201
|
Zhou A, Lin K, Zhang S, Ma L, Xue J, Morris SA, Aldape KD, Huang S. Gli1-induced deubiquitinase USP48 aids glioblastoma tumorigenesis by stabilizing Gli1. EMBO Rep 2017. [PMID: 28623188 DOI: 10.15252/embr.201643124] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aberrant activation of the Hedgehog (Hh) signaling pathway drives the tumorigenesis of multiple cancers. In this study, we screened a panel of deubiquitinases that may regulate the Hh pathway. We find that deubiquitinase USP48 activates Gli-dependent transcription by stabilizing Gli1 protein. Mechanistically, USP48 interacts with Gli1 and cleaves its ubiquitin off directly. In glioblastoma cells, knockdown of USP48 inhibits cell proliferation and the expression of Gli1's downstream targets, which leads to repressed glioblastoma tumorigenesis. Importantly, USP48's effect on cell proliferation and tumorigenesis depends to some extent on Gli1. In addition, we find that the Sonic Hedgehog (SHH) pathway induces USP48 expression through Gli1-mediated transcriptional activation, which forms thus a positive feedback loop to regulate Hh signaling. In human glioblastoma specimens, the expression levels of USP48 and Gli1 proteins are clinically relevant, and high expression of USP48 correlates with glioma malignancy. In summary, our study reveals that the USP48-Gli1 regulatory axis is critical for glioma cell proliferation and glioblastoma tumorigenesis.
Collapse
Affiliation(s)
- Aidong Zhou
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kangyu Lin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sicong Zhang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Program in Cancer Biology, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Li Ma
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Neuro-oncology and Neurosurgery, National Clinical Research Center for Cancer, Tianjin Medical University Institute and Hospital, Tianjin, China
| | - Jianfei Xue
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Saint-Aaron Morris
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth D Aldape
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Suyun Huang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA .,Program in Cancer Biology, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
202
|
Lee DH, Lee SY, Oh SC. Hedgehog signaling pathway as a potential target in the treatment of advanced gastric cancer. Tumour Biol 2017. [DOI: 10.1177/1010428317692266] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Dae-Hee Lee
- Division of Brain Korea 21 Program for Biomedicine Science, College of Medicine, Korea University, Seoul, Republic of Korea
- Division of Oncology and Hematology, Department of Internal Medicine, College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Suk-young Lee
- Division of Oncology and Hematology, Department of Internal Medicine, College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Sang Cheul Oh
- Division of Brain Korea 21 Program for Biomedicine Science, College of Medicine, Korea University, Seoul, Republic of Korea
- Division of Oncology and Hematology, Department of Internal Medicine, College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| |
Collapse
|
203
|
Shao S, Wang GL, Raymond C, Deng XH, Zhu XL, Wang D, Hong LP. Activation of Sonic hedgehog signal by Purmorphamine, in a mouse model of Parkinson's disease, protects dopaminergic neurons and attenuates inflammatory response by mediating PI3K/AKt signaling pathway. Mol Med Rep 2017. [PMID: 28627590 PMCID: PMC5562000 DOI: 10.3892/mmr.2017.6751] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In Parkinson's disease (PD), microglial activation-mediated neuroinflammation is associated with dopaminergic neurons degeneration in the substantia nigra pars compacta. Previous studies that have investigated this neurodegenerative disease have reported that the Sonic hedgehog (SHH) signaling pathway, through inhibiting the inflammatory processes, exerts a beneficial neuroprotective effect. However, the mechanisms underlying the anti-inflammatory and neuroprotective effects of this signaling pathway remain poorly understood. The present study aimed to further investigate these mechanisms in vitro and in vivo. At first, BV2 microglial cells treated with lipopolysaccharide (LPS) were used to induce an inflammatory response. It was observed that the activation of SHH signaling by Purmorphamine attenuated the LPS-induced inflammatory response, increased the expression of transforming growth factor-β1 through the phosphatidylinositol 3-kinase (PI3K)/AKT serine/threonine kinase (Akt) intracellular signaling pathway and inhibited nuclear receptor subfamily 4 group A member 2, independently of the PI3K/Akt signaling pathway. Furthermore, the blockade of the PI3K/Akt signaling pathway by intranasal administration of LY294002, significantly reduced the SHH-associated neuroprotective effects on dopaminergic neurons, improved motor functions, and increased the microglial activation and inflammatory response in a mouse model of PD induced using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. In conclusion, the data of the present study reported that anti-inflammatory and neuroprotective effects can be obtained in BV2 microglial cells and in a mouse model of PD by successive activation of the SHH and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Shuai Shao
- Department of Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Guang-Liang Wang
- Department of Histology and Embryology, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Cespuglio Raymond
- Neuroscience Research Center of Lyon (CRNL), Neurochem, Claude Bernard University, F-69373 Lyon, France
| | - Xue-Hua Deng
- Department of Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xiao-Lan Zhu
- Department of Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Di Wang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Le-Peng Hong
- Department of Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| |
Collapse
|
204
|
Marwarha G, Raza S, Hammer K, Ghribi O. 27-hydroxycholesterol: A novel player in molecular carcinogenesis of breast and prostate cancer. Chem Phys Lipids 2017; 207:108-126. [PMID: 28583434 DOI: 10.1016/j.chemphyslip.2017.05.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 12/13/2022]
Abstract
Several studies have suggested an etiological role for hypercholesterolemia in the pathogenesis of breast cancer and prostate cancer (PCa). However, the molecular mechanisms that underlie and mediate the hypercholesterolemia-fostered increased risk for breast cancer and PCa are yet to be determined. The discovery that the most abundant cholesterol oxidized metabolite in the plasma, 27 hydroxycholesterol (27-OHC), is a selective estrogen receptor modulator (SERM) and an agonist of Liver X receptors (LXR) partially fills the void in our understanding and knowledge of the mechanisms that may link hypercholesterolemia to development and progression of breast cancer and PCa. The wide spectrum and repertoire of SERM and LXR-dependent effects of 27-OHC in the context of all facets and aspects of breast cancer and prostate cancer biology are reviewed in this manuscript in a very comprehensive manner. This review highlights recent findings pertaining to the role of 27-OHC in breast cancer and PCa and delineates the signaling mechanisms involved in the governing of different facets of tumor biology, that include tumor cell proliferation, epithelial-mesenchymal transition (EMT), as well as tumor cell invasion, migration, and metastasis. We also discuss the limitations of contemporary studies and lack of our comprehension of the entire gamut of effects exerted by 27-OHC that may be relevant to the pathogenesis of breast cancer and PCa. We unveil and propose potential future directions of research that may further our understanding of the role of 27-OHC in breast cancer and PCa and help design therapeutic interventions against endocrine therapy-resistant breast cancer and PCa.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA
| | - Shaneabbas Raza
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA
| | - Kimberly Hammer
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA; Department of Veteran Affairs, Fargo VA Health Care System, Fargo, North Dakota 58102, USA
| | - Othman Ghribi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA.
| |
Collapse
|
205
|
Singh AK, Sharma N, Ghosh M, Park YH, Jeong DK. Emerging importance of dietary phytochemicals in fight against cancer: Role in targeting cancer stem cells. Crit Rev Food Sci Nutr 2017; 57:3449-3463. [DOI: 10.1080/10408398.2015.1129310] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Amit Kumar Singh
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| | - Neelesh Sharma
- Division of Veterinary Medicine, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, R. S. Pura, Jammu, India
| | - Mrinmoy Ghosh
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| | | | - Dong Kee Jeong
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
206
|
Tech K, Tikunov AP, Farooq H, Morrissy AS, Meidinger J, Fish T, Green SC, Liu H, Li Y, Mungall AJ, Moore RA, Ma Y, Jones SJM, Marra MA, Vander Heiden MG, Taylor MD, Macdonald JM, Gershon TR. Pyruvate Kinase Inhibits Proliferation during Postnatal Cerebellar Neurogenesis and Suppresses Medulloblastoma Formation. Cancer Res 2017; 77:3217-3230. [PMID: 28515149 DOI: 10.1158/0008-5472.can-16-3304] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/20/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022]
Abstract
Aerobic glycolysis supports proliferation through unresolved mechanisms. We have previously shown that aerobic glycolysis is required for the regulated proliferation of cerebellar granule neuron progenitors (CGNP) and for the growth of CGNP-derived medulloblastoma. Blocking the initiation of glycolysis via deletion of hexokinase-2 (Hk2) disrupts CGNP proliferation and restricts medulloblastoma growth. Here, we assessed whether disrupting pyruvate kinase-M (Pkm), an enzyme that acts in the terminal steps of glycolysis, would alter CGNP metabolism, proliferation, and tumorigenesis. We observed a dichotomous pattern of PKM expression, in which postmitotic neurons throughout the brain expressed the constitutively active PKM1 isoform, while neural progenitors and medulloblastomas exclusively expressed the less active PKM2. Isoform-specific Pkm2 deletion in CGNPs blocked all Pkm expression. Pkm2-deleted CGNPs showed reduced lactate production and increased SHH-driven proliferation. 13C-flux analysis showed that Pkm2 deletion reduced the flow of glucose carbons into lactate and glutamate without markedly increasing glucose-to-ribose flux. Pkm2 deletion accelerated tumor formation in medulloblastoma-prone ND2:SmoA1 mice, indicating the disrupting PKM releases CGNPs from a tumor-suppressive effect. These findings show that distal and proximal disruptions of glycolysis have opposite effects on proliferation, and that efforts to block the oncogenic effect of aerobic glycolysis must target reactions upstream of PKM. Cancer Res; 77(12); 3217-30. ©2017 AACR.
Collapse
Affiliation(s)
- Katherine Tech
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina.,Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Andrey P Tikunov
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina
| | - Hamza Farooq
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - A Sorana Morrissy
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jessica Meidinger
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Taylor Fish
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Sarah C Green
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina
| | - Hedi Liu
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Yisu Li
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Andrew J Mungall
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Richard A Moore
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Yussanne Ma
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Steven J M Jones
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Marco A Marra
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Matthew G Vander Heiden
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michael D Taylor
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jeffrey M Macdonald
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina
| | - Timothy R Gershon
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina. .,Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina.,UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
207
|
Design, Synthesis and Biological Evaluation of novel Hedgehog Inhibitors for treating Pancreatic Cancer. Sci Rep 2017; 7:1665. [PMID: 28490735 PMCID: PMC5431907 DOI: 10.1038/s41598-017-01942-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 04/05/2017] [Indexed: 12/12/2022] Open
Abstract
Hedgehog (Hh) pathway is involved in epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) maintenance resulting in tumor progression. GDC-0449, an inhibitor of Hh pathway component smoothened (Smo) has shown promise in the treatment of various cancers including pancreatic cancer. However, the emergence of resistance during GDC-0449 treatment with numerous side effects limits its use. Therefore, here we report the design, synthesis and evaluation of novel GDC-0449 analogs using N-[3-(2-pyridinyl) phenyl] benzamide scaffold. Cell-based screening followed by molecular simulation revealed 2-chloro-N1-[4-chloro-3-(2-pyridinyl)phenyl]-N4,N4-bis(2-pyridinylmethyl)-1,4-benzenedicarboxamide (MDB5) as most potent analog, binding with an extra interactions in seven-transmembrane (7-TM) domain of Smo due to an additional 2-pyridylmethyl group than GDC-0449. Moreover, MDB5 was more efficient in inhibiting Hh pathway components as measured by Gli-1 and Shh at transcriptional and translational levels. Additionally, a significant reduction of ALDH1, CD44 and Oct-3/4, key markers of pancreatic CSC was observed when MIA PaCa-2 cells were treated with MDB5 compared to GDC-0449. In a pancreatic tumor mouse model, MDB5 containing nanoparticles treated group showed significant inhibition of tumor growth without loss in body weight. These evidence highlight the enhanced Hh pathway inhibition and anticancer properties of MDB5 leaving a platform for mono and/or combination therapy.
Collapse
|
208
|
Guo M, Hong KH, Lv Y, Ding Y, Li C, Xu H, Qi W, Chen J, Ji M, Cai J. A Novel and Efficient Route for Synthesis of Taladegib. JOURNAL OF CHEMICAL RESEARCH 2017. [DOI: 10.3184/003685017x14859543105069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Taladegib (LY-2940680), a small molecule Hedgehog signalling pathway inhibitor, was obtained from N-benzyl-4-piperidone via Borch reductive amination, acylation with 4-fluoro-2-(trifluoromethyl)benzoyl chloride, debenzylation, substitution with 1,4-dichlorophthalazine and Suzuki cross-coupling reaction with 1-methyl-1H-pyrazole-5-boronic acid. The advantages of this synthesis route were the elimination of Boc protection and deprotection and the inexpensive starting materials. Furthermore, the debenzylation reaction was achieved with simplified operational procedure using ammonium formate as hydrogen source that provided high reaction yield. This synthetic procedure was suitable for large-scale production of the compound for biological evaluation and further study.
Collapse
Affiliation(s)
- Mingliang Guo
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, 211189, P.R. China
| | - Kwon Ho Hong
- Department of Medicinal Chemistry and the Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN 55414, USA
| | - Yongfeng Lv
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, 211189, P.R. China
| | - Yu Ding
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, 211189, P.R. China
| | - Congcong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, 211189, P.R. China
| | - Hua Xu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, 211189, P.R. China
| | - Wenxiu Qi
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, 211189, P.R. China
| | - Junqing Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, 211189, P.R. China
| | - Min Ji
- School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, P.R. China
- Suzhou Key Laboratory of Biomaterials and Technologies & Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215123, P.R. China
| | - Jin Cai
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, 211189, P.R. China
- Suzhou Key Laboratory of Biomaterials and Technologies & Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215123, P.R. China
| |
Collapse
|
209
|
Weidle UH, Birzele F, Kollmorgen G, Rüger R. Long Non-coding RNAs and their Role in Metastasis. Cancer Genomics Proteomics 2017; 14:143-160. [PMID: 28446530 PMCID: PMC5420816 DOI: 10.21873/cgp.20027] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 02/08/2023] Open
Abstract
The perception of long non-coding RNAs as chunk RNA and transcriptional noise has been steadily replaced by their role as validated targets for a diverse set of physiological processes in the past few years. However, for the vast majority of lncRNAs their precise mode of action and physiological function remain to be uncovered. A large body of evidence has revealed their essential role in all stages of cancirogenesis and metastasis. In this review we focus on the role of lncRNAs in metastasis. We grouped selected lncRNAs into three categories based on in vitro and in vivo mode of action-related studies and clinical relevance for metastasis. Grouped according to their mode of action, in category I we discuss lncRNAs such as CCAT2, DREH, LET, NKILA, treRNA, HOTAIR, H19, FENDRR, lincROR, MALAT, GClnc1, BCAR4, SCHLAP1 and lncRNA ATP, all lncRNAs with in vitro and in vivo metastasis-related data and clinical significance. In category II we discuss lncRNAs CCAT1, PCAT1, PTENgp1, GPLINC, MEG3, ZEB2-AS, LCT13, ANRIL, NBAT1 and lncTCF7 all characterized by their mode of action in vitro and clinical significance, but pending or preliminary in vivo data. Finally, under category III, we discuss lncRNAs BANCR, FRLnc1, SPRY4-IT1 and LIMT with partially or poorly-resolved mode of action and varying degree of validation in clinical metastasis. Finally we discuss metastasis-related translational aspects of lncRNAs.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Fabian Birzele
- Roche Innovation Center Basel, F. Hofman La Roche, Basel, Switzerland
| | - Gwen Kollmorgen
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Rüdiger Rüger
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
210
|
Yan C, Yang Q, Huo X, Li H, Zhou L, Gong Z. Chemical inhibition reveals differential requirements of signaling pathways in kras V12- and Myc-induced liver tumors in transgenic zebrafish. Sci Rep 2017; 7:45796. [PMID: 28378824 PMCID: PMC5381109 DOI: 10.1038/srep45796] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022] Open
Abstract
Previously we have generated inducible liver tumor models by transgenic expression of an oncogene and robust tumorigenesis can be rapidly induced by activation of the oncogene in both juvenile and adult fish. In the present study, we aimed at chemical intervention of tumorigenesis for understanding molecular pathways of tumorigenesis and for potential development of a chemical screening tool for anti-cancer drug discovery. Thus, we evaluated the roles of several major signaling pathways in krasV12- or Myc-induced liver tumors by using several small molecule inhibitors: SU5402 and SU6668 for VEGF/FGF signaling; IWR1 and cardionogen 1 for Wnt signaling; and cyclopamine and Gant61 for Hedgehog signaling. Inhibition of VEGF/FGF signaling was found to deter both Myc- and krasV12-induced liver tumorigenesis while suppression of Wnt signaling relaxed only Myc- but not krasV12-induced liver tumorigenesis. Inhibiting Hedgehog signaling did not suppress either krasV12 or Myc-induced tumors. The suppression of liver tumorigenesis was accompanied with a decrease of cell proliferation, increase of apoptosis, distorted liver histology. Collectively, our observations suggested the requirement of VEGF/FGF signaling but not the hedgehog signaling in liver tumorigenesis in both transgenic fry. However, Wnt signaling appeared to be required for liver tumorigenesis only in Myc but not krasV12 transgenic zebrafish.
Collapse
Affiliation(s)
- Chuan Yan
- Department of Biological Sciences, National University of Singapore, Singapore
- National University of Singapore graduate school for integrative sciences and engineering, National University of Singapore, Singapore
| | - Qiqi Yang
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Xiaojing Huo
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Hankun Li
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Li Zhou
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, Singapore
- National University of Singapore graduate school for integrative sciences and engineering, National University of Singapore, Singapore
| |
Collapse
|
211
|
Noncanonical GLI1 signaling promotes stemness features and in vivo growth in lung adenocarcinoma. Oncogene 2017; 36:4641-4652. [PMID: 28368412 PMCID: PMC5558095 DOI: 10.1038/onc.2017.91] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 02/04/2017] [Accepted: 02/26/2017] [Indexed: 02/07/2023]
Abstract
Aberrant Hedgehog/GLI signaling has been implicated in a diverse spectrum of human cancers, but its role in lung adenocarcinoma (LAC) is still under debate. We show that the downstream effector of the Hedgehog pathway, GLI1, is expressed in 76% of LACs, but in roughly half of these tumors, the canonical pathway activator, Smoothened, is expressed at low levels, possibly owing to epigenetic silencing. In LAC cells including the cancer stem cell compartment, we show that GLI1 is activated noncanonically by MAPK/ERK signaling. Different mechanisms can trigger the MAPK/ERK/GLI1 cascade including KRAS mutation and stimulation of NRP2 by VEGF produced by the cancer cells themselves in an autocrine loop or by stromal cells as paracrine cross talk. Suppression of GLI1, by silencing or drug-mediated, inhibits LAC cells proliferation, attenuates their stemness and increases their susceptibility to apoptosis in vitro and in vivo. These findings provide insight into the growth of LACs and point to GLI1 as a downstream effector for oncogenic pathways. Thus, strategies involving direct inhibition of GLI1 may be useful in the treatment of LACs.
Collapse
|
212
|
Ye Y, Wei Y, Xu Y, Li Y, Wang R, Chen J, Zhou Y, Fu Z, Chen Y, Wang X, Yu R, Bai C, Li G, Chen R, Chen T. Induced MiR-1249 expression by aberrant activation of Hedegehog signaling pathway in hepatocellular carcinoma. Exp Cell Res 2017; 355:9-17. [PMID: 28365245 DOI: 10.1016/j.yexcr.2017.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 03/04/2017] [Accepted: 03/07/2017] [Indexed: 12/29/2022]
Abstract
Aberrant activations of Hedegehog (Hh) signaling were found in hepatocellular carcinoma (HCC) and some other cancer types. However, the details have not been completely understood and the underlying mechanism remains unclear. Here we reported that miR-1249 transcription in HCC cells was regulated through direct binding to the conserved sequences in miR-1249 promoter region by Gli1, which functions as a transcription factor and is a component in the Hh signaling pathway. Interestingly, expression of tumor suppressor PTCH1, which is another component of the Hh signaling pathway, was inhibited by miR-1249 through targeting its 3'-untranslated region. Down-regulation of PTCH1 further enhanced the downstream effects mediated by Gli1. In consistent with these findings, miR-1249 expression level was correlated with degree of prognosis (p=0.005) in HCC patients. Taken together, our results suggested the existence of a positive feedback loop comprised of Gli1, miR-1249 and PTCH1. During the process of HCC progression, this positive feedback loop could be continuously activated to enhance tumor cell growth, migration and invasion.
Collapse
Affiliation(s)
- Yibiao Ye
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunping Wei
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunxiuxiu Xu
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanshan Li
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruomei Wang
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie Chen
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Zhou
- Department of General Surgery, Guangdong General Hospital, Guangzhou, China
| | - Zhiqiang Fu
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Chen
- Pearl Laboratory Animal Sci. & Tech. Co., Ltd., Guangzhou, China
| | - Xin Wang
- Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Huhhot, PR China; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA; Hepatoscience Inc., Sunnyvale, CA 94085, USA
| | - Ruiping Yu
- Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Huhhot, PR China
| | - Chunling Bai
- Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Huhhot, PR China
| | - Guangpeng Li
- Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Huhhot, PR China
| | - Rufu Chen
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Tao Chen
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
213
|
Phase I trial of the oral smoothened inhibitor sonidegib in combination with paclitaxel in patients with advanced solid tumors. Invest New Drugs 2017; 35:766-772. [PMID: 28317088 DOI: 10.1007/s10637-017-0454-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 03/06/2017] [Indexed: 02/04/2023]
Abstract
Purpose To establish a recommended phase II dose (RP2D) for the oral smoothened inhibitor sonidegib in combination with paclitaxel; secondary objectives include evaluation of safety, tolerability, markers of Hedgehog (Hh) signaling and preliminary antitumor activity. Methods Patients with advanced solid tumors were enrolled in cohorts of escalating sonidegib dose levels (400mg, 600mg and 800mg orally, once daily on days 1-28) in combination with paclitaxel 80 mg/m2 on days 1, 8 and 15 in 4-weekly cycles. Dose-limiting toxicities (DLTs) were assessed using CTCAE v4. Once the RP2D was defined, patients with advanced ovarian carcinoma were treated at this dose level in an expansion phase. Biomarkers of Hh signaling were assessed by immunohistochemistry in archival tissue and antitumor activity evaluated using RECIST 1.1. Results 18 patients were treated: 3 at 400 mg, 3 at 600 mg and 12 at 800 mg sonidegib. Only one patient treated at 800 mg presented a DLT (prolonged neutropenia resulting in failure to receive 75% of the planned sonidegib dose). However, 4 of 12 patients treated at 800 mg had their sonidegib dose reduced for toxicity after cycle 1. Hh biomarker (SHH, Patched, SMO and GLI1) staining did not correlate with clinical activity. Best response was partial response in 3 patients (2 ovarian, 1 breast cancer) and stable disease >4 cycles in 3 patients (2 ovarian, 1 anal cancer). Conclusions The combination of sonidegib and paclitaxel is tolerable and evidence of antitumor activity was identified. The RP2D of sonidegib was 800 mg in combination with paclitaxel 80mg/m2.
Collapse
|
214
|
Jain S, Song R, Xie J. Sonidegib: mechanism of action, pharmacology, and clinical utility for advanced basal cell carcinomas. Onco Targets Ther 2017; 10:1645-1653. [PMID: 28352196 PMCID: PMC5360396 DOI: 10.2147/ott.s130910] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The Hedgehog (Hh) pathway is critical for cell differentiation, tissue polarity, and stem cell maintenance during embryonic development, but is silent in adult tissues under normal conditions. However, aberrant Hh signaling activation has been implicated in the development and promotion of certain types of cancer, including basal cell carcinoma (BCC), medulloblastoma, and gastrointestinal cancers. In 2015, the US Food and Drug Administration (FDA) approved sonidegib, a smoothened (SMO) antagonist, for treatment of advanced BCC (aBCC) after a successful Phase II clinical trial. Sonidegib, also named Odomzo, is the second Hh signaling inhibitor approved by the FDA to treat BCCs following approval of the first SMO antagonist vismodegib in 2012. What are the major features of sonidegib (mechanism of action; metabolic profiles, clinical efficacy, safety, and tolerability profiles)? Will the sonidegib experience help other clinical trials using Hh signaling inhibitors in the future? In this review, we will summarize current understanding of BCCs and Hh signaling. We will focus on sonidegib and its use in the clinic, and we will discuss ways to improve its clinical application in cancer therapeutics.
Collapse
Affiliation(s)
| | - Ruolan Song
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA
| | - Jingwu Xie
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA
| |
Collapse
|
215
|
|
216
|
Wu F, Zhang Y, Sun B, McMahon AP, Wang Y. Hedgehog Signaling: From Basic Biology to Cancer Therapy. Cell Chem Biol 2017; 24:252-280. [PMID: 28286127 DOI: 10.1016/j.chembiol.2017.02.010] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/29/2016] [Accepted: 02/10/2017] [Indexed: 02/07/2023]
Abstract
The Hedgehog (HH) signaling pathway was discovered originally as a key pathway in embryonic patterning and development. Since its discovery, it has become increasingly clear that the HH pathway also plays important roles in a multitude of cancers. Therefore, HH signaling has emerged as a therapeutic target of interest for cancer therapy. In this review, we provide a brief overview of HH signaling and the key molecular players involved and offer an up-to-date summary of our current knowledge of endogenous and exogenous small molecules that modulate HH signaling. We discuss experiences and lessons learned from the decades-long efforts toward the development of cancer therapies targeting the HH pathway. Challenges to develop next-generation cancer therapies are highlighted.
Collapse
Affiliation(s)
- Fujia Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bo Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
217
|
Monkkonen T, Landua JD, Visbal AP, Lewis MT. Epithelial and non-epithelial Ptch1 play opposing roles to regulate proliferation and morphogenesis of the mouse mammary gland. Development 2017; 144:1317-1327. [PMID: 28275010 PMCID: PMC5399619 DOI: 10.1242/dev.140434] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 02/07/2017] [Indexed: 12/14/2022]
Abstract
Patched 1 (Ptch1) has epithelial, stromal and systemic roles in murine mammary gland organogenesis, yet specific functions remain undefined. Cre-recombinase-mediated Ptch1 ablation in mammary epithelium increased proliferation and branching, but did not phenocopy transgenic expression of activated smoothened (SmoM2). The epithelium showed no evidence of canonical hedgehog signaling, and hyperproliferation was not blocked by smoothened (SMO) inhibition, suggesting a non-canonical function of PTCH1. Consistent with this possibility, nuclear localization of cyclin B1 was increased. In non-epithelial cells, heterozygous Fsp-Cre-mediated Ptch1 ablation increased proliferation and branching, with dysplastic terminal end buds (TEB) and ducts. By contrast, homozygous Ptch1 ablation decreased proliferation and branching, producing stunted ducts filled with luminal cells showing altered ovarian hormone receptor expression. Whole-gland transplantation into wild-type hosts or estrogen/progesterone treatment rescued outgrowth and hormone receptor expression, but not the histological changes. Bone marrow transplantation failed to rescue outgrowth. Ducts of Fsp-Cre;Ptch1fl/fl mice were similar to Fsp-Cre;SmoM2 ducts, but Fsp-Cre;SmoM2 outgrowths were not stunted, suggesting that the histology might be mediated by Smo in the local stroma, with systemic Ptch1 required for ductal outgrowth and proper hormone receptor expression in the mammary epithelium. Summary: Systemic and tissue-specific depletion of patched 1 in epithelial and stromal compartments of the mammary gland defines functions in ductal patterning, proliferation and gene expression.
Collapse
Affiliation(s)
- Teresa Monkkonen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - John D Landua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Adriana P Visbal
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA .,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
218
|
Diarylheptanoids suppress proliferation of pancreatic cancer PANC-1 cells through modulating shh-Gli-FoxM1 pathway. Arch Pharm Res 2017; 40:509-517. [PMID: 28258481 DOI: 10.1007/s12272-017-0905-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 02/22/2017] [Indexed: 12/18/2022]
Abstract
Pancreatic cancer is one of the leading causes of cancer, and it has the lowest 5-year survival rates. It is necessary to develop more potent anti-pancreatic cancer drugs to overcome the fast metastasis and resistance to surgery, radiotherapy, chemotherapy, and combinations of these. We have identified several diarylheptanoids as anti-pancreatic cancer agents from Alpinia officinarum (lesser galangal) and Alnus japonica. These diarylheptanoids suppressed cell proliferation and induced the cell cycle arrest of pancreatic cancer cells (PANC-1). Among them, the most potent compounds 1 and 7 inhibited the shh-Gli-FoxM1 pathway and their target gene expression in PANC-1 cells. Furthermore, they suppressed the expression of the cell cycle associated genes that were rescued by the overexpression of exogenous FoxM1. Taken together, (E)-7-(4-hydroxy-3-methoxyphenyl)-1-phenylhept-4-en-3-one (1) from Alpinia officinarum (lesser galangal) and platyphyllenone (7) from Alnus japonica inhibit PANC-1 cell proliferation by suppressing the shh-Gli-FoxM1 pathway, and they can be potential candidates for anti-pancreatic cancer drug development.
Collapse
|
219
|
Jia G, Chandriani S, Abbas AR, DePianto DJ, N'Diaye EN, Yaylaoglu MB, Moore HM, Peng I, DeVoss J, Collard HR, Wolters PJ, Egen JG, Arron JR. CXCL14 is a candidate biomarker for Hedgehog signalling in idiopathic pulmonary fibrosis. Thorax 2017; 72:780-787. [PMID: 28250200 DOI: 10.1136/thoraxjnl-2015-207682] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 01/31/2017] [Accepted: 02/03/2017] [Indexed: 11/04/2022]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is associated with aberrant expression of developmental pathways, including Hedgehog (Hh). As Hh signalling contributes to multiple pro-fibrotic processes, Hh inhibition may represent a therapeutic option for IPF. However, no non-invasive biomarkers are available to monitor lung Hh activity. METHODS We assessed gene and protein expression in IPF and control lung biopsies, mouse lung, fibroblasts stimulated in vitro with sonic hedgehog (SHh), and plasma in IPF patients versus controls, and cancer patients before and after treatment with vismodegib, a Hh inhibitor. RESULTS Lung tissue from IPF patients exhibited significantly greater expression of Hh-related genes versus controls. The gene most significantly upregulated in both IPF lung biopsies and fibroblasts stimulated in vitro with SHh was CXCL14, which encodes a soluble secreted chemokine whose expression is inhibited in vitro by the addition of vismodegib. CXCL14 expression was induced by SHh overexpression in mouse lung. Circulating CXCL14 protein levels were significantly higher in plasma from IPF patients than controls. In cancer patients, circulating CXCL14 levels were significantly reduced upon vismodegib treatment. CONCLUSIONS CXCL14 is a systemic biomarker that could be used to identify IPF patients with increased Hh pathway activity and monitor the pharmacodynamic effects of Hh antagonist therapy in IPF. TRIAL REGISTRATION NUMBER Post-results, NCT00968981.
Collapse
Affiliation(s)
- Guiquan Jia
- Genentech, Inc., South San Francisco, California, USA
| | | | | | | | | | | | | | - Ivan Peng
- Genentech, Inc., South San Francisco, California, USA
| | - Jason DeVoss
- Genentech, Inc., South San Francisco, California, USA
| | - Harold R Collard
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Paul J Wolters
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | | | | |
Collapse
|
220
|
Kambach DM, Halim AS, Cauer A, Sun Q, Tristan CA, Celiku O, Kesarwala AH, Shankavaram U, Batchelor E, Stommel JM. Disabled cell density sensing leads to dysregulated cholesterol synthesis in glioblastoma. Oncotarget 2017; 8:14860-14875. [PMID: 28118603 PMCID: PMC5362450 DOI: 10.18632/oncotarget.14740] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 01/10/2017] [Indexed: 01/09/2023] Open
Abstract
A hallmark of cellular transformation is the evasion of contact-dependent inhibition of growth. To find new therapeutic targets for glioblastoma, we looked for pathways that are inhibited by high cell density in astrocytes but not in glioma cells. Here we report that glioma cells have disabled the normal controls on cholesterol synthesis. At high cell density, astrocytes turn off cholesterol synthesis genes and have low cholesterol levels, but glioma cells keep this pathway on and maintain high cholesterol. Correspondingly, cholesterol pathway upregulation is associated with poor prognosis in glioblastoma patients. Densely-plated glioma cells increase oxygen consumption, aerobic glycolysis, and the pentose phosphate pathway to synthesize cholesterol, resulting in a decrease in reactive oxygen species, TCA cycle intermediates, and ATP. This constitutive cholesterol synthesis is controlled by the cell cycle, as it can be turned off by cyclin-dependent kinase inhibitors and it correlates with disabled cell cycle control though loss of p53 and RB. Finally, glioma cells, but not astrocytes, are sensitive to cholesterol synthesis inhibition downstream of the mevalonate pathway, suggesting that specifically targeting cholesterol synthesis might be an effective treatment for glioblastoma.
Collapse
Affiliation(s)
- Diane M. Kambach
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alan S. Halim
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - A.Gesine Cauer
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qian Sun
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carlos A. Tristan
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Orieta Celiku
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aparna H. Kesarwala
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eric Batchelor
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jayne M. Stommel
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
221
|
Polychronidou G, Karavasilis V, Pollack SM, Huang PH, Lee A, Jones RL. Novel therapeutic approaches in chondrosarcoma. Future Oncol 2017; 13:637-648. [PMID: 28133974 DOI: 10.2217/fon-2016-0226] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chondrosarcoma is a malignant tumor of bones, characterized by the production of cartilage matrix. Due to lack of effective treatment for advanced disease, the clinical management of chondrosarcomas is exceptionally challenging. Current research focuses on elucidating the molecular events underlying the pathogenesis of this rare bone malignancy, with the goal of developing new molecularly targeted therapies. Signaling pathways suggested to have a role in chondrosarcoma include Hedgehog, Src, PI3k-Akt-mTOR and angiogenesis. Mutations in IDH1/2, present in more than 50% of primary conventional chondrosarcomas, make the development of IDH inhibitors a promising treatment option. The present review discusses the preclinical and early clinical data on novel targeted therapeutic approaches in chondrosarcoma.
Collapse
Affiliation(s)
| | | | - Seth M Pollack
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Paul H Huang
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Alex Lee
- Sarcoma Unit, Royal Marsden Hospital, London, UK
| | - Robin L Jones
- Sarcoma Unit, Royal Marsden Hospital, London, UK.,Division of Clinical Studies, The Institute of Cancer Research, London, UK
| |
Collapse
|
222
|
Pietanza MC, Zimmerman S, Peters S, Curran WJ. Seeking New Approaches to Patients With Small Cell Lung Cancer. Am Soc Clin Oncol Educ Book 2017; 35:e477-82. [PMID: 27249756 DOI: 10.1200/edbk_158710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fundamental approach to the treatment of small cell lung cancer (SCLC) has not changed in the last several decades, with most advances being restricted to improved radiation approaches. The standard first-line chemotherapy regimen in the United States and Europe remains cisplatin or carboplatin plus etoposide in the treatment of limited stage (LS-SCLC) and extensive stage (ES-SCLC) disease. Radiation therapy is administered to those patients with LS-SCLC, whose cancer is confined to the chest in a single tolerable radiation field. This article will summarize a number of exciting observations regarding the biology of SCLC and how a deeper understanding of newly integrated targets and target pathways may lead to new and better therapeutic approaches in the near future.
Collapse
Affiliation(s)
- Marie Catherine Pietanza
- From the Merck Research Laboratories, Rahway, NJ; Department of Oncology, HFR Fribourg, Fribourg, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Winship Cancer Institute of Emory University, Atlanta, GA
| | - Stefan Zimmerman
- From the Merck Research Laboratories, Rahway, NJ; Department of Oncology, HFR Fribourg, Fribourg, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Winship Cancer Institute of Emory University, Atlanta, GA
| | - Solange Peters
- From the Merck Research Laboratories, Rahway, NJ; Department of Oncology, HFR Fribourg, Fribourg, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Winship Cancer Institute of Emory University, Atlanta, GA
| | - Walter J Curran
- From the Merck Research Laboratories, Rahway, NJ; Department of Oncology, HFR Fribourg, Fribourg, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Winship Cancer Institute of Emory University, Atlanta, GA
| |
Collapse
|
223
|
Bhattarai D, Jung JH, Han S, Lee H, Oh SJ, Ko HW, Lee K. Design, synthesis, and biological evaluation of structurally modified isoindolinone and quinazolinone derivatives as hedgehog pathway inhibitors. Eur J Med Chem 2017; 125:1036-1050. [DOI: 10.1016/j.ejmech.2016.10.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 09/13/2016] [Accepted: 10/17/2016] [Indexed: 01/19/2023]
|
224
|
The Hedgehog Signaling Networks in Lung Cancer: The Mechanisms and Roles in Tumor Progression and Implications for Cancer Therapy. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7969286. [PMID: 28105432 PMCID: PMC5220431 DOI: 10.1155/2016/7969286] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023]
Abstract
Lung cancer is the most common cause of cancer-related death worldwide and is classified into small cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC). Several gene mutations that contribute to aberrant cell proliferation have been identified in lung adenocarcinoma, a part of NSCLC. Various anticancer drugs that target these mutated molecules have been developed for NSCLC treatment. However, although molecularly targeted drugs are initially effective for patients, the 5-year survival rate remains low because of tumor relapse. Therefore, more effective drugs for lung cancer treatment should be developed. The hedgehog (HH) signaling pathway contributes to organ development and stem cell maintenance, and aberrant activation of this signaling pathway is observed in various cancers including lung cancer. In lung cancer, HH signaling pathway upregulates cancer cell proliferation and maintains cancer stem cells as well as cancer-associated fibroblasts (CAFs). Furthermore, physical contact between CAFs and NSCLC cells induces HH signaling pathway activation in NSCLC cells to enhance their metastatic potential. Therefore, HH signaling pathway inhibitors could be a useful option for lung cancer therapy.
Collapse
|
225
|
Liu X, Shen Q, Yu T, Huang H, Zhang Z, Ding J, Tang Y, Xu N, Yue S. Small GTPase Arl6 controls RH30 rhabdomyosarcoma cell growth through ciliogenesis and Hedgehog signaling. Cell Biosci 2016; 6:61. [PMID: 27999656 PMCID: PMC5154108 DOI: 10.1186/s13578-016-0126-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 11/28/2016] [Indexed: 12/04/2022] Open
Abstract
Background Rhabdomyosarcoma (RMS) originates from skeletal muscle precursors that fail to differentiate. Hedgehog (Hh) signaling and primary cilia contribute to the pathobiology of RMS. Results Here we showed ADP ribosylation factor like GTPase 6 (ARL6) localizes at the base of primary cilium, controls ciliogenesis and Hh signaling. The transcription of Arl6 is dynamic during the differentiation of myoblasts, companying with the growth and elimination of primary cilia. Arl6 expression is significantly up regulated in cilia-dependent RMS cells and tissues. Knockdown of Arl6 inhibits proliferation and promotes apoptosis of RMS RH30 cells through defected ciliogenesis and reduced Hh activity. Conclusions Taken together, the functions of Arl6 in ciliogenesis and Hh signaling suggest it as a potential RMS drug target. Electronic supplementary material The online version of this article (doi:10.1186/s13578-016-0126-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaotong Liu
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 Jiangsu People's Republic of China
| | - Qiuhong Shen
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 Jiangsu People's Republic of China
| | - Tingting Yu
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 Jiangsu People's Republic of China
| | - Huijie Huang
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 Jiangsu People's Republic of China
| | - Ziyu Zhang
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 Jiangsu People's Republic of China
| | - Jie Ding
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 Jiangsu People's Republic of China
| | - Ying Tang
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 Jiangsu People's Republic of China.,Central Laboratory, The First People's Hospital of Wujiang District, Suzhou, 215200 People's Republic of China
| | - Ning Xu
- Department of Pathology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 Jiangsu People's Republic of China
| | - Shen Yue
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166 Jiangsu People's Republic of China
| |
Collapse
|
226
|
Mangum R, Varga E, Boué DR, Capper D, Benesch M, Leonard J, Osorio DS, Pierson CR, Zumberge N, Sahm F, Schrimpf D, Pfister SM, Finlay JL. SHH desmoplastic/nodular medulloblastoma and Gorlin syndrome in the setting of Down syndrome: case report, molecular profiling, and review of the literature. Childs Nerv Syst 2016; 32:2439-2446. [PMID: 27444290 DOI: 10.1007/s00381-016-3185-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 07/07/2016] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Individuals with Down syndrome (DS) have an increased risk of acute leukemia compared to a markedly decreased incidence of solid tumors. Medulloblastoma, the most common malignant brain tumor of childhood, is particularly rare in the DS population, with only one published case. As demonstrated in a mouse model, DS is associated with cerebellar hypoplasia and a decreased number of cerebellar granule neuron progenitor cells (CGNPs) in the external granule cell layer (EGL). Treatment of these mice with sonic hedgehog signaling pathway (Shh) agonists promote normalization of CGNPs and improved cognitive functioning. CASE REPORT We describe a 21-month-old male with DS and concurrent desmoplastic/nodular medulloblastoma (DNMB)-a tumor derived from Shh dysregulation and over-activation of CGNPs. Molecular profiling further classified the tumor into the new consensus SHH molecular subgroup. Additional testing revealed a de novo heterozygous germ line mutation in the PTCH1 gene encoding a tumor suppressor protein in the Shh pathway. DISCUSSION The developmental failure of CGNPs in DS patients offers a plausible explanation for the rarity of medulloblastoma in this population. Conversely, patients with PTCH1 germline mutations experience Shh overstimulation resulting in Gorlin (Nevoid Basal Cell Carcinoma) syndrome and an increased incidence of malignant transformation of CGNPs leading to medulloblastoma formation. This represents the first documented report of an individual with DS simultaneously carrying PTCH1 germline mutation. CONCLUSION We have observed a highly unusual circumstance in which the PTCH1 mutation appears to "trump" the effects of DS in causation of Shh-activated medulloblastoma.
Collapse
Affiliation(s)
- Ross Mangum
- The Divisions of Hematology/Oncology/BMT, Neurosurgery and Neuropathology, the Departments of Pediatrics, Surgery and Pathology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA.
| | - Elizabeth Varga
- The Divisions of Hematology/Oncology/BMT, Neurosurgery and Neuropathology, the Departments of Pediatrics, Surgery and Pathology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - Daniel R Boué
- The Divisions of Hematology/Oncology/BMT, Neurosurgery and Neuropathology, the Departments of Pediatrics, Surgery and Pathology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - David Capper
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Benesch
- Division of Pediatric Hematology/Oncology, Medical University of Graz, Graz, Austria
| | - Jeffrey Leonard
- The Divisions of Hematology/Oncology/BMT, Neurosurgery and Neuropathology, the Departments of Pediatrics, Surgery and Pathology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - Diana S Osorio
- The Divisions of Hematology/Oncology/BMT, Neurosurgery and Neuropathology, the Departments of Pediatrics, Surgery and Pathology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - Christopher R Pierson
- The Divisions of Hematology/Oncology/BMT, Neurosurgery and Neuropathology, the Departments of Pediatrics, Surgery and Pathology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - Nicholas Zumberge
- The Divisions of Hematology/Oncology/BMT, Neurosurgery and Neuropathology, the Departments of Pediatrics, Surgery and Pathology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - Felix Sahm
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Schrimpf
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan M Pfister
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jonathan L Finlay
- The Divisions of Hematology/Oncology/BMT, Neurosurgery and Neuropathology, the Departments of Pediatrics, Surgery and Pathology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| |
Collapse
|
227
|
Pavan AR, Silva GDBD, Jornada DH, Chiba DE, Fernandes GFDS, Man Chin C, Dos Santos JL. Unraveling the Anticancer Effect of Curcumin and Resveratrol. Nutrients 2016; 8:nu8110628. [PMID: 27834913 PMCID: PMC5133053 DOI: 10.3390/nu8110628] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 09/24/2016] [Accepted: 09/27/2016] [Indexed: 12/16/2022] Open
Abstract
Resveratrol and curcumin are natural products with important therapeutic properties useful to treat several human diseases, including cancer. In the last years, the number of studies describing the effect of both polyphenols against cancer has increased; however, the mechanism of action in all of those cases is not completely comprehended. The unspecific effect and the ability to interfere in assays by both polyphenols make this challenge even more difficult. Herein, we analyzed the anticancer activity of resveratrol and curcumin reported in the literature in the last 11 years, in order to unravel the molecular mechanism of action of both compounds. Molecular targets and cellular pathways will be described. Furthermore, we also discussed the ability of these natural products act as chemopreventive and its use in association with other anticancer drugs.
Collapse
Affiliation(s)
- Aline Renata Pavan
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| | | | | | - Diego Eidy Chiba
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| | | | - Chung Man Chin
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| | - Jean Leandro Dos Santos
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| |
Collapse
|
228
|
Rifai Y, Tani HB, Nur M, Aswad M, Lallo S, Wahyudin E. Synthesis, Molecular Mechanism and Pharmacokinetic Studies of New Epoxy Lignan-Based Derivatives. Arch Pharm (Weinheim) 2016; 349:848-852. [PMID: 27699826 DOI: 10.1002/ardp.201600069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 09/16/2016] [Accepted: 09/16/2016] [Indexed: 01/14/2023]
Abstract
The oxidative demethylation procedure for a new epoxy lignan isolated from Piper nigrum was applied to the synthesis of 3'-methoxy-3",4"-(methylenedioxy)-2,5-epoxylignan-4'-ol-6'-one. This compound inhibited the mRNA expression of the protein patched homolog (Ptch) in human pancreatic cancer cells (PANC1) and therefore might be valuable as a probe for tumor-related disease. The pharmacokinetic profile of 3'-methoxy-3",4"-(methylenedioxy)-2,5-epoxylignan-4'-ol-6'-one was rapidly determined using ultra-fast liquid chromatography. The compound was rapidly absorbed in blood.
Collapse
Affiliation(s)
- Yusnita Rifai
- Pharmaceutical Chemistry Laboratory Faculty of Pharmacy Hasanuddin University, Makassar, Indonesia.
| | - Harold B Tani
- Pharmaceutical Chemistry Laboratory Faculty of Pharmacy Hasanuddin University, Makassar, Indonesia
| | - Muhammad Nur
- Pharmaceutical Chemistry Laboratory Faculty of Pharmacy Hasanuddin University, Makassar, Indonesia
| | - Muhammad Aswad
- Pharmaceutical Chemistry Laboratory Faculty of Pharmacy Hasanuddin University, Makassar, Indonesia
| | - Subehan Lallo
- Pharmaceutical Chemistry Laboratory Faculty of Pharmacy Hasanuddin University, Makassar, Indonesia
| | - Elly Wahyudin
- Pharmaceutical Chemistry Laboratory Faculty of Pharmacy Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
229
|
Schimmack S, Kneller S, Dadabaeva N, Bergmann F, Taylor A, Hackert T, Werner J, Strobel O. Epithelial to Stromal Re-Distribution of Primary Cilia during Pancreatic Carcinogenesis. PLoS One 2016; 11:e0164231. [PMID: 27783689 PMCID: PMC5081192 DOI: 10.1371/journal.pone.0164231] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/21/2016] [Indexed: 12/30/2022] Open
Abstract
Background The Hedgehog (HH) pathway is a mediator in pancreatic ductal adenocarcinoma (PDAC). Surprisingly, previous studies suggested that primary cilia (PC), the essential organelles for HH signal transduction, were lost in PDAC. The aim of this study was to determine the presence of PC in human normal pancreas, chronic pancreatitis, and during carcinogenesis to PDAC with focus on both epithelia and stroma. Methods PC were analyzed in paraffin sections from normal pancreas, chronic pancreatitis, intraductal papillary-mucinous neoplasia, and PDAC, as well as in primary human pancreatic stellate cells (PSC) and pancreatic cancer cell lines by double immunofluorescence staining for acetylated α-tubuline and γ-tubuline. Co-staining for the HH receptors PTCH1, PTCH2 and SMO was also performed. Results PC are gradually lost during pancreatic carcinogenesis in the epithelium: the fraction of cells with PC gradually and significantly decreased from 32% in ducts of normal pancreas, to 21% in ducts of chronic pancreatitis, to 18% in PanIN1a, 6% in PanIN2, 3% in PanIN3 and to 1.2% in invasive PDAC. However, this loss of PC in the neoplastic epithelium is accompanied by a gain of PC in the surrounding stroma. The fraction of stromal cells with PC significantly increased from 13% around normal ducts to about 30% around PanIN and PDAC. HH-receptors were detected in tumor stroma but not in epithelial cells. PC are also present in PSC and pancreatic cancer cell lines. Conclusion PC are not lost during pancreatic carcinogenesis but re-distributed from the epithelium to the stroma. This redistribution may explain the re-direction of HH signaling towards the stroma during pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Simon Schimmack
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Sarah Kneller
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Nigora Dadabaeva
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Frank Bergmann
- Institute of Pathology Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Andrew Taylor
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Jens Werner
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
- University Hospital of General, Visceral, Transplantation, Vascular and Thoracic Surgery of Munich, Campus Großhadern, Marchioninistraße 15, 81377 Munich, Germany
| | - Oliver Strobel
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
- * E-mail:
| |
Collapse
|
230
|
Abstract
The casein kinase 1 (CK1) family of serine (Ser)/threonine (Thr) protein kinases participates in a myriad of cellular processes including developmental signaling. Hedgehog (Hh) and Wnt pathways are two major and evolutionarily conserved signaling pathways that control embryonic development and adult tissue homeostasis. Deregulation of these pathways leads to many human disorders including birth defects and cancer. Here, I review the role of CK1 in the regulation of Hh and Wnt signal transduction cascades from the membrane reception systems to the transcriptional effectors. In both Hh and Wnt pathways, multiple CK1 family members regulate signal transduction at several levels of the pathways and play either positive or negative roles depending on the signaling status, individual CK1 isoforms involved, and the specific substrates they phosphorylate. A common mechanism underlying the control of CK1-mediated phosphorylation of Hh and Wnt pathway components is the regulation of CK1/substrate interaction within large protein complexes. I will highlight this feature in the context of Hh signaling and draw interesting parallels between the Hh and Wnt pathways.
Collapse
Affiliation(s)
- Jin Jiang
- University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States.
| |
Collapse
|
231
|
Gao L, Chen F, Li X, Xu S, Huang W, Ye Y. Three new alkaloids from Veratrum grandiflorum Loes with inhibition activities on Hedgehog pathway. Bioorg Med Chem Lett 2016; 26:4735-4738. [DOI: 10.1016/j.bmcl.2016.08.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/10/2016] [Accepted: 08/13/2016] [Indexed: 10/21/2022]
|
232
|
Chen J, Lv H, Hu J, Ji M, Xue N, Li C, Ma S, Zhou Q, Lin B, Li Y, Yu S, Chen X. CAT3, a novel agent for medulloblastoma and glioblastoma treatment, inhibits tumor growth by disrupting the Hedgehog signaling pathway. Cancer Lett 2016; 381:391-403. [DOI: 10.1016/j.canlet.2016.07.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 05/31/2016] [Accepted: 07/21/2016] [Indexed: 01/20/2023]
|
233
|
Perli SD, Cui CH, Lu TK. Continuous genetic recording with self-targeting CRISPR-Cas in human cells. Science 2016; 353:aag0511. [PMID: 27540006 DOI: 10.1126/science.aag0511] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/27/2016] [Indexed: 01/02/2023]
Abstract
The ability to record molecular events in vivo would enable monitoring of signaling dynamics within cellular niches and critical factors that orchestrate cellular behavior. We present a self-contained analog memory device for longitudinal recording of molecular stimuli into DNA mutations in human cells. This device consists of a self-targeting guide RNA (stgRNA) that repeatedly directs Streptococcus pyogenes Cas9 nuclease activity toward the DNA that encodes the stgRNA, enabling localized, continuous DNA mutagenesis as a function of stgRNA expression. We demonstrate programmable and multiplexed memory storage in human cells triggered by exogenous inducers or inflammation, both in vitro and in vivo. This tool, Mammalian Synthetic Cellular Recorder Integrating Biological Events (mSCRIBE), provides a distinct strategy for investigating cell biology in vivo and enables continuous evolution of targeted DNA sequences.
Collapse
Affiliation(s)
- Samuel D Perli
- Synthetic Biology Group, MIT Synthetic Biology Center, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Research Laboratory of Electronics, MIT, Cambridge, MA 02139, USA. Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139, USA
| | - Cheryl H Cui
- Synthetic Biology Group, MIT Synthetic Biology Center, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Research Laboratory of Electronics, MIT, Cambridge, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Timothy K Lu
- Synthetic Biology Group, MIT Synthetic Biology Center, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Research Laboratory of Electronics, MIT, Cambridge, MA 02139, USA. Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139, USA. Department of Biological Engineering, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
234
|
Tamoxifen Resistance: Emerging Molecular Targets. Int J Mol Sci 2016; 17:ijms17081357. [PMID: 27548161 PMCID: PMC5000752 DOI: 10.3390/ijms17081357] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 12/12/2022] Open
Abstract
17β-Estradiol (E2) plays a pivotal role in the development and progression of breast cancer. As a result, blockade of the E2 signal through either tamoxifen (TAM) or aromatase inhibitors is an important therapeutic strategy to treat or prevent estrogen receptor (ER) positive breast cancer. However, resistance to TAM is the major obstacle in endocrine therapy. This resistance occurs either de novo or is acquired after an initial beneficial response. The underlying mechanisms for TAM resistance are probably multifactorial and remain largely unknown. Considering that breast cancer is a very heterogeneous disease and patients respond differently to treatment, the molecular analysis of TAM’s biological activity could provide the necessary framework to understand the complex effects of this drug in target cells. Moreover, this could explain, at least in part, the development of resistance and indicate an optimal therapeutic option. This review highlights the implications of TAM in breast cancer as well as the role of receptors/signal pathways recently suggested to be involved in the development of TAM resistance. G protein—coupled estrogen receptor, Androgen Receptor and Hedgehog signaling pathways are emerging as novel therapeutic targets and prognostic indicators for breast cancer, based on their ability to mediate estrogenic signaling in ERα-positive or -negative breast cancer.
Collapse
|
235
|
Angelaud R, Reynolds M, Venkatramani C, Savage S, Trafelet H, Landmesser T, Demel P, Levis M, Ruha O, Rueckert B, Jaeggi H. Manufacturing Development and Genotoxic Impurity Control Strategy of the Hedgehog Pathway Inhibitor Vismodegib. Org Process Res Dev 2016. [DOI: 10.1021/acs.oprd.6b00208] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | | | | | | | - Peter Demel
- Siegfried AG, Untere Brühlstrasse
4, CH-4800 Zofingen, Switzerland
| | - Michael Levis
- Siegfried AG, Untere Brühlstrasse
4, CH-4800 Zofingen, Switzerland
| | - Olivier Ruha
- Siegfried AG, Untere Brühlstrasse
4, CH-4800 Zofingen, Switzerland
| | - Baerbel Rueckert
- Siegfried AG, Untere Brühlstrasse
4, CH-4800 Zofingen, Switzerland
| | - Heinz Jaeggi
- Siegfried AG, Untere Brühlstrasse
4, CH-4800 Zofingen, Switzerland
| |
Collapse
|
236
|
Liu R, Wong W, IJzerman AP. Human G protein-coupled receptor studies in Saccharomyces cerevisiae. Biochem Pharmacol 2016; 114:103-15. [DOI: 10.1016/j.bcp.2016.02.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/12/2016] [Indexed: 12/22/2022]
|
237
|
Schröder P, Bauer JO, Strohmann C, Kumar K, Waldmann H. Synthesis of an Iridoid-Inspired Compound Collection and Discovery of Autophagy Inhibitors. J Org Chem 2016; 81:10242-10255. [DOI: 10.1021/acs.joc.6b01185] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Peter Schröder
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Jonathan O. Bauer
- Faculty
of Chemistry and Chemical Biology, Technical University Dortmund, Otto-Hahn-Straßa 6, 44221 Dortmund, Germany
| | - Carsten Strohmann
- Faculty
of Chemistry and Chemical Biology, Technical University Dortmund, Otto-Hahn-Straßa 6, 44221 Dortmund, Germany
| | - Kamal Kumar
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Herbert Waldmann
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Faculty
of Chemistry and Chemical Biology, Technical University Dortmund, Otto-Hahn-Straßa 6, 44221 Dortmund, Germany
| |
Collapse
|
238
|
Houot R, Soussain C, Tilly H, Haioun C, Thieblemont C, Casasnovas O, Bouabdallah K, Morschhauser F, Le Gouill S, Salles G, Hoang-Xuan K, Choquet S, Marchand T, Laurent C, Pangault C, Lamy T. Inhibition of Hedgehog signaling for the treatment of lymphoma and CLL: a phase II study from the LYSA. Ann Oncol 2016; 27:1349-50. [DOI: 10.1093/annonc/mdw138] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
239
|
Kötzner L, Leutzsch M, Sievers S, Patil S, Waldmann H, Zheng Y, Thiel W, List B. Organokatalytische Synthese von enantiomerenreinen 2H- und 3H-Pyrrolen: Inhibitoren des Hedgehog-Signalwegs. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201602932] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Lisa Kötzner
- Max-Planck-Institut für Kohlenforschung; Kaiser-Wilhelm-Platz 1 45470 Mülheim an der Ruhr Deutschland
| | - Markus Leutzsch
- Max-Planck-Institut für Kohlenforschung; Kaiser-Wilhelm-Platz 1 45470 Mülheim an der Ruhr Deutschland
| | - Sonja Sievers
- Max-Planck-Institut für Molekulare Physiologie; Compound Management and Screening Center (COMAS); Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Sumersing Patil
- Max-Planck-Institut für Molekulare Physiologie; Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Herbert Waldmann
- Max-Planck-Institut für Molekulare Physiologie; Otto-Hahn-Straße 11 44227 Dortmund Deutschland
| | - Yiying Zheng
- Max-Planck-Institut für Kohlenforschung; Kaiser-Wilhelm-Platz 1 45470 Mülheim an der Ruhr Deutschland
| | - Walter Thiel
- Max-Planck-Institut für Kohlenforschung; Kaiser-Wilhelm-Platz 1 45470 Mülheim an der Ruhr Deutschland
| | - Benjamin List
- Max-Planck-Institut für Kohlenforschung; Kaiser-Wilhelm-Platz 1 45470 Mülheim an der Ruhr Deutschland
| |
Collapse
|
240
|
Kötzner L, Leutzsch M, Sievers S, Patil S, Waldmann H, Zheng Y, Thiel W, List B. The Organocatalytic Approach to Enantiopure 2H- and 3H-Pyrroles: Inhibitors of the Hedgehog Signaling Pathway. Angew Chem Int Ed Engl 2016; 55:7693-7. [DOI: 10.1002/anie.201602932] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Lisa Kötzner
- Max-Planck-Institut für Kohlenforschung; Kaiser Wilhelm-Platz 1 45470 Mülheim an der Ruhr Germany
| | - Markus Leutzsch
- Max-Planck-Institut für Kohlenforschung; Kaiser Wilhelm-Platz 1 45470 Mülheim an der Ruhr Germany
| | - Sonja Sievers
- Max-Planck-Institut für Molekulare Physiologie; Compound Management and Screening Center (COMAS); Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Sumersing Patil
- Max-Planck-Institut für Molekulare Physiologie; Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Herbert Waldmann
- Max-Planck-Institut für Molekulare Physiologie; Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Yiying Zheng
- Max-Planck-Institut für Kohlenforschung; Kaiser Wilhelm-Platz 1 45470 Mülheim an der Ruhr Germany
| | - Walter Thiel
- Max-Planck-Institut für Kohlenforschung; Kaiser Wilhelm-Platz 1 45470 Mülheim an der Ruhr Germany
| | - Benjamin List
- Max-Planck-Institut für Kohlenforschung; Kaiser Wilhelm-Platz 1 45470 Mülheim an der Ruhr Germany
| |
Collapse
|
241
|
Gonnissen A, Isebaert S, Haustermans K. Targeting the Hedgehog signaling pathway in cancer: beyond Smoothened. Oncotarget 2016; 6:13899-913. [PMID: 26053182 PMCID: PMC4546439 DOI: 10.18632/oncotarget.4224] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/13/2015] [Indexed: 12/20/2022] Open
Abstract
An essential role for Hedgehog (Hh) signaling in human cancer has been established beyond doubt. At present, targeting Hh signaling has mainly been investigated with SMO inhibitors. Unfortunately, resistance against currently used SMO inhibitors has already been observed in basal cell carcinoma (BCC) patients. Therefore, the use of Hh inhibitors targeting the signaling cascade more downstream of SMO could represent a more promising strategy. Furthermore, besides the classical canonical way of Hh signaling activation, non-canonical activation of the GLI transcription factors by multiple important signaling pathways (e.g. MAPK, PI3K, TGFβ) has also been described, pinpointing the importance of targeting the transcription factors GLI1/2. The most promising agent in this context is probably the GLI1/2 inhibitor GANT61 which has been investigated preclinically in numerous tumor types in the last few years. In this review, the emerging role of Hh signaling in cancer is critically evaluated focusing on the potential of targeting Hh signaling more downstream of SMO, i.e. at the level of the GLI transcription factors. Furthermore, the working mechanism and therapeutic potential of the most extensively studied GLI inhibitor in human cancer, i.e. GANT61, is discussed in detail. In conclusion, GANT61 appears to be highly effective against human cancer cells and in xenograft mouse models, targeting almost all of the classical hallmarks of cancer and could hence represent a promising treatment option for human cancer.
Collapse
Affiliation(s)
- Annelies Gonnissen
- University of Leuven (KU Leuven), Department of Oncology, Laboratory of Experimental Radiotherapy, Leuven, Belgium
| | - Sofie Isebaert
- University of Leuven (KU Leuven), Department of Oncology, Laboratory of Experimental Radiotherapy, Leuven, Belgium
| | - Karin Haustermans
- University of Leuven (KU Leuven), Department of Oncology, Laboratory of Experimental Radiotherapy, Leuven, Belgium.,University Hospitals Leuven, Department of Radiation Oncology, Leuven, Belgium
| |
Collapse
|
242
|
Belani CP, Dahlberg SE, Rudin CM, Fleisher M, Chen HX, Takebe N, Velasco MR, Tester WJ, Sturtz K, Hann CL, Shanks JC, Monga M, Ramalingam SS, Schiller JH. Vismodegib or cixutumumab in combination with standard chemotherapy for patients with extensive-stage small cell lung cancer: A trial of the ECOG-ACRIN Cancer Research Group (E1508). Cancer 2016; 122:2371-8. [PMID: 27163943 DOI: 10.1002/cncr.30062] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/30/2016] [Accepted: 04/01/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND Preclinical targeting of the hedgehog pathway by vismodegib and of insulin-like growth factor 1 receptor by cixutumumab enhances the efficacy of chemotherapy and also demonstrates activity against the tumor cell fraction responsible for disease recurrence in small cell lung cancer. METHODS Patients with newly diagnosed extensive-stage small cell lung cancer (SCLC-ED) were randomized to receive four 21-day cycles of cisplatin and etoposide alone (cisplatin at 75 mg/m(2) on day 1 and etoposide at 100 mg/m(2) on days 1-3; arm A) or in combination with either vismodegib (150 mg/d by mouth; arm B) or cixutumumab (6 mg/kg/wk intravenously on day 1; arm C). The primary endpoint was progression-free survival (PFS). Circulating tumor cells (CTCs) were isolated/enumerated with the Veridex CellSearch platform at the baseline. RESULTS One hundred fifty-two eligible patients were treated. Patient demographics and disease characteristics were well balanced between the 3 arms except for the higher rate with a performance status of 0 in arm B (P = .03). The median PFS times in arms A, B, and C were 4.4, 4.4, and 4.6 months, respectively; the median overall survival (OS) times were 8.8, 9.8, and 10.1 months, respectively; and the response rates were 48%, 56%, and 50%, respectively. None of the comparisons of these outcomes were statistically significant. The median OS was 10.5 months for those with low CTC counts (≤100/7.5 mL) at baseline and 7.2 months for those with high CTC counts (hazard ratio, 1.74; P = .006). CONCLUSIONS There was no significant improvement in PFS or OS with the addition of either vismodegib or cixutumumab to chemotherapy in patients with SCLC-ED. A low baseline CTC count was associated with a favorable prognosis. Cancer 2016;122:2371-2378. © 2016 American Cancer Society.
Collapse
Affiliation(s)
| | - Suzanne E Dahlberg
- ECOG-ACRIN Biostatistics Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | | | | | | | | | - Keren Sturtz
- Colorado Cancer Research Program, Denver, Colorado
| | | | | | - Manish Monga
- West Virginia University, Morgantown, West Virginia
| | | | - Joan H Schiller
- University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
243
|
Fu L, Lv X, Xiong Y, Zhao Y. Investigation of Protein-Protein Interactions and Conformational Changes in Hedgehog Signaling Pathway by FRET. Methods Mol Biol 2016; 1322:61-70. [PMID: 26179039 DOI: 10.1007/978-1-4939-2772-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Protein-protein interactions and signal-induced protein conformational changes are fundamental molecular events that are considered as essential in modern life sciences. Among various techniques developed to study such phenomena, fluorescence resonance energy transfer (FRET) is a widely used method with many advantages in detecting these molecular events. Here, we describe the application of FRET in the mechanistic investigation of cell signal transduction, taking the example of the Hh signaling pathway, which plays a critical role in embryonic development and tissue homeostasis. A number of general guidelines as well as some key notes have been summarized as a protocol for reader's reference.
Collapse
Affiliation(s)
- Lin Fu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, People's Republic of China
| | | | | | | |
Collapse
|
244
|
Shi Y, Gao S. Recent advances of synthesis of fluorenone and fluorene containing natural products. Tetrahedron 2016. [DOI: 10.1016/j.tet.2016.02.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
245
|
Abstract
Cancer poses a serious health problem in society and is increasingly surpassing cardiovascular disease as the leading cause of mortality in the United States. Current therapeutic strategies for cancer are extreme and harsh to patients and often have limited success; the danger of cancer is intensified as it metastasizes to secondary locations such as lung, bone, and liver, posing a dire threat to patient treatment and survival. Hedgehog signaling is an important pathway for normal development. Initially identified in Drosophila, the vertebrate and mammalian equivalent of the pathway has been studied extensively for its role in cancer development and progression. As this pathway regulates key target genes involved in development, its action also allows for the modulation of the microenvironment to prepare a tumor-suitable niche by manipulating tumor cell growth, differentiation, and immune regulation, thus creating an enabling environment for progression and metastasis. In this review, we will summarize recent scientific discoveries reporting the impact of the Hedgehog signaling pathway on the tumor initiation process and metastatic cascade, shedding light on the ability of the tumor to take over a mechanism crucially intended for development and normal function.
Collapse
Affiliation(s)
- Ann Hanna
- Department of Pathology and Comprehensive Cancer Center, The University of Alabama at Birmingham, Wallace Tumor Institute 320D, 1824 6th Avenue South, Birmingham, 35233, Alabama, USA
| | - Lalita A Shevde
- Department of Pathology and Comprehensive Cancer Center, The University of Alabama at Birmingham, Wallace Tumor Institute 320D, 1824 6th Avenue South, Birmingham, 35233, Alabama, USA.
| |
Collapse
|
246
|
Hanna A, Shevde LA. Hedgehog signaling: modulation of cancer properies and tumor mircroenvironment. Mol Cancer 2016; 15:24. [PMID: 26988232 PMCID: PMC4797362 DOI: 10.1186/s12943-016-0509-3] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/11/2016] [Indexed: 12/16/2022] Open
Abstract
Cancer poses a serious health problem in society and is increasingly surpassing cardiovascular disease as the leading cause of mortality in the United States. Current therapeutic strategies for cancer are extreme and harsh to patients and often have limited success; the danger of cancer is intensified as it metastasizes to secondary locations such as lung, bone, and liver, posing a dire threat to patient treatment and survival. Hedgehog signaling is an important pathway for normal development. Initially identified in Drosophila, the vertebrate and mammalian equivalent of the pathway has been studied extensively for its role in cancer development and progression. As this pathway regulates key target genes involved in development, its action also allows for the modulation of the microenvironment to prepare a tumor-suitable niche by manipulating tumor cell growth, differentiation, and immune regulation, thus creating an enabling environment for progression and metastasis. In this review, we will summarize recent scientific discoveries reporting the impact of the Hedgehog signaling pathway on the tumor initiation process and metastatic cascade, shedding light on the ability of the tumor to take over a mechanism crucially intended for development and normal function.
Collapse
Affiliation(s)
- Ann Hanna
- Department of Pathology and Comprehensive Cancer Center, The University of Alabama at Birmingham, Wallace Tumor Institute 320D, 1824 6th Avenue South, Birmingham, 35233, Alabama, USA
| | - Lalita A Shevde
- Department of Pathology and Comprehensive Cancer Center, The University of Alabama at Birmingham, Wallace Tumor Institute 320D, 1824 6th Avenue South, Birmingham, 35233, Alabama, USA.
| |
Collapse
|
247
|
Filocamo G, Brunetti M, Colaceci F, Sasso R, Tanori M, Pasquali E, Alfonsi R, Mancuso M, Saran A, Lahm A, Di Marcotullio L, Steinkühler C, Pazzaglia S. MK-4101, a Potent Inhibitor of the Hedgehog Pathway, Is Highly Active against Medulloblastoma and Basal Cell Carcinoma. Mol Cancer Ther 2016; 15:1177-89. [DOI: 10.1158/1535-7163.mct-15-0371] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 02/27/2016] [Indexed: 11/16/2022]
|
248
|
Christ A, Herzog K, Willnow TE. LRP2, an auxiliary receptor that controls sonic hedgehog signaling in development and disease. Dev Dyn 2016; 245:569-79. [PMID: 26872844 DOI: 10.1002/dvdy.24394] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/03/2016] [Accepted: 02/07/2016] [Indexed: 12/31/2022] Open
Abstract
To fulfill their multiple roles in organ development and adult tissue homeostasis, hedgehog (HH) morphogens act through their receptor Patched (PTCH) on target cells. However, HH actions also require HH binding proteins, auxiliary cell surface receptors that agonize or antagonize morphogen signaling in a context-dependent manner. Here, we discuss recent findings on the LDL receptor-related protein 2 (LRP2), an exemplary HH binding protein that modulates sonic hedgehog activities in stem and progenitor cell niches in embryonic and adult tissues. LRP2 functions are crucial for developmental processes in a number of tissues, including the brain, the eye, and the heart, and defects in this receptor pathway are the cause of devastating congenital diseases in humans. Developmental Dynamics 245:569-579, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Annabel Christ
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Katja Herzog
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| |
Collapse
|
249
|
Bai XY, Zhang XC, Yang SQ, An SJ, Chen ZH, Su J, Xie Z, Gou LY, Wu YL. Blockade of Hedgehog Signaling Synergistically Increases Sensitivity to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small-Cell Lung Cancer Cell Lines. PLoS One 2016; 11:e0149370. [PMID: 26943330 PMCID: PMC4778934 DOI: 10.1371/journal.pone.0149370] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 01/31/2016] [Indexed: 12/20/2022] Open
Abstract
Aberrant activation of the hedgehog (Hh) signaling pathway has been implicated in the epithelial-to-mesenchymal transition (EMT) and cancer stem-like cell (CSC) maintenance; both processes can result in tumor progression and treatment resistance in several types of human cancer. Hh cooperates with the epidermal growth factor receptor (EGFR) signaling pathway in embryogenesis. We found that the Hh signaling pathway was silenced in EGFR-TKI-sensitive non-small-cell lung cancer (NSCLC) cells, while it was inappropriately activated in EGFR-TKI-resistant NSCLC cells, accompanied by EMT induction and ABCG2 overexpression. Upregulation of Hh signaling through extrinsic SHH exposure downregulated E-cadherin expression and elevated Snail and ABCG2 expression, resulting in gefitinib tolerance (P < 0.001) in EGFR-TKI-sensitive cells. Blockade of the Hh signaling pathway using the SMO antagonist SANT-1 restored E-cadherin expression and downregulate Snail and ABCG2 in EGFR-TKI-resistant cells. A combination of SANT-1 and gefitinib markedly inhibited tumorigenesis and proliferation in EGFR-TKI-resistant cells (P < 0.001). These findings indicate that hyperactivity of Hh signaling resulted in EGFR-TKI resistance, by EMT introduction and ABCG2 upregulation, and blockade of Hh signaling synergistically increased sensitivity to EGFR-TKIs in primary and secondary resistant NSCLC cells. E-cadherin expression may be a potential biomarker of the suitability of the combined application of an Hh inhibitor and EGFR-TKIs in EGFR-TKI-resistant NSCLCs.
Collapse
Affiliation(s)
- Xiao-Yan Bai
- Department of Pulmonary Oncology, Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Science, Guangzhou 510080, China
| | - Xu-Chao Zhang
- Department of Pulmonary Oncology, Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Science, Guangzhou 510080, China
| | - Su-Qing Yang
- Department of Pulmonary Oncology, Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Science, Guangzhou 510080, China
| | - She-Juan An
- Department of Pulmonary Oncology, Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Science, Guangzhou 510080, China
| | - Zhi-Hong Chen
- Department of Pulmonary Oncology, Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Science, Guangzhou 510080, China
| | - Jian Su
- Department of Pulmonary Oncology, Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Science, Guangzhou 510080, China
| | - Zhi Xie
- Department of Pulmonary Oncology, Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Science, Guangzhou 510080, China
| | - Lan-Ying Gou
- Department of Pulmonary Oncology, Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Science, Guangzhou 510080, China
- Southern Medical University, Guangzhou 510515, China
| | - Yi-Long Wu
- Department of Pulmonary Oncology, Guangdong Lung Cancer Institute, Guangdong General Hospital & Guangdong Academy of Medical Science, Guangzhou 510080, China
- * E-mail:
| |
Collapse
|
250
|
Li Q, Liu Z, Xu M, Xue Y, Yao B, Dou C, Jia Y, Wang Y, Tu K, Zheng X, Yao Y. PCAF inhibits hepatocellular carcinoma metastasis by inhibition of epithelial-mesenchymal transition by targeting Gli-1. Cancer Lett 2016; 375:190-198. [PMID: 26945969 DOI: 10.1016/j.canlet.2016.02.053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/24/2016] [Accepted: 02/24/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED The p300-CBP-associated factor (PCAF), other than its histone acetyltransferase (HAT) activity, possesses an intrinsic ubiquitination activity that is involved in various transcriptional regulators, including the transcription factor glioma-associated oncogene 1 (Gli1), a well-known regulator of epithelial-mesenchymal transition (EMT) in cancer. In present research, we detected that PCAF was down-regulated in hepatocellular carcinoma (HCC) tissues compared with the adjacent non-tumor tissues and significantly associated with malignant portal vein invasion (p < 0.05) and poor survival (p < 0.05) of HCC patients. Moreover, functional study demonstrated that downregulation of PCAF facilitated tumor cell migration, invasion via EMT. Further study found that Gli1 as a direct target of PCAF induced EMT and promoted tumor metastasis and invasion. CONCLUSION PCAF is an anti-oncogene that plays an important role in the development of HCC by suppressing HCC cell metastasis and EMT by targeting Gli1, which indicates the potential therapeutic value of PCAF for suppression of metastasis of HCC.
Collapse
Affiliation(s)
- Qing Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhikui Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Meng Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yumo Xue
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Bowen Yao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Changwei Dou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuli Jia
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yufeng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Xin Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Yingmin Yao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|