NOD‐scid‐IL2rgnull mice expressing human IL15.
- Author
-
Aryee, Ken‐Edwin, Burzenski, Lisa M., Yao, Li‐Chin, Keck, James G., Greiner, Dale L., Shultz, Leonard D., and Brehm, Michael A.
- Published
- 2022
- Full Text
- View/download PDF
10. Dynamics of Airway Blood Vessels and Lymphatics: Lessons from Development and Inflammation
- Author
-
McDonald, Donald M., Yao, Li-Chin, and Baluk, Peter
- Published
- 2011
- Full Text
- View/download PDF
11. Rapamycin reversal of VEGF-C-driven lymphatic anomalies in the respiratory tract
- Author
-
Baluk, Peter, Yao, Li-Chin, Flores, Julio C, Choi, Dongwon, Hong, Young-Kwon, and McDonald, Donald M
- Subjects
Vascular Biology - Abstract
Lymphatic malformations are serious but poorly understood conditions that present therapeutic challenges. The goal of this study was to compare strategies for inducing regression of abnormal lymphatics and explore underlying mechanisms. CCSP-rtTA/tetO-VEGF-C mice, in which doxycycline regulates VEGF-C expression in the airway epithelium, were used as a model of pulmonary lymphangiectasia. After doxycycline was stopped, VEGF-C expression returned to normal, but lymphangiectasia persisted for at least 9 months. Inhibition of VEGFR-2/VEGFR-3 signaling, Notch, β-adrenergic receptors, or autophagy and antiinflammatory steroids had no noticeable effect on the amount or severity of lymphangiectasia. However, rapamycin inhibition of mTOR reduced lymphangiectasia by 76% within 7 days without affecting normal lymphatics. Efficacy of rapamycin was not increased by coadministration with the other agents. In prevention trials, rapamycin suppressed VEGF-C-driven mTOR phosphorylation and lymphatic endothelial cell sprouting and proliferation. However, in reversal trials, no lymphatic endothelial cell proliferation was present to block in established lymphangiectasia, and rapamycin did not increase caspase-dependent apoptosis. However, rapamycin potently suppressed Prox1 and VEGFR-3. These experiments revealed that lymphangiectasia is remarkably resistant to regression but is responsive to rapamycin, which rapidly reduces and normalizes the abnormal lymphatics without affecting normal lymphatics.
- Published
- 2017
- Full Text
- View/download PDF
12. Direct interaction of two homeoproteins, Homothorax and Extradenticle, is essential for EXD nuclear localization and function
- Author
-
Jaw, Thomas J, You, Li-Ru, Knoepfler, Paul S, Yao, Li-Chin, Pai, Chi-Yun, Tang, Chiou-Yang, Chang, Li-Ping, Berthelsen, Jens, Blasi, Francesco, Kamps, Mark P, and Sun, Y.Henry
- Published
- 2000
- Full Text
- View/download PDF
13. A Common Mechanism for Antenna-to-Leg Transformation in Drosophila: Suppression of homothorax Transcription by Four HOM-C Genes
- Author
-
Yao, Li-Chin, Liaw, Gwo-Jen, Pai, Chi-Yun, and Sun, Y.Henry
- Published
- 1999
- Full Text
- View/download PDF
14. Pulmonary Lymphangiectasia Resulting from Vegf-C Overexpression During a Critical Period
- Author
-
Yao, Li-Chin, Testini, Chiara, Tvorogov, Denis, Anisimov, Andrey, Vargas, Sara O., Baluk, Peter, Pytowski, Bronislaw, Claesson-Welsh, Lena, Alitalo, Kari, and McDonald, Donald M.
- Subjects
Lung Diseases ,Male ,Lymphangiectasis ,Vascular Endothelial Growth Factor C ,Infant ,Mice, Inbred Strains ,Mice, Transgenic ,Pulmonary Edema ,Vascular Endothelial Growth Factor Receptor-3 ,Vascular Endothelial Growth Factor Receptor-2 ,Article ,Trachea ,Mice ,Pregnancy ,Animals ,Humans ,Uteroglobin ,Female ,Lymphatic Vessels ,Signal Transduction - Abstract
Lymphatic vessels in the respiratory tract normally mature into a functional network during the neonatal period, but under some pathological conditions they can grow as enlarged, dilated sacs that result in the potentially lethal condition of pulmonary lymphangiectasia.We sought to determine whether overexpression of the lymphangiogenic growth factor (vascular endothelial growth factor-C [VEGF-C]) can promote lymphatic growth and maturation in the respiratory tract. Unexpectedly, perinatal overexpression of VEGF-C in the respiratory epithelium led to a condition resembling human pulmonary lymphangiectasia, a life-threatening disorder of the newborn characterized by respiratory distress and the presence of widely dilated lymphatics.Administration of doxycycline to Clara cell secretory protein-reverse tetracycline-controlled transactivator/tetracycline operator-VEGF-C double-transgenic mice during a critical period from embryonic day 15.5 to postnatal day 14 was accompanied by respiratory distress, chylothorax, pulmonary lymphangiectasia, and high mortality. Enlarged sac-like lymphatics were abundant near major airways, pulmonary vessels, and visceral pleura. Side-by-side comparison revealed morphological features similar to pulmonary lymphangiectasia in humans. The condition was milder in mice given doxycycline after age postnatal day 14 and did not develop after postnatal day 35. Mechanistic studies revealed that VEGF recptor (VEGFR)-3 alone drove lymphatic growth in adult mice, but both VEGFR-2 and VEGFR-3 were required for the development of lymphangiectasia in neonates. VEGFR-2/VEGFR-3 heterodimers were more abundant in the dilated lymphatics, consistent with the involvement of both receptors. Despite the dependence of lymphangiectasia on VEGFR-2 and VEGFR-3, the condition was not reversed by blocking both receptors together or by withdrawing VEGF-C.The findings indicate that VEGF-C overexpression can induce pulmonary lymphangiectasia during a critical period in perinatal development.
- Published
- 2014
15. Plasticity of Airway Lymphatics in Development and Disease.
- Author
-
Yao, Li-Chin and McDonald, Donald M.
- Published
- 2014
- Full Text
- View/download PDF
16. Pulmonary Lymphangiectasia Resulting From Vascular Endothelial Growth Factor-C Overexpression During a Critical Period.
- Author
-
Yao, Li-Chin, Testini, Chiara, Tvorogov, Denis, Anisimov, Andrey, Vargas, Sara O., Baluk, Peter, Pytowski, Bronislaw, Claesson-Welsh, Lena, Alitalo, Kari, and McDonald, Donald M.
- Published
- 2014
- Full Text
- View/download PDF
17. Rapid remodeling of airway vascular architecture at birth.
- Author
-
Ni, Amy, Lashnits, Erin, Yao, Li-Chin, Baluk, Peter, and McDonald, Donald M.
- Abstract
Recent advances have documented the development of lung vasculature before and after birth, but less is known of the growth and maturation of airway vasculature. We sought to determine whether airway vasculature changes during the perinatal period and when the typical adult pattern develops. On embryonic day 16.5 mouse tracheas had a primitive vascular plexus unlike the adult airway vasculature, but instead resembling the yolk sac vasculature. Soon after birth (P0), the primitive vascular plexus underwent abrupt and extensive remodeling. Blood vessels overlying tracheal cartilage rings regressed from P1 to P3 but regrew from P4 to P7 to form the hierarchical, segmented, ladder-like adult pattern. Hypoxia and HIF-1α were present in tracheal epithelium over vessels that survived but not where they regressed. These findings reveal the plasticity of airway vasculature after birth and show that these vessels can be used to elucidate factors that promote postnatal vascular remodeling and maturation. Developmental Dynamics 239:2354-2366, 2010. © 2010 Wiley-Liss, Inc. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
18. Creation of PDX-Bearing Humanized Mice to Study Immuno-oncology.
- Author
-
Yao LC, Aryee KE, Cheng M, Kaur P, Keck JG, and Brehm MA
- Subjects
- Animals, Flow Cytometry methods, Hematopoietic Stem Cell Transplantation methods, Humans, Immunity, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating pathology, Mice, Inbred NOD, Mice, SCID, Neoplasms pathology, Neoplasm Transplantation methods, Neoplasms immunology, Transplantation, Heterologous methods
- Abstract
A significant obstacle to the study of human cancer biology and the testing of human specific immunotherapeutics is the paucity of translational models that recapitulate both the growth of human tumors and the functionality of human immune systems. Humanized mice engrafted with human hematopoietic stem cells (HSC) and patient-derived xenografts (PDX) enable preclinical investigation of the interactions between the human immune system and human cancer. We use immunodeficient non-obese diabetic (NOD, scid, gamma) NSG™ or NSG™-SGM3 mice as hosts for establishment of human immunity following HSC injection and for engraftment of human tumors. Here we describe a refined protocol for the subcutaneous implant of solid PDX tumors into humanized mice. Protocols to recover infiltrating immune cells from growing tumors and to evaluate the immune cell subsets by flow cytometry are also described.
- Published
- 2019
- Full Text
- View/download PDF
19. Characterization of the Selective Indoleamine 2,3-Dioxygenase-1 (IDO1) Catalytic Inhibitor EOS200271/PF-06840003 Supports IDO1 as a Critical Resistance Mechanism to PD-(L)1 Blockade Therapy.
- Author
-
Gomes B, Driessens G, Bartlett D, Cai D, Cauwenberghs S, Crosignani S, Dalvie D, Denies S, Dillon CP, Fantin VR, Guo J, Letellier MC, Li W, Maegley K, Marillier R, Miller N, Pirson R, Rabolli V, Ray C, Streiner N, Torti VR, Tsaparikos K, Van den Eynde BJ, Wythes M, Yao LC, Zheng X, Tumang J, and Kraus M
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized, Antineoplastic Agents pharmacology, B7-H1 Antigen metabolism, CTLA-4 Antigen metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Female, Humans, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Interferon-gamma metabolism, Kynurenine blood, Lymphocytes, Tumor-Infiltrating drug effects, Mice, Inbred BALB C, Mice, Inbred C57BL, Stereoisomerism, Substrate Specificity drug effects, T-Lymphocytes cytology, T-Lymphocytes drug effects, B7-H1 Antigen antagonists & inhibitors, Biocatalysis, Enzyme Inhibitors pharmacology, Immunotherapy, Indoleamine-Pyrrole 2,3,-Dioxygenase antagonists & inhibitors, Indoles pharmacology, Succinimides pharmacology
- Abstract
Tumors use indoleamine 2,3-dioxygenase-1 (IDO1) as a major mechanism to induce an immunosuppressive microenvironment. IDO1 expression is upregulated in many cancers and considered to be a resistance mechanism to immune checkpoint therapies. IDO1 is induced in response to inflammatory stimuli such as IFNγ and promotes immune tolerance by depleting tryptophan and producing tryptophan catabolites, including kynurenine, in the tumor microenvironment. This leads to effector T-cell anergy and enhanced T
reg function through upregulation of FoxP3. As a nexus for the induction of key immunosuppressive mechanisms, IDO1 represents an important immunotherapeutic target in oncology. Here, we report the identification and characterization of the novel selective, orally bioavailable IDO1 inhibitor EOS200271/PF-06840003. It reversed IDO1-induced T-cell anergy in vitro In mice carrying syngeneic tumor grafts, PF-06840003 reduced intratumoral kynurenine levels by over 80% and inhibited tumor growth both in monotherapy and, with an increased efficacy, in combination with antibodies blocking the immune checkpoint ligand PD-L1. We demonstrate that anti-PD-L1 therapy results in increased IDO1 metabolic activity thereby providing additional mechanistic rationale for combining PD-(L)1 blockade with IDO1 inhibition in cancer immunotherapies. Supported by these preclinical data and favorable predicted human pharmacokinetic properties of PF-06840003, a phase I open-label, multicenter clinical study (NCT02764151) has been initiated., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
20. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy.
- Author
-
Wang M, Yao LC, Cheng M, Cai D, Martinek J, Pan CX, Shi W, Ma AH, De Vere White RW, Airhart S, Liu ET, Banchereau J, Brehm MA, Greiner DL, Shultz LD, Palucka K, and Keck JG
- Subjects
- Animals, CD8-Positive T-Lymphocytes pathology, Cell Line, Tumor, Female, Humans, Mice, Mice, Inbred NOD, Neoplasms immunology, Neoplasms pathology, Xenograft Model Antitumor Assays, Antibodies, Monoclonal, Humanized pharmacology, CD8-Positive T-Lymphocytes immunology, Immunity, Cellular drug effects, Immunotherapy, Neoplasms therapy, Programmed Cell Death 1 Receptor immunology
- Abstract
Establishment of an in vivo small animal model of human tumor and human immune system interaction would enable preclinical investigations into the mechanisms underlying cancer immunotherapy. To this end, nonobese diabetic (NOD).Cg- Prkdc
scid IL2rgtm1Wjl /Sz (null; NSG) mice were transplanted with human (h)CD34+ hematopoietic progenitor and stem cells, which leads to the development of human hematopoietic and immune systems [humanized NSG (HuNSG)]. HuNSG mice received human leukocyte antigen partially matched tumor implants from patient-derived xenografts [PDX; non-small cell lung cancer (NSCLC), sarcoma, bladder cancer, and triple-negative breast cancer (TNBC)] or from a TNBC cell line-derived xenograft (CDX). Tumor growth curves were similar in HuNSG compared with nonhuman immune-engrafted NSG mice. Treatment with pembrolizumab, which targets programmed cell death protein 1, produced significant growth inhibition in both CDX and PDX tumors in HuNSG but not in NSG mice. Finally, inhibition of tumor growth was dependent on hCD8+ T cells, as demonstrated by antibody-mediated depletion. Thus, tumor-bearing HuNSG mice may represent an important, new model for preclinical immunotherapy research.-Wang, M., Yao, L.-C., Cheng, M., Cai, D., Martinek, J., Pan, C.-X., Shi, W., Ma, A.-H., De Vere White, R. W., Airhart, S., Liu, E. T., Banchereau, J., Brehm, M. A., Greiner, D. L., Shultz, L. D., Palucka, K., Keck, J. G. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy.- Published
- 2018
- Full Text
- View/download PDF
21. Rapamycin reversal of VEGF-C-driven lymphatic anomalies in the respiratory tract.
- Author
-
Baluk P, Yao LC, Flores JC, Choi D, Hong YK, and McDonald DM
- Abstract
Lymphatic malformations are serious but poorly understood conditions that present therapeutic challenges. The goal of this study was to compare strategies for inducing regression of abnormal lymphatics and explore underlying mechanisms. CCSP-rtTA/tetO-VEGF-C mice, in which doxycycline regulates VEGF-C expression in the airway epithelium, were used as a model of pulmonary lymphangiectasia. After doxycycline was stopped, VEGF-C expression returned to normal, but lymphangiectasia persisted for at least 9 months. Inhibition of VEGFR-2/VEGFR-3 signaling, Notch, β-adrenergic receptors, or autophagy and antiinflammatory steroids had no noticeable effect on the amount or severity of lymphangiectasia. However, rapamycin inhibition of mTOR reduced lymphangiectasia by 76% within 7 days without affecting normal lymphatics. Efficacy of rapamycin was not increased by coadministration with the other agents. In prevention trials, rapamycin suppressed VEGF-C-driven mTOR phosphorylation and lymphatic endothelial cell sprouting and proliferation. However, in reversal trials, no lymphatic endothelial cell proliferation was present to block in established lymphangiectasia, and rapamycin did not increase caspase-dependent apoptosis. However, rapamycin potently suppressed Prox1 and VEGFR-3. These experiments revealed that lymphangiectasia is remarkably resistant to regression but is responsive to rapamycin, which rapidly reduces and normalizes the abnormal lymphatics without affecting normal lymphatics.
- Published
- 2017
- Full Text
- View/download PDF
22. Plasticity of airway lymphatics in development and disease.
- Author
-
Yao LC and McDonald DM
- Subjects
- Animals, Humans, Respiratory System physiopathology, Lymphangiogenesis, Lymphatic Vessels embryology, Lymphatic Vessels physiology, Respiratory System embryology, Respiratory Tract Diseases physiopathology
- Abstract
The dynamic nature of lymphatic vessels is reflected by structural and functional modifications that coincide with changes in their environment. Lymphatics in the respiratory tract undergo rapid changes around birth, during adaptation to air breathing, when lymphatic endothelial cells develop button-like intercellular junctions specialized for efficient fluid uptake and transport. In inflammatory conditions, lymphatic vessels proliferate and undergo remodeling to accommodate greater plasma leakage and immune cell trafficking. However, the newly formed lymphatics are abnormal, and resolution of inflammation is not accompanied by complete reversal of the lymphatic vessel changes back to the baseline. As the understanding of lymphatic plasticity advances, approaches for eliminating the abnormal vessels and improving the functionality of those that remain move closer to reality. This chapter provides an overview of what is known about lymphatic vessel growth, remodeling, and other forms of plasticity that occur during development or inflammation, with an emphasis on the respiratory tract. Also addressed is the limited reversibility of changes in lymphatics during the resolution of inflammation.
- Published
- 2014
- Full Text
- View/download PDF
23. Dynamics of airway blood vessels and lymphatics: lessons from development and inflammation.
- Author
-
McDonald DM, Yao LC, and Baluk P
- Subjects
- Animals, Capillaries physiology, Humans, Lymphatic Vessels physiology, Mice, Models, Animal, Inflammation physiopathology, Respiratory System blood supply, Respiratory Tract Diseases physiopathology
- Abstract
Blood vessels and lymphatic vessels in the respiratory tract play key roles in inflammation. By undergoing adaptive remodeling and growth, blood vessels undergo changes that enable the extravasation of plasma and leukocytes into inflamed tissues, and lymphatic vessels adjust to the increased fluid clearance and cell traffic involved in immune responses. Blood vessels and lymphatics in adult airways are strikingly different from those of late-stage embryos. Before birth, blood vessels in mouse airways make up a primitive plexus similar to that of the yolk sac. This plexus undergoes rapid and extensive remodeling at birth. In the early neonatal period, parts of the plexus regress. Capillaries then rapidly regrow, and with arterioles and venules form the characteristic adult vascular pattern. Lymphatic vessels of the airways also undergo rapid changes around birth, when lymphatic endothelial cells develop button-like intercellular junctions specialized for efficient fluid uptake. Among the mechanisms that underlie the onset of rapid vascular remodeling at birth, changes in tissue oxygen tension and mechanical forces associated with breathing are likely to be involved, along with growth factors that promote the growth and maturation of blood vessels and lymphatics. Whatever the mechanisms, the dynamic nature of airway blood vessels and lymphatics during perinatal development foretells the extraordinary vascular plasticity found in many diseases.
- Published
- 2011
- Full Text
- View/download PDF
24. VEGF and c-Met blockade amplify angiogenesis inhibition in pancreatic islet cancer.
- Author
-
You WK, Sennino B, Williamson CW, Falcón B, Hashizume H, Yao LC, Aftab DT, and McDonald DM
- Subjects
- Adenoma, Islet Cell pathology, Anilides pharmacology, Animals, Apoptosis drug effects, Basement Membrane drug effects, Basement Membrane metabolism, Basement Membrane pathology, Cell Hypoxia drug effects, Mice, Mice, Inbred C57BL, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Pancreatic Neoplasms pathology, Proto-Oncogene Proteins c-met metabolism, Pyridines pharmacology, Quinolines pharmacology, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-2 biosynthesis, Vascular Endothelial Growth Factor Receptor-3 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-3 biosynthesis, Adenoma, Islet Cell blood supply, Adenoma, Islet Cell drug therapy, Pancreatic Neoplasms blood supply, Pancreatic Neoplasms drug therapy, Proto-Oncogene Proteins c-met antagonists & inhibitors, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
Angiogenesis inhibitors that block VEGF receptor (VEGFR) signaling slow the growth of many types of tumors, but eventually the disease progresses. Multiple strategies are being explored to improve efficacy by concurrent inhibition of other functionally relevant receptor tyrosine kinases (RTK). XL880 (foretinib, GSK1363089) and XL184 (cabozantinib) are small-molecule inhibitors that potently block multiple RTKs, including VEGFR and the receptor of hepatocyte growth factor c-Met, which can drive tumor invasion and metastasis. This study compared the cellular effects of XL880 and XL184 with those of an RTK inhibitor (XL999) that blocks VEGFR but not c-Met. Treatment of RIP-Tag2 mice with XL999 resulted in 43% reduction in vascularity of spontaneous pancreatic islet tumors over 7 days, but treatment with XL880 or XL184 eliminated approximately 80% of the tumor vasculature, reduced pericytes and empty basement membrane sleeves, caused widespread intratumoral hypoxia and tumor cell apoptosis, and slowed regrowth of the tumor vasculature after drug withdrawal. Importantly, XL880 and XL184 also decreased invasiveness of primary tumors and reduced metastasis. Overall, these findings indicate that inhibition of c-Met and functionally related kinases amplifies the effects of VEGFR blockade and leads to rapid, robust, and progressive regression of tumor vasculature, increased intratumoral hypoxia and apoptosis, and reduced tumor invasiveness and metastasis., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
25. Multiple modular promoter elements drive graded brinker expression in response to the Dpp morphogen gradient.
- Author
-
Yao LC, Phin S, Cho J, Rushlow C, Arora K, and Warrior R
- Subjects
- Animals, Drosophila embryology, Drosophila genetics, Drosophila Proteins genetics, Embryo, Nonmammalian, Gene Expression Regulation, Developmental, Insect Proteins genetics, Repressor Proteins genetics, Transcription Factors genetics, Drosophila Proteins metabolism, Insect Proteins metabolism, Promoter Regions, Genetic, Repressor Proteins metabolism, Transcription Factors metabolism
- Abstract
Morphogen gradients play fundamental roles in patterning and cell specification during development by eliciting differential transcriptional responses in target cells. In Drosophila, Decapentaplegic (Dpp), the BMP2/4 homolog, downregulates transcription of the nuclear repressor brinker (brk) in a concentration-dependent manner to generate an inverse graded distribution. Both Dpp and Brk are crucial for directing Dpp target gene expression in defined domains and the consequent execution of distinct developmental programs. Thus, determining the mechanism by which the brk promoter interprets the Dpp activity gradient is essential for understanding both Dpp-dependent patterning and how graded signaling activity can generate different responses through transcriptional repression. We have uncovered key features of the brk promoter that suggest it uses a complex enhancer logic not represented in current models. First, we find that the regulatory region contains multiple compact modules that can independently drive brk-like expression patterns. Second, each module contains binding sites for the Schnurri/Mad/Medea (SMM) complex, which mediates Dpp-dependent repression, linked to regions that direct activation. Third, the SMM repression complex acts through a distance-dependent mechanism that probably uses the canonical co-repressor C-terminal Binding Protein (CtBP). Finally, our data suggest that inputs from multiple regulatory modules are integrated to generate the final pattern. This unusual promoter organization may be necessary for brk to respond to the Dpp gradient in a precise and robust fashion.
- Published
- 2008
- Full Text
- View/download PDF
26. Schnurri transcription factors from Drosophila and vertebrates can mediate Bmp signaling through a phylogenetically conserved mechanism.
- Author
-
Yao LC, Blitz IL, Peiffer DA, Phin S, Wang Y, Ogata S, Cho KW, Arora K, and Warrior R
- Subjects
- Animals, Base Sequence, Conserved Sequence genetics, DNA-Binding Proteins genetics, Drosophila embryology, Drosophila genetics, Embryo, Nonmammalian metabolism, Gene Expression Regulation, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Phylogeny, Signal Transduction, Smad Proteins metabolism, Transcription Factors genetics, Vertebrates genetics, Vertebrates metabolism, Xenopus genetics, Xenopus metabolism, Xenopus Proteins genetics, Xenopus Proteins metabolism, Bone Morphogenetic Proteins genetics, DNA-Binding Proteins classification, DNA-Binding Proteins metabolism, Drosophila metabolism, Drosophila Proteins classification, Drosophila Proteins genetics, Drosophila Proteins metabolism, Response Elements genetics, Transcription Factors classification, Transcription Factors metabolism
- Abstract
Bone Morphogenetic Proteins (Bmps) are secreted growth factors that play crucial roles in animal development across the phylogenetic spectrum. Bmp signaling results in the phosphorylation and nuclear translocation of Smads, downstream signal transducers that bind DNA. In Drosophila, the zinc finger protein Schnurri (Shn) plays a key role in signaling by the Bmp2/Bmp4 homolog Decapentaplegic (Dpp), by forming a Shn/Smad repression complex on defined promoter elements in the brinker (brk) gene. Brk is a transcriptional repressor that downregulates Dpp target genes. Thus, brk inhibition by Shn results in the upregulation of Dpp-responsive genes. We present evidence that vertebrate Shn homologs can also mediate Bmp responsiveness through a mechanism similar to Drosophila Shn. We find that a Bmp response element (BRE) from the Xenopus Vent2 promoter drives Dpp-dependent expression in Drosophila. However, in sharp contrast to its activating role in vertebrates, the frog BRE mediates repression in Drosophila. Remarkably, despite these opposite transcriptional polarities, sequence changes that abolish cis-element activity in Drosophila also affect BRE function in Xenopus. These similar cis requirements reflect conservation of trans-acting factors, as human Shn1 (hShn1; HIVEP1) can interact with Smad1/Smad4 and assemble an hShn1/Smad complex on the BRE. Furthermore, both Shn and hShn1 activate the BRE in Xenopus embryos, and both repress brk and rescue embryonic patterning defects in shn mutants. Our results suggest that vertebrate Shn proteins function in Bmp signal transduction, and that Shn proteins recruit coactivators and co-repressors in a context-dependent manner, rather than acting as dedicated activators or repressors.
- Published
- 2006
- Full Text
- View/download PDF
27. Two Pax genes, eye gone and eyeless, act cooperatively in promoting Drosophila eye development.
- Author
-
Jang CC, Chao JL, Jones N, Yao LC, Bessarab DA, Kuo YM, Jun S, Desplan C, Beckendorf SK, and Sun YH
- Subjects
- Animals, DNA-Binding Proteins genetics, Drosophila Proteins genetics, Drosophila melanogaster anatomy & histology, Drosophila melanogaster embryology, Drosophila melanogaster genetics, Embryonic Structures anatomy & histology, Gene Expression Regulation, Developmental, Genes, Insect, In Situ Hybridization, Phenotype, Photoreceptor Cells, Invertebrate anatomy & histology, Photoreceptor Cells, Invertebrate embryology, Photoreceptor Cells, Invertebrate growth & development, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Transcription, Genetic, Transgenes, Wnt1 Protein, DNA-Binding Proteins metabolism, Drosophila Proteins metabolism, Drosophila melanogaster growth & development, Embryonic Structures growth & development
- Abstract
We report the identification of a Drosophila Pax gene, eye gone (eyg), which is required for eye development. Loss-of-function eyg mutations cause reduction or absence of the eye. Similar to the Pax6 eyeless (ey) gene, ectopic expression of eyg induces extra eye formation, but at sites different from those induced by ey. Several lines of evidence suggest that eyg and ey act cooperatively: (1) eyg expression is not regulated by ey, nor does it regulate ey expression, (2) eyg-induced ectopic morphogenetic furrow formation does not require ey, nor does ey-induced ectopic eye production require eyg, (3) eyg and ey can partially substitute for the function of the other, and (4) coexpression of eyg and ey has a synergistic enhancement of ectopic eye formation. Our results also show that eyg has two major functions: to promote cell proliferation in the eye disc and to promote eye development through suppression of wg transcription.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.