1
|
Mechanism of Peitu Shengjin Formula Shenlingbaizhu Powder in Treating Bronchial Asthma and Allergic Colitis through Different Diseases with Simultaneous Treatment Based on Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4687788. [PMID: 35586697 PMCID: PMC9110165 DOI: 10.1155/2022/4687788] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/08/2022] [Accepted: 03/26/2022] [Indexed: 11/18/2022]
Abstract
Background Shenlingbaizhu powder (SLBZP), one of the classic Earth-cultivating and gold-generating prescriptions of traditional Chinese medicine, is widely used to treat various diseases. However, the pharmacological mechanisms of SLBZP on bronchial asthma (BA) and allergic colitis (AC) remain to be elucidated. Methods Network pharmacology and molecular docking technology were used to explore the potential mechanism of SLBZP in treating BA and AC with the simultaneous treatment of different diseases. The potential active compounds of SLBZP and their corresponding targets were obtained from BATMAN-TCM, ETCM, SymMap TCM@TAIWAN, and TCMSP databases. BA and AC disease targets were collected through DisGeNET, TTD, GeneCards, PharmGKB, OMIM, NCBI, The Human Phenotype Ontology, and DrugBank databases. Common targets for drugs and diseases were screened by using the bioinformatics and evolutionary genomics platform. The analyses and visualizations of Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment of common targets were carried out by R software. The key targets were screened by using the plug-in “cytoHubba” of Cytoscape software, and the “active compound-key target” network was constructed. Molecular docking analysis was performed using AutoDock software. The miRTarBase database was used to predict microRNAs (miRNAs) targeting key targets, and the key target-miRNA network was constructed. Result Through screening, 246 active compounds and 281 corresponding targets were obtained. Common targets were mainly enriched in 2933 biological processes and 182 signal pathways to play the role of treating BA and AC. There were 131 active compounds related to key targets. The results of molecular docking showed that the important active compounds in SLBZP had good binding ability with the key targets. The key target-miRNA network showed that 94 miRNAs were predicted. Conclusion SLBZP has played the role of treating different diseases with the same treatment on BA and AC through the characteristics of multicompound, multitarget, and multipathway of traditional Chinese medicine, which provides a theoretical basis for explaining the mechanism and clinical application of SLBZP treating different diseases with the same treatment in BA and AC.
Collapse
|
2
|
Goodman LD, Cope H, Nil Z, Ravenscroft TA, Charng WL, Lu S, Tien AC, Pfundt R, Koolen DA, Haaxma CA, Veenstra-Knol HE, Wassink-Ruiter JSK, Wevers MR, Jones M, Walsh LE, Klee VH, Theunis M, Legius E, Steel D, Barwick KES, Kurian MA, Mohammad SS, Dale RC, Terhal PA, van Binsbergen E, Kirmse B, Robinette B, Cogné B, Isidor B, Grebe TA, Kulch P, Hainline BE, Sapp K, Morava E, Klee EW, Macke EL, Trapane P, Spencer C, Si Y, Begtrup A, Moulton MJ, Dutta D, Kanca O, Wangler MF, Yamamoto S, Bellen HJ, Tan QKG. TNPO2 variants associate with human developmental delays, neurologic deficits, and dysmorphic features and alter TNPO2 activity in Drosophila. Am J Hum Genet 2021; 108:1669-1691. [PMID: 34314705 PMCID: PMC8456166 DOI: 10.1016/j.ajhg.2021.06.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/27/2021] [Indexed: 12/11/2022] Open
Abstract
Transportin-2 (TNPO2) mediates multiple pathways including non-classical nucleocytoplasmic shuttling of >60 cargoes, such as developmental and neuronal proteins. We identified 15 individuals carrying de novo coding variants in TNPO2 who presented with global developmental delay (GDD), dysmorphic features, ophthalmologic abnormalities, and neurological features. To assess the nature of these variants, functional studies were performed in Drosophila. We found that fly dTnpo (orthologous to TNPO2) is expressed in a subset of neurons. dTnpo is critical for neuronal maintenance and function as downregulating dTnpo in mature neurons using RNAi disrupts neuronal activity and survival. Altering the activity and expression of dTnpo using mutant alleles or RNAi causes developmental defects, including eye and wing deformities and lethality. These effects are dosage dependent as more severe phenotypes are associated with stronger dTnpo loss. Interestingly, similar phenotypes are observed with dTnpo upregulation and ectopic expression of TNPO2, showing that loss and gain of Transportin activity causes developmental defects. Further, proband-associated variants can cause more or less severe developmental abnormalities compared to wild-type TNPO2 when ectopically expressed. The impact of the variants tested seems to correlate with their position within the protein. Specifically, those that fall within the RAN binding domain cause more severe toxicity and those in the acidic loop are less toxic. Variants within the cargo binding domain show tissue-dependent effects. In summary, dTnpo is an essential gene in flies during development and in neurons. Further, proband-associated de novo variants within TNPO2 disrupt the function of the encoded protein. Hence, TNPO2 variants are causative for neurodevelopmental abnormalities.
Collapse
Affiliation(s)
- Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Heidi Cope
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Zelha Nil
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Thomas A Ravenscroft
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Wu-Lin Charng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - An-Chi Tien
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Rolph Pfundt
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, PO Box 9101, Nijmegen, the Netherlands
| | - David A Koolen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, PO Box 9101, Nijmegen, the Netherlands
| | - Charlotte A Haaxma
- Department of Pediatric Neurology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid 10, 6525 GA, PO Box 9101, the Netherlands
| | - Hermine E Veenstra-Knol
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Jolien S Klein Wassink-Ruiter
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Marijke R Wevers
- Department of Genetics, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Melissa Jones
- Houston Area Pediatric Neurology, 24514 Kingsland Blvd, Katy, TX 77494, USA
| | - Laurence E Walsh
- Department of Pediatric Neurology, Riley Hospital for Children, Indianapolis, IN 46202, USA
| | - Victoria H Klee
- Department of Pediatric Neurology, Riley Hospital for Children, Indianapolis, IN 46202, USA
| | - Miel Theunis
- Center for Human Genetics, University Hospital Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Eric Legius
- Department of Human Genetics, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Dora Steel
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; Department of Neurology, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Katy E S Barwick
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Manju A Kurian
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; Department of Neurology, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Shekeeb S Mohammad
- T.Y. Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Westmead, NSW 2145, Australia
| | - Russell C Dale
- T.Y. Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Westmead, NSW 2145, Australia
| | - Paulien A Terhal
- Department of Genetics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Ellen van Binsbergen
- Department of Genetics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Brian Kirmse
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bethany Robinette
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Benjamin Cogné
- Centre hospitalier universitaire (CHU) de Nantes, Service de Génétique Médicale, 9 quai Moncousu, 44093 Nantes, France; INSERM, CNRS, UNIV Nantes, Centre hospitalier universitaire (CHU) de Nantes, l'institut du thorax, 44007 Nantes, France
| | - Bertrand Isidor
- Centre hospitalier universitaire (CHU) de Nantes, Service de Génétique Médicale, 9 quai Moncousu, 44093 Nantes, France; INSERM, CNRS, UNIV Nantes, Centre hospitalier universitaire (CHU) de Nantes, l'institut du thorax, 44007 Nantes, France
| | - Theresa A Grebe
- Phoenix Children's Hospital, Phoenix, AZ 85016, USA; Department of Child Health, University of Arizona College of Medicine Phoenix, Phoenix, AZ 85004, USA
| | - Peggy Kulch
- Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - Bryan E Hainline
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Katherine Sapp
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eva Morava
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Erica L Macke
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Pamela Trapane
- University of Florida, College of Medicine, Jacksonville, Jacksonville, FL 32209, USA
| | - Christopher Spencer
- University of Florida, College of Medicine, Jacksonville, Jacksonville, FL 32209, USA
| | - Yue Si
- GeneDx, Gaithersburg, MD 20877, USA
| | | | - Matthew J Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Debdeep Dutta
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Queenie K-G Tan
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
3
|
Ge CY, Wei LY, Tian Y, Wang HH. A Seven-NF-κB-Related Gene Signature May Distinguish Patients with Ulcerative Colitis-Associated Colorectal Carcinoma. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:707-718. [PMID: 33299340 PMCID: PMC7719442 DOI: 10.2147/pgpm.s274258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/11/2020] [Indexed: 01/07/2023]
Abstract
Purpose Ulcerative colitis (UC) patients have an increased risk of colorectal cancer (CRC), and compared with sporadic CRC, ulcerative colitis-associated colorectal cancer (CAC) is more aggressive with a worse prognosis. This study aimed to identify a gene signature to predict the risk of CAC for patients with UC in remission. Patients and Methods Series of quiescent UC-related transcriptome data obtained from the Gene Expression Omnibus (GEO) data set were divided into a training set and a validation set. Gene Set Variation Analysis (GSVA), Gene Set Enrichment Analysis (GSEA), and \Weighted Correlation Network Analysis (WGCNA) combined with protein-protein interaction (PPI) analysis were used to identify the pathways and gene signatures related to tumorigenesis among quiescent UC patients. A generalized linear model (GLM) of Poisson regression based on the training set was applied to estimate the diagnostic power of the gene signature in our validation set. Results The tumor necrosis factor (TNF) signaling via NF-κB pathway was significantly augmented with the highest normalized enrichment score (NES). The genes in the brown module from WGCNA have shown a significant correlation with CAC (Pearson coefficient = 0.83, p = 6e-06). A subset of NF-κB related genes (FOS, CCL4, CXCL1, MYC, CEBPB, ATF3, and JUNB) were identified with a relatively higher expression level in CAC samples. The diagnostic value of this 7-gene biomarker was estimated by the receiver operating characteristic (ROC) curve with an area under the ROC curve (AUC) at 0.82 (p<0.0001, 95% CI: 0.7098-0.9400) in the validation cohort. Conclusion In summary, the increased expression of this seven-NF-κB-related gene signature may act as a powerful index for tumorigenesis prediction among patients with UC in remission.
Collapse
Affiliation(s)
- Chao-Yi Ge
- Department of Gastroenterology, Peking University First Hospital, Beijing, People's Republic of China
| | - Li-Yuan Wei
- Department of Breast Surgery, Shanxi Bethune Hospital, Taiyuan, People's Republic of China
| | - Yu Tian
- Department of Gastroenterology, Peking University First Hospital, Beijing, People's Republic of China
| | - Hua-Hong Wang
- Department of Gastroenterology, Peking University First Hospital, Beijing, People's Republic of China
| |
Collapse
|
4
|
Cheng YF, Wang XM, Yan M, Xiao JG. [Expression of the Fra-1 gene in the peripheral blood of children with Wilms tumor]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:161-164. [PMID: 30782279 PMCID: PMC7389833 DOI: 10.7499/j.issn.1008-8830.2019.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/07/2019] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To study the expression of the Fra-1 gene in the peripheral blood of children with Wilms tumor and its clinical significance. METHODS Fifty children pathologically diagnosed with Wilms tumor between December 2012 and January 2018 were enrolled as the case group, and 40 healthy children for physical examination were selected as the control group. Among the 45 children with Wilms tumor who were followed up, the children with continuous remission were included in the ideal efficacy group (n=33), and those with recurrence, metastasis or death were included in the poor efficacy group (n=12). Peripheral blood samples were collected from all subjects. Quantitative real-time PCR was used to measure the mRNA expression of Fra-1. RESULTS The case group had significantly higher mRNA expression of Fra-1 in peripheral blood than the control group (P<0.05). In the case group, Fra-1 mRNA expression was significantly different between the individuals with and without distant metastasis and those with different TNM stages (P<0.05), but was not significantly different between the individuals with different sexes, ages, tumor diabetes, tumor locations and alpha-fetoprotein levels (P>0.05). The mRNA expression of Fra-1 was significantly lower in the ideal efficacy group than in the poor efficacy group (P<0.05). CONCLUSIONS Fra-1 may be involved in the development of Wilms tumor and plays a certain role in its development, invasion and metastasis, but the mechanism remains to be further studied.
Collapse
Affiliation(s)
- Yong-Feng Cheng
- Department of Pediatrics, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China.
| | | | | | | |
Collapse
|
5
|
Sabzevary-Ghahfarokhi M, Shohan M, Shirzad H, Rahimian G, Bagheri N, Soltani A, Deris F, Ghatreh-Samani M, Razmara E. The expression analysis of Fra-1 gene and IL-11 protein in Iranian patients with ulcerative colitis. BMC Immunol 2018; 19:17. [PMID: 29914371 PMCID: PMC6006762 DOI: 10.1186/s12865-018-0257-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 06/11/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Fra-1 (fosl1) belongs to the activator protein1 (AP-1) family inducing IL-11 expression in oxidative stress condition. IL-11 plays a pivotal role in protecting epithelial barriers integrity. In this study, we investigated the Fra-1 gene expression in the inflamed mucosa of patients with ulcerative colitis (UC) as well as its relation to IL-11 expression. MATERIALS AND METHODS We enrolled 20 patients and 20 healthy controls with definite UC based on the clinical criteria. Fra-1 gene expression in inflamed and non-inflamed colonic biopsies was determined by real-time polymerase chain reaction (RT-PCR). The IL-11 protein concentration was measured by Enzyme-Linked Immunosorbent Assay (ELISA) method. Pearson correlation was applied to calculate the relation between Fra-1 and IL-11. RESULTS An increased level of Fra-1 gene expression was observed in patients with mild ulcerative colitis. The protein concentration of IL-11 was also increased in mild UC patients. Conversely, a significant decrease of IL-11 protein level was detected in severe UC patients compared to control group. CONCLUSION Oxidative stress in inflamed intestinal biopsies can induce fra-1 gene expression. Our findings suggest that Fra-1 transcription factor leads to the production of IL-11 protein in UC patients.
Collapse
Affiliation(s)
- Milad Sabzevary-Ghahfarokhi
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mojtaba Shohan
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hedayatollah Shirzad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Ghorbanali Rahimian
- Department of Internal Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nader Bagheri
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amin Soltani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Deris
- Department of Epidemiology and Biostatistics, School of Health, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahdi Ghatreh-Samani
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ehsan Razmara
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
6
|
Toosendanin inhibits adipogenesis by activating Wnt/β-catenin signaling. Sci Rep 2018; 8:4626. [PMID: 29545541 PMCID: PMC5854628 DOI: 10.1038/s41598-018-22873-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/02/2018] [Indexed: 12/15/2022] Open
Abstract
Toosendanin (TSN), a triterpenoid extracted from Melia toosendan, has been reported to possess anti-oxidant, anti-inflammatory, anti-allergic, and anti-arthritic activities. However, its anti-adipogenic effect remains unknown. Here, we found that TSN dose-dependently attenuated lipid accumulation in preadipocytes 3T3-L1 as evidenced by Oil Red O staining. TSN also significantly downregulated mRNA and protein levels of adipocytokines (adiponectin and leptin), CCAAT/enhancer binding proteins α (C/EBP-α), peroxisome proliferator-activated receptor γ (PPAR-γ), fatty acid synthase, and acetyl-CoA carboxylase in adipocytes. To understand the mechanism, we observed that TSN effectively activated Wnt/β-catenin pathway, in which TSN increased low density lipoprotein receptor related protein 6, disheveled 2, β-catenin, and cyclin D1 expression levels, while it inactivated glycogen synthase kinase 3β by enhancing its phosphorylation. Moreover, TSN reduced weight of gonadal white fat and serum triacylglycerol (TAG) content in high-fat diet (HFD)-fed mice. Interestingly, the in vivo studies also demonstrated that TSN promoted the expression of β-catenin, but accordingly repressed C/EBP-α and PPAR-γ expression in HFD-induced mice. Overall, TSN is capable of inhibiting the lipogenesis of adipocytes by activating the Wnt/β-catenin pathway, suggesting potential application of TSN as a natural anti-obesity agent.
Collapse
|
7
|
Yang Y, Huycke MM, Herman TS, Wang X. Glutathione S-transferase alpha 4 induction by activator protein 1 in colorectal cancer. Oncogene 2016; 35:5795-5806. [PMID: 27065323 DOI: 10.1038/onc.2016.113] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 12/25/2015] [Accepted: 01/04/2016] [Indexed: 01/01/2023]
Abstract
Glutathione S-transferase alpha 4 (GSTA4) is a phase II detoxifying enzyme that metabolizes electrophiles and carcinogens including 4-hydroxy-2-nonenal (4-HNE), an endogenous carcinogen that contributes to colorectal carcinogenesis. In this study, we investigated GSTA4 expression and regulation in murine primary colonic epithelial cells, microbiome-driven murine colitis and human carcinomas. Exposure of YAMC cells to 4-HNE induced Gsta4 expression. Using an inflammation-associated model of colorectal cancer (CRC), Gsta4 expression increased in vivo in colon macrophages and serum after 2 weeks of colonization of IL-10 deficient (Il10-/-) mice with Enterococcus faecalis. Increased expression was noted after 9 months of colonization in colon macrophages and epithelia in areas of inflammation. In human colon biopsies, immunohistochemistry showed no GSTA4 expression in normal epithelial cells, whereas GSTA4 was strongly expressed in the neoplastic epithelia of invasive carcinomas. For tubular adenomas, increased expression was primarily noted in stromal macrophages. Increased GSTA4 was confirmed in established human CRC cell lines and associated with 4-HNE-protein adducts in human colon adenomas and CRC. Next, we showed that 4-HNE induced activation of c-Jun and Nrf2, two components of the oncogenic transcription factor AP-1. AP-1 inhibitors and gene-specific small interfering RNAs partially suppressed GSTA4 expression. Co-immunoprecipitation confirmed interactions between c-Jun and Nrf2 supporting a role for AP-1 in regulating 4-HNE-induced GSTA4 expression. These findings demonstrate GSTA4 activation during 4-HNE-induced neoplastic transformation in colorectal carcinogenesis. GSTA4 is a potential surrogate biomarker for CRC screening and should provide novel approaches for chemoprevention.
Collapse
Affiliation(s)
- Y Yang
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,The Muchmore Laboratories for Infectious Diseases Research, Oklahoma City VA Health Care System, Oklahoma City, OK, USA
| | - M M Huycke
- The Muchmore Laboratories for Infectious Diseases Research, Oklahoma City VA Health Care System, Oklahoma City, OK, USA.,Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - T S Herman
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - X Wang
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,The Muchmore Laboratories for Infectious Diseases Research, Oklahoma City VA Health Care System, Oklahoma City, OK, USA
| |
Collapse
|
8
|
Zhang J, Yan B, Späth SS, Qun H, Cornelius S, Guan D, Shao J, Hagiwara K, Van Waes C, Chen Z, Su X, Bi Y. Integrated transcriptional profiling and genomic analyses reveal RPN2 and HMGB1 as promising biomarkers in colorectal cancer. Cell Biosci 2015; 5:53. [PMID: 26388988 PMCID: PMC4574027 DOI: 10.1186/s13578-015-0043-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/16/2015] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease that is associated with a gradual accumulation of genetic and epigenetic alterations. Among all CRC stages, stage II tumors are highly heterogeneous with a high relapse rate in about 20–25 % of stage II CRC patients following surgery. Thus, a comprehensive analysis of gene signatures to identify aggressive and metastatic phenotypes in stage II CRC is desired for a more accurate disease classification and outcome prediction. By utilizing a Cancer Array, containing 440 oncogenes and tumor suppressors to profile mRNA expression, we identified a larger number of differentially expressed genes in poorly differentiated stage II colorectal adenocarcinoma tissues, compared to their matched normal tissues. Ontology and Ingenuity Pathway Analysis (IPA) indicated that these genes are involved in functional mechanisms associated with several transcription factors. Genomic alterations of these genes were also investigated through The Cancer Genome Atlas (TCGA) database, utilizing 195 published CRC specimens. The percentage of genomic alterations in these genes was ranked based on their mRNA expression, copy number variations and mutations. This data was further combined with published microarray studies from a large set of CRC tumors classified based on prognostic features. This led to the identification of eight candidate genes including RPN2, HMGB1, AARS, IGFBP3, STAT1, HYOU1, NQO1 and PEA15 that were associated with the progressive phenotype. In particular, RPN2 and HMGB1 displayed a higher genomic alteration frequency in CRC, compared to eight other major solid cancers. Immunohistochemistry was performed on additional 78 stage I–IV CRC samples, where RPN2 protein immunostaining exhibited a significant association with stage III/IV tumors, distant metastasis, and poor differentiation, indicating that RPN2 expression is associated with poor prognosis. Further, our study revealed significant transcriptional regulatory mechanisms, networks and gene signatures, underlying CRC malignant progression and phenotype warranting future clinical investigations.
Collapse
Affiliation(s)
- Jialing Zhang
- School of Public Health, Wuhan University, Wuhan, China ; Clinical Medicine Research Center, The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, China ; Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Bin Yan
- Laboratory for Food Safety and Environmental Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Stephan Stanislaw Späth
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Hu Qun
- Department of Oncology, The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, China
| | - Shaleeka Cornelius
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Daogang Guan
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Jiaofang Shao
- Department of Bioinformatics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Koichi Hagiwara
- Department of Respiratory Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Xiulan Su
- Clinical Medicine Research Center, The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, China
| | - Yongyi Bi
- School of Public Health, Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Camp JG, Frank CL, Lickwar CR, Guturu H, Rube T, Wenger AM, Chen J, Bejerano G, Crawford GE, Rawls JF. Microbiota modulate transcription in the intestinal epithelium without remodeling the accessible chromatin landscape. Genome Res 2014; 24:1504-16. [PMID: 24963153 PMCID: PMC4158762 DOI: 10.1101/gr.165845.113] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Microbiota regulate intestinal physiology by modifying host gene expression along the length of the intestine, but the underlying regulatory mechanisms remain unresolved. Transcriptional specificity occurs through interactions between transcription factors (TFs) and cis-regulatory regions (CRRs) characterized by nucleosome-depleted accessible chromatin. We profiled transcriptome and accessible chromatin landscapes in intestinal epithelial cells (IECs) from mice reared in the presence or absence of microbiota. We show that regional differences in gene transcription along the intestinal tract were accompanied by major alterations in chromatin accessibility. Surprisingly, we discovered that microbiota modify host gene transcription in IECs without significantly impacting the accessible chromatin landscape. Instead, microbiota regulation of host gene transcription might be achieved by differential expression of specific TFs and enrichment of their binding sites in nucleosome-depleted CRRs near target genes. Our results suggest that the chromatin landscape in IECs is preprogrammed by the host in a region-specific manner to permit responses to microbiota through binding of open CRRs by specific TFs.
Collapse
Affiliation(s)
- J Gray Camp
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Department of Developmental Biology, Stanford University, Stanford, California 94305, USA; Computer Science Department, Stanford University, Stanford, California 94305, USA
| | - Christopher L Frank
- Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina 27708, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina 27710, USA
| | - Colin R Lickwar
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina 27710, USA
| | - Harendra Guturu
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, USA
| | - Tomas Rube
- Physics Department, Stanford University, Stanford, California 94305, USA
| | - Aaron M Wenger
- Computer Science Department, Stanford University, Stanford, California 94305, USA
| | - Jenny Chen
- Biomedical Informatics Program, Stanford University, Stanford, California 94305, USA
| | - Gill Bejerano
- Department of Developmental Biology, Stanford University, Stanford, California 94305, USA; Computer Science Department, Stanford University, Stanford, California 94305, USA
| | - Gregory E Crawford
- Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina 27708, USA; Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, North Carolina 27708, USA
| | - John F Rawls
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina 27708, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina 27710, USA;
| |
Collapse
|
10
|
Colon Macrophages Polarized by Commensal Bacteria Cause Colitis and Cancer through the Bystander Effect. Transl Oncol 2013; 6:596-606. [PMID: 24151540 DOI: 10.1593/tlo.13412] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 05/20/2013] [Accepted: 06/24/2013] [Indexed: 02/07/2023] Open
Abstract
Intestinal commensal bacteria have recently been shown to trigger macrophages to produce diffusible clastogens (or chromosome-breaking factors) through a bystander effect (BSE) that mediates DNA damage and induces chromosomal instability in neighboring cells. Colon macrophages appear central to colon carcinogenesis and BSE through the expression of tumor necrosis factor-α (TNF-α) and cyclooxygenase-2 (COX-2). The former induces netrin-1, a regulator of intestinal epithelial cell apoptosis, and the latter generates trans-4-hydroxy-2-nonenal (4-HNE), an endogenous mutagen. To test whether colon macrophages are key effectors for BSE, we depleted these cells in interleukin-10 knockout mice colonized with Enterococcus faecalis using encapsulated liposomal clodronate (ELC), a bisphosphonate that causes macrophage apoptosis. We observed that E. faecalis polarizes colon macrophages to an M1 phenotype. In addition, depleting these cells suppressed COX-2 and TNF-α, blocked the formation of 4-HNE protein adducts, and inhibited up-regulation of netrin-1-all markers for BSE. Finally, treatment with ELC prevented colitis, β-catenin activation, and cancer formation. These results show that selected human commensals can polarize colon macrophages to the M1 phenotype and, when activated, serve as the key effector for bacterial-induced BSE. Our findings suggest that depleting M1-polarized macro-phages is a mechanism for the chemopreventive activity of bisphosphonates and that it represents a new strategy for preventing colon cancer induced by intestinal commensals.
Collapse
|
11
|
Zeller E, Hammer K, Kirschnick M, Braeuning A. Mechanisms of RAS/β-catenin interactions. Arch Toxicol 2013; 87:611-32. [PMID: 23483189 DOI: 10.1007/s00204-013-1035-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 02/28/2013] [Indexed: 12/20/2022]
Abstract
Signaling through the WNT/β-catenin and the RAS (rat sarcoma)/MAPK (mitogen-activated protein kinase) pathways plays a key role in the regulation of various physiological cellular processes including proliferation, differentiation, and cell death. Aberrant mutational activation of these signaling pathways is closely linked to the development of cancer in many organs, in humans as well as in laboratory animals. Over the past years, more and more evidence for a close linkage of the two oncogenic signaling cascades has accumulated. Using different experimental approaches, model systems, and experimental conditions, a variety of molecular mechanisms have been identified by which signal transduction through WNT/β-catenin and RAS interact, either in a synergistic or an antagonistic manner. Mechanisms of interaction comprise an upstream crosstalk at the level of pathway-activating ligands and their receptors, interrelations of cytosolic kinases involved in either pathways, as well as interaction in the nucleus related to the joint regulation of target gene transcription. Here, we present a comprehensive review of the current knowledge on the interaction of RAS/MAPK- and WNT/β-catenin-driven signal transduction in mammalian cells.
Collapse
Affiliation(s)
- Eva Zeller
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, University of Tübingen, Germany
| | | | | | | |
Collapse
|
12
|
Trierweiler C, Blum HE, Hasselblatt P. The transcription factor c-Jun protects against liver damage following activated β-Catenin signaling. PLoS One 2012; 7:e40638. [PMID: 22792392 PMCID: PMC3391288 DOI: 10.1371/journal.pone.0040638] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 06/11/2012] [Indexed: 01/20/2023] Open
Abstract
Background The Wnt/β-Catenin signaling pathway is central for liver functions and frequently deregulated in hepatocellular carcinoma (HCC). Analysis of the early phenotypes and molecular events following β-Catenin activation is therefore essential for better understanding HCC pathogenesis. The AP-1 transcription factor c-Jun is a putative β-Catenin target gene and promotes hepatocyte survival, proliferation, and liver tumorigenesis, suggesting that c-Jun may be a key target of β-Catenin signaling in the liver. Methodology/Principal Findings To address this issue, the immediate hepatic phenotypes following deletion of the tumor suppressor Apc and subsequent β-Catenin activation were analyzed in mice. The contribution of c-Jun to these phenotypes was dissected in double mutant animals lacking both, Apc and c-Jun. β-Catenin was rapidly activated in virtually all Apc mutant hepatocytes while c-Jun was induced only after several days, suggesting that its expression was rather a secondary event following Apc deletion in the liver. Loss of Apc resulted in increased hepatocyte proliferation, hepatomegaly, deregulated protein metabolism, and premature death. Interestingly, additional deletion of c-Jun did not affect hepatocyte proliferation but resulted in increased liver damage and mortality. This phenotype correlated with impaired expression of hepatoprotective genes such as Birc5, Egfr Igf1 and subsequently deregulated Akt signaling. Conclusions/Significance These data indicate that c-Jun is not a primary target of β-Catenin signaling in the liver, but rather protects against liver damage, which in turn may promote liver tumorigenesis.
Collapse
Affiliation(s)
- Claudia Trierweiler
- Department of Medicine II, University Hospital Freiburg, Freiburg, Germany
- Faculty of Biology, Freiburg University, Freiburg, Germany
| | - Hubert E. Blum
- Department of Medicine II, University Hospital Freiburg, Freiburg, Germany
| | - Peter Hasselblatt
- Department of Medicine II, University Hospital Freiburg, Freiburg, Germany
- * E-mail:
| |
Collapse
|
13
|
Takeda K, Naguro I, Nishitoh H, Matsuzawa A, Ichijo H. Apoptosis signaling kinases: from stress response to health outcomes. Antioxid Redox Signal 2011; 15:719-61. [PMID: 20969480 DOI: 10.1089/ars.2010.3392] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Apoptosis is a highly regulated process essential for the development and homeostasis of multicellular organisms. Whereas caspases, a large family of intracellular cysteine proteases, play central roles in the execution of apoptosis, other proapoptotic and antiapoptotic regulators such as the members of the Bcl-2 family are also critically involved in the regulation of apoptosis. A large body of evidence has revealed that a number of protein kinases are among such regulators and regulate cellular sensitivity to various proapoptotic signals at multiple steps in apoptosis. However, recent progress in the analysis of these apoptosis signaling kinases demonstrates that they generally act as crucial regulators of diverse cellular responses to a wide variety of stressors, beyond their roles in apoptosis regulation. In this review, we have cataloged apoptosis signaling kinases involved in cellular stress responses on the basis of their ability to induce apoptosis and discuss their roles in stress responses with particular emphasis on health outcomes upon their dysregulation.
Collapse
Affiliation(s)
- Kohsuke Takeda
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, Strategic Approach to Drug Discovery and Development in Pharmaceutical Sciences, Global Center of Excellence Program and Core Research for Evolutional Science and Technology, Japan Science and Technology Corporation, The University of Tokyo, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
14
|
Warfel NA, Niederst M, Stevens MW, Brennan PM, Frame MC, Newton AC. Mislocalization of the E3 ligase, β-transducin repeat-containing protein 1 (β-TrCP1), in glioblastoma uncouples negative feedback between the pleckstrin homology domain leucine-rich repeat protein phosphatase 1 (PHLPP1) and Akt. J Biol Chem 2011; 286:19777-88. [PMID: 21454620 PMCID: PMC3103356 DOI: 10.1074/jbc.m111.237081] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 03/25/2011] [Indexed: 12/16/2022] Open
Abstract
The PH domain leucine-rich repeat protein phosphatase, PHLPP, plays a central role in controlling the amplitude of growth factor signaling by directly dephosphorylating and thereby inactivating Akt. The cellular levels of PHLPP1 have recently been shown to be enhanced by its substrate, activated Akt, via modulation of a phosphodegron recognized by the E3 ligase β-TrCP1, thus providing a negative feedback loop to tightly control cellular Akt output. Here we show that this feedback loop is lost in aggressive glioblastoma but not less aggressive astrocytoma. Overexpression and pharmacological studies reveal that loss of the feedback loop does not result from a defect in PHLPP1 protein or in the upstream kinases that control its phosphodegron. Rather, the defect arises from altered localization of β-TrCP1; in astrocytoma cell lines and in normal brain tissue the E3 ligase is predominantly cytoplasmic, whereas in glioblastoma cell lines and patient-derived tumor neurospheres, the E3 ligase is confined to the nucleus and thus spatially separated from PHLPP1, which is cytoplasmic. Restoring the localization of β-TrCP1 to the cytosol of glioblastoma cells rescues the ability of Akt to regulate PHLPP1 stability. Additionally, we show that the degradation of another β-TrCP1 substrate, β-catenin, is impaired and accumulates in the cytosol of glioblastoma cell lines. Our findings reveal that the cellular localization of β-TrCP1 is altered in glioblastoma, resulting in dysregulation of PHLPP1 and other substrates such as β-catenin.
Collapse
Affiliation(s)
- Noel A. Warfel
- From the Department of Pharmacology and
- Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California 92093 and
| | - Matt Niederst
- From the Department of Pharmacology and
- Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California 92093 and
| | | | - Paul M. Brennan
- the Edinburgh Cancer Research Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Margaret C. Frame
- the Edinburgh Cancer Research Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | | |
Collapse
|
15
|
Fujishita T, Aoki M, Taketo MM. JNK signaling promotes intestinal tumorigenesis through activation of mTOR complex 1 in Apc(Δ716) mice. Gastroenterology 2011; 140:1556-63.e6. [PMID: 21320501 DOI: 10.1053/j.gastro.2011.02.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Revised: 01/11/2011] [Accepted: 02/01/2011] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Signaling by the mammalian target of rapamycin complex 1 (mTORC1) has been implicated in various human cancers. mTORC1 signaling is activated in intestinal tumors of adenomatous polyposis coli (Apc(Δ716)) mice, a model of familial adenomatous polyposis; in these mice, the mTORC1 inhibitor RAD001 can block tumor formation. However, the precise mechanism of mTORC1 signaling in intestinal tumors is not clear. We investigated whether c-Jun-NH(2) terminal kinase (JNK) is involved in the mTORC1 activation. METHODS We investigated the effects of an inhibitor and an activator of JNK, as well as small interfering RNA against JNK, on mTORC1 in Apc(Δ716) mice and colon cancer cell lines. We also determined the role of JNK in mTORC1 signaling using in vitro kinase assays. RESULTS JNK was activated in intestinal polyps of Apc((Δ716) mice); the JNK inhibitor SP600125 significantly suppressed tumor formation. In colorectal cancer cell lines, the JNK activator anisomycin activated mTORC1, whereas SP600125 or small interfering RNAs against JNK suppressed signaling. Importantly, JNK stimulated the mTORC1 kinase activity in vitro, through direct phosphorylation of Raptor at serine 863. CONCLUSIONS JNK is required for activation of mTORC1 in intestinal tumor cells. JNK inhibitors might be developed as therapeutics or to prevent development of intestinal tumors.
Collapse
Affiliation(s)
- Teruaki Fujishita
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Yoshida-Konoé-Cho, Sakyo-Ku, Kyoto, Japan
| | | | | |
Collapse
|
16
|
Lee S, Song YA, Park YL, Cho SB, Lee WS, Lee JH, Chung IJ, Kim KK, Rew JS, Joo YE. Expression of KITENIN in human colorectal cancer and its relation to tumor behavior and progression. Pathol Int 2011; 61:210-20. [DOI: 10.1111/j.1440-1827.2011.02646.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
17
|
Talero E, Sánchez-Fidalgo S, Villegas I, de la Lastra CA, Illanes M, Motilva V. Role of different inflammatory and tumor biomarkers in the development of ulcerative colitis-associated carcinogenesis. Inflamm Bowel Dis 2011; 17:696-710. [PMID: 20722052 DOI: 10.1002/ibd.21420] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 06/08/2010] [Indexed: 12/21/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is the most severe complication in inflammatory bowel disease (IBD). In the present study we investigated different mechanistic links between chronic colonic inflammation and its progression to adenocarcinoma. Along these lines, given that adrenomedullin (AM) has been implicated in carcinogenesis, we also analyzed changes in its colonic expression. METHODS Mice were exposed to 5, 10, and 15 cycles of dextran sulfate sodium (DSS); each cycle consisted of 0.7% DSS for 1 week followed by distilled water for 10 days. After each period, macroscopic and histological studies, as well as characterization of inflammatory and tumor biomarkers, were carried out. RESULTS The disease activity index (DAI) showed that the disease was present from the third cycle and it gradually increased during the course of DSS treatment. Macroscopic tumors were only seen after 15 cycles, and microscopic study showed that inflammation, dysplasia, and adenocarcinomas correlated with DSS cycles. β-Catenin and proliferating cell nuclear antigen expressions progressively increased in animals treated with the different cycles of DSS. TNF-α and IFN-γ showed the highest production at the tenth cycle. COX-2, mPGES-1, and iNOS levels were also appreciably higher at the fifth and tenth cycles. Moreover, we observed a progressive enhancement in AM expression and changes in its intracellular location during the progression of the disease. CONCLUSIONS Our results show an early induction of proinflammatory factors, which may contribute to the development of colon cancer, as well as demonstrate, for the first time, the expression of AM in IBD-derived CRC.
Collapse
Affiliation(s)
- E Talero
- Department of Pharmacology, School of Pharmacy, University of Seville, Seville, Spain.
| | | | | | | | | | | |
Collapse
|
18
|
Bottomly D, Kyler SL, McWeeney SK, Yochum GS. Identification of {beta}-catenin binding regions in colon cancer cells using ChIP-Seq. Nucleic Acids Res 2010; 38:5735-45. [PMID: 20460455 PMCID: PMC2943592 DOI: 10.1093/nar/gkq363] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Deregulation of the Wnt/β-catenin signaling pathway is a hallmark of colon cancer. Mutations in the adenomatous polyposis coli (APC) gene occur in the vast majority of colorectal cancers and are an initiating event in cellular transformation. Cells harboring mutant APC contain elevated levels of the β-catenin transcription coactivator in the nucleus which leads to abnormal expression of genes controlled by β-catenin/T-cell factor 4 (TCF4) complexes. Here, we use chromatin immunoprecipitation coupled with massively parallel sequencing (ChIP-Seq) to identify β-catenin binding regions in HCT116 human colon cancer cells. We localized 2168 β-catenin enriched regions using a concordance approach for integrating the output from multiple peak alignment algorithms. Motif discovery algorithms found a core TCF4 motif (T/A–T/A–C–A–A–A–G), an extended TCF4 motif (A/T/G–C/G–T/A–T/A–C–A–A–A–G) and an AP-1 motif (T–G–A–C/T–T–C–A) to be significantly represented in β-catenin enriched regions. Furthermore, 417 regions contained both TCF4 and AP-1 motifs. Genes associated with TCF4 and AP-1 motifs bound β-catenin, TCF4 and c-Jun in vivo and were activated by Wnt signaling and serum growth factors. Our work provides evidence that Wnt/β-catenin and mitogen signaling pathways intersect directly to regulate a defined set of target genes.
Collapse
Affiliation(s)
- Daniel Bottomly
- Oregon Clinical and Translational Research Institute, Oregon Health and Science University, Portland, OR, USA
| | | | | | | |
Collapse
|
19
|
Kress E, Skah S, Sirakov M, Nadjar J, Gadot N, Scoazec JY, Samarut J, Plateroti M. Cooperation between the thyroid hormone receptor TRalpha1 and the WNT pathway in the induction of intestinal tumorigenesis. Gastroenterology 2010; 138:1863-74. [PMID: 20114049 DOI: 10.1053/j.gastro.2010.01.041] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 12/30/2009] [Accepted: 01/21/2010] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Colorectal tumorigenesis is a multistep process involving the alteration of oncogenes and tumor suppressor genes, leading to the deregulation of molecular pathways that govern intestinal homeostasis. We have previously shown that the thyroid hormone receptor alpha1 (TRalpha1) controls intestinal development and homeostasis through the WNT pathway. More precisely, TRalpha1 directly enhances the transcription of several components of this pathway, allowing increased expression of beta-catenin/Tcf4 target genes and stimulation of cell proliferation. Because the WNT pathway is a major player in controlling intestinal homeostasis, we addressed whether the TRalpha1 receptor has tumor-inducing potential. METHODS We generated mice overexpressing TRalpha1 specifically in the intestinal epithelium in a wild-type (vil-TRalpha1) or a WNT-activated (vil-TRalpha1/Apc(+/1638N)) genetic background. RESULTS The intestine of vil-TRalpha1 mice presents aberrant intestinal mucosal architecture and increased cell proliferation and develops adenoma at a low rate. However, TRalpha1 overexpression is unable to induce cancer development. On the contrary, we observed accelerated tumorigenesis in vil-TRalpha1/Apc(+/1638N) mice compared with the Apc(+/1638N) mutants. CONCLUSION Our results suggest that this phenotype is due to cooperation between the activated TRalpha1 and WNT pathways. This is the first report describing the tumor-inducing function of TRalpha1 in the intestine.
Collapse
Affiliation(s)
- Elsa Kress
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Newton VA, Ramocki NM, Scull BP, Simmons JG, McNaughton K, Lund PK. Suppressor of cytokine signaling-2 gene disruption promotes Apc(Min/+) tumorigenesis and activator protein-1 activation. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2320-32. [PMID: 20348236 DOI: 10.2353/ajpath.2010.090684] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Epigenetic in vitro and in vivo studies suggest that suppressor of cytokine signaling-2 (SOCS2) may normally limit tumorigenesis in the intestine; however, this theory has not been directly tested. We hypothesized that SOCS2 deficiency promotes spontaneous intestinal tumorigenesis in Apc(Min/+) mice. Therefore, we quantified tumor number, size, and load in the small intestine and colon using SOCS2(+/+)/Apc(Min/+), SOCS2(+/-)/Apc(Min/+), and SOCS2(-/-)/Apc(Min/+) mice and assayed hematocrit as an indirect marker of disease severity. Biochemical and histological assays were used to assess mechanisms. Heterozygous and homozygous disruption of SOCS2 alleles promoted 166 and 441% increases in tumor load in the small intestine, respectively, accelerated development of colon tumors, and caused severe anemia. SOCS2 deletion promoted significant increases in intestinal insulin-like growth factor-I mRNA but did not affect plasma insulin-like growth factor-I. Western blots and immunohistochemical analysis demonstrated that tumor and nontumor intestinal tissue of SOCS2(-/-)/Apc(Min/+) mice had increased serine 727 phosphorylation of signal transducer and activator of transcription 3 compared with SOCS2(+/+)/Apc(Min/+) mice. Moreover, electromobility shift assays showed that SOCS2 deletion did not alter signal transducer and activator of transcription 3 DNA binding. However, tumors and small intestine from SOCS2(-/-)/Apc(Min/+) showed dramatic increases in activator protein-1 (AP-1) DNA binding, and SOCS2 overexpression in vitro reduced levels of AP-1. These studies indicate that SOCS2 deletion promotes the spontaneous development of intestinal tumors driven by mutations in the adenomatous polyposis coli/beta-catenin pathway and activates AP-1. Therefore, reduced expression or epigenetic silencing of SOCS2 may serve as a useful biomarker for colorectal cancer risk.
Collapse
Affiliation(s)
- Victoria A Newton
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599-7545, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Shaulian E. AP-1--The Jun proteins: Oncogenes or tumor suppressors in disguise? Cell Signal 2010; 22:894-9. [PMID: 20060892 DOI: 10.1016/j.cellsig.2009.12.008] [Citation(s) in RCA: 516] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 12/31/2009] [Indexed: 10/20/2022]
Abstract
Since its discovery more than two decades ago the involvement of the Activating protein 1 (AP-1) in proliferation, inflammation, differentiation, apoptosis, cellular migration and wound healing has been intensively studied. A model based on the early studies suggested antagonistic roles for the Jun proteins in proliferation and transformation. c-Jun was suggested to enhance transformation whereas JunB suggested to inhibit it in an antagonistic manner. Surprisingly, despite accumulation of data obtained from animal models regarding the role of Jun proteins in cancer and identification of oncogenic pathways regulating them, their involvement in human cancer was not demonstrated until recently. Here, we will describe the current knowledge about the roles of Jun proteins in human neoplasia. We will focus on the pathological examples demonstrating that the initial dogma has to be reexamined. For example, like c-Jun, JunB seems to play an oncogenic role in lymphomas, particularly in Hodgkin's lympomas. Furthermore, unlike the antagonistic activities of c-Jun and JunB in the transcription of genes coding for major cell cycle regulators such as CyclinD or p16INK4A, the transcription of other cell cycle regulating genes is modified similarly by c-Jun or JunB. Interestingly, some of these genes such as the ones coding for CyclinA or p19(ARF) are important players in either positive or negative regulation of cellular proliferation and survival. Finally, we will also discuss results posing JNK, known so far as the major activator of c-Jun, as a negative regulator of c-Jun level and activity. These recent findings suggest that the role of each Jun protein in neoplasia as well as in cellular survival should be examined in a context-dependent manner.
Collapse
Affiliation(s)
- Eitan Shaulian
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research-Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
22
|
A beta-catenin/TCF-coordinated chromatin loop at MYC integrates 5' and 3' Wnt responsive enhancers. Proc Natl Acad Sci U S A 2009; 107:145-50. [PMID: 19966299 DOI: 10.1073/pnas.0912294107] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aberrant MYC gene expression by the Wnt/beta-catenin pathway is implicated in colorectal carcinogenesis. Wnt/beta-catenin signaling stimulates association of the beta-catenin coactivator complex with two Wnt responsive enhancers (WREs) located in close proximity to MYC gene boundaries. Each enhancer directly binds members of the TCF/Lef family of transcription factors that, in turn, recruit beta-catenin. In a previous report, we showed that the downstream MYC enhancer (MYC 3' WRE) cooperated with the upstream enhancer (MYC 5' WRE) to activate expression of a heterologous reporter gene in response to Wnt/beta-catenin and mitogen signaling. Here we use chromatin conformation capture (3C) to show that the MYC 5' and 3' WREs are juxtaposed at the genomic MYC locus during active transcription. This MYC 5'3' chromatin loop is present in HCT116 human colorectal cancer cells that contain high levels of nuclear beta-catenin and is absent in HEK293 cells that contain trace amounts of nuclear beta-catenin. Depletion of functional beta-catenin/TCF complexes blocks formation of the MYC 5'3 chromatin loop. Furthermore, we find that the chromatin loop is absent in quiescent cells, but is rapidly and transiently induced by serum mitogens in a beta-catenin-dependent manner. Thus, we propose that a distinct chromatin architecture coordinated by beta-catenin/TCF-bound WREs accompanies transcriptional activation of MYC gene expression.
Collapse
|
23
|
Abstract
Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) family members function in a cell context-specific and cell type-specific manner to integrate signals that affect proliferation, differentiation, survival and migration. Consistent with the importance of these events in tumorigenesis, JNK and p38 MAPK signalling is associated with cancers in humans and mice. Studies in mouse models have been essential to better understand how these MAPKs control cancer development, and these models are expected to provide new strategies for the design of improved therapeutic approaches. In this Review we highlight the recent progress made in defining the functions of the JNK and p38 MAPK pathways in different cancers.
Collapse
|
24
|
Abstract
Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) family members function in a cell context-specific and cell type-specific manner to integrate signals that affect proliferation, differentiation, survival and migration. Consistent with the importance of these events in tumorigenesis, JNK and p38 MAPK signalling is associated with cancers in humans and mice. Studies in mouse models have been essential to better understand how these MAPKs control cancer development, and these models are expected to provide new strategies for the design of improved therapeutic approaches. In this Review we highlight the recent progress made in defining the functions of the JNK and p38 MAPK pathways in different cancers.
Collapse
Affiliation(s)
- Erwin F Wagner
- Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro 3, Madrid 28029, Spain.
| | | |
Collapse
|
25
|
Sancho R, Nateri AS, de Vinuesa AG, Aguilera C, Nye E, Spencer-Dene B, Behrens A. JNK signalling modulates intestinal homeostasis and tumourigenesis in mice. EMBO J 2009; 28:1843-54. [PMID: 19521338 DOI: 10.1038/emboj.2009.153] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Accepted: 05/12/2009] [Indexed: 01/08/2023] Open
Abstract
Wnt signalling is a crucial signalling pathway controlling intestinal homeostasis and cancer. We show here that the JNK MAP kinase pathway and one of its most important substrates, the AP-1 transcription factor c-Jun, modulates Wnt signalling strength in the intestine. Transgenic gut-specific augmentation of JNK signalling stimulated progenitor cell proliferation and migration, resulting in increased villus length. In the crypt, c-Jun protein was highly expressed in progenitor cells and the absence of c-Jun resulted in decreased proliferation and villus length. In addition to several known c-Jun/AP-1 target genes, expression of Wnt target genes Axin2 and Lgr5 were stimulated by JNK activation, suggesting a cross talk of JNK to Wnt signalling. Expression of the Wnt pathway component TCF4 was controlled by JNK activity, and chromatin immunoprecipitation and reporter assays identified tcf4 as a direct c-Jun target gene. Consequently, increased JNK activity accelerated tumourigenesis in a model of colorectal carcinogenesis. As c-jun is a direct target of the TCF4/beta-catenin complex, the control of tcf4 expression by JNK/c-Jun leads to a positive feedback loop that connects JNK and Wnt signalling. This mechanism regulates the physiological function of progenitor cells and oncogenic transformation.
Collapse
Affiliation(s)
- Rocio Sancho
- Mammalian Genetics Laboratory, CRUK London Research Institute, Lincoln's Inn Fields Laboratories, London, UK
| | | | | | | | | | | | | |
Collapse
|
26
|
Hui L, Zatloukal K, Scheuch H, Stepniak E, Wagner EF. Proliferation of human HCC cells and chemically induced mouse liver cancers requires JNK1-dependent p21 downregulation. J Clin Invest 2008; 118:3943-53. [PMID: 19033664 PMCID: PMC2579707 DOI: 10.1172/jci37156] [Citation(s) in RCA: 254] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 10/01/2008] [Indexed: 12/11/2022] Open
Abstract
JNK proteins have been shown to be involved in liver carcinogenesis in mice, but the extent of their involvement in the development of human liver cancers is unknown. Here, we show that activation of JNK1 but not JNK2 was increased in human primary hepatocellular carcinomas (HCCs). Further, JNK1 was required for human HCC cell proliferation in vitro and tumorigenesis after xenotransplantation. Importantly, mice lacking JNK1 displayed decreased tumor cell proliferation in a mouse model of liver carcinogenesis and decreased hepatocyte proliferation in a mouse model of liver regeneration. In both cases, impaired proliferation was caused by increased expression of p21, a cell-cycle inhibitor, and reduced expression of c-Myc, a negative regulator of p21. Genetic inactivation of p21 in JNK1-/- mice restored hepatocyte proliferation in models of both liver carcinogenesis and liver regeneration, and overexpression of c-Myc increased proliferation of JNK1-/- liver cells. Similarly, JNK1 was found to control the proliferation of human HCC cells by affecting p21 and c-Myc expression. Pharmacologic inhibition of JNK reduced the growth of both xenografted human HCC cells and chemically induced mouse liver cancers. These findings provide a mechanistic link between JNK activity and liver cell proliferation via p21 and c-Myc and suggest JNK targeting can be considered as a new therapeutic approach for HCC treatment.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/therapy
- Cell Line, Tumor
- Cell Proliferation
- Cyclin-Dependent Kinase Inhibitor p21/genetics
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
- Female
- Hepatocytes/metabolism
- Humans
- Liver Neoplasms/chemically induced
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/therapy
- Liver Regeneration/drug effects
- Liver Regeneration/genetics
- Male
- Mice
- Mice, Knockout
- Mice, Nude
- Mitogen-Activated Protein Kinase 8/genetics
- Mitogen-Activated Protein Kinase 8/metabolism
- Neoplasms, Experimental/chemically induced
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/therapy
Collapse
Affiliation(s)
- Lijian Hui
- Research Institute of Molecular Pathology, Vienna, Austria.
Institute of Pathology, Medical University of Graz, Graz, Austria.
Spanish National Cancer Research Center, Madrid, Spain
| | - Kurt Zatloukal
- Research Institute of Molecular Pathology, Vienna, Austria.
Institute of Pathology, Medical University of Graz, Graz, Austria.
Spanish National Cancer Research Center, Madrid, Spain
| | - Harald Scheuch
- Research Institute of Molecular Pathology, Vienna, Austria.
Institute of Pathology, Medical University of Graz, Graz, Austria.
Spanish National Cancer Research Center, Madrid, Spain
| | - Ewa Stepniak
- Research Institute of Molecular Pathology, Vienna, Austria.
Institute of Pathology, Medical University of Graz, Graz, Austria.
Spanish National Cancer Research Center, Madrid, Spain
| | - Erwin F. Wagner
- Research Institute of Molecular Pathology, Vienna, Austria.
Institute of Pathology, Medical University of Graz, Graz, Austria.
Spanish National Cancer Research Center, Madrid, Spain
| |
Collapse
|