1
|
Franz C, Jötten L, Wührl M, Hartmann S, Klupp F, Schmidt T, Schneider M. Protective effect of miR-18a in resected liver metastases of colorectal cancer and FOLFOX treatment. Cancer Rep (Hoboken) 2023; 6:e1899. [PMID: 37698257 PMCID: PMC10728504 DOI: 10.1002/cnr2.1899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/22/2023] [Accepted: 08/27/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Colorectal cancer ranks second in terms of cancer associated deaths worldwide, whereas miRNA play a pivotal role in the etiology of cancer and its metastases. AIMS Studying the expression and cellular function of miR-18a in metastatic colorectal cancer and association to progression-free survival. METHODS AND RESULTS Colorectal liver metastases (N = 123) and primary colorectal cancer (N = 27) where analyzed by RT-PCR and correlated with clinical follow up data. Invasion and migration assays were performed with the liver metastatic cell line LIM2099 after miR-18a knockdown. Cell viability under FOLFOX treatment and knockdown was measured. We found that the expression of miR-18a was increased 4.38-fold in liver metastases and 3.86-fold in colorectal tumor tissue compared to healthy liver tissue and colorectal mucosa, respectively (p ≤ .001). Patients with a high miR-18a expression in liver metastases had a progression-free survival (PFS) of 13.6 months versus 8.9 months in patients with low expression (N = 123; p = .024). In vitro migration of LIM2099 cells was reduced after miR-18a knockdown and cell viability was significantly increased after miR-18a knockdown and treatment with folinic acid or oxaliplatin. Subgroup analysis of PFS revealed significant benefits for patients with high miR-18a expression receiving 5-FU, folinic acid or oxaliplatin. CONCLUSIONS High expression of miR-18a in colorectal liver metastases might have a protective effect after resection of metastases and FOLFOX treatment regarding PFS.
Collapse
Affiliation(s)
- Clemens Franz
- Department of General, Visceral and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Laila Jötten
- Department of General, Visceral and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Wührl
- Department of General, Visceral and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Sibylle Hartmann
- Department of General, Visceral and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Fee Klupp
- Department of General, Visceral and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
2
|
Manohar SM, Joshi KS. Molecular Pharmacology of Multitarget Cyclin-Dependent Kinase Inhibitors in Human Colorectal Carcinoma Cells. Expert Opin Ther Targets 2023; 27:251-261. [PMID: 37015886 DOI: 10.1080/14728222.2023.2199924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer death. Certain signaling pathways are implicated in colorectal carcinogenesis. Cyclin-dependent kinases (CDKs) are commonly hyperactivated in CRC and hence multitarget CDK inhibitors serve as promising therapeutic drugs against CRC. OBJECTIVE Off-target effects of multitarget CDK inhibitors with differential CDK inhibitory spectrum viz. P276-00 (also known as riviciclib), roscovitine and UCN-01 on CRC cell lines of varied genetic background were delineated. METHOD Protein expression was analyzed for key signaling proteins by western blotting. β-catenin localization was assessed using immunofluorescence. HIF-1 transcriptional activity and target gene expression were studied by reporter gene assay and RT-PCR respectively. Anti-migratory and anti-angiogenic potential was evaluated by wound healing assay and endothelial tube formation assay. RESULTS CDK inhibitors modulated various signaling pathways in CRC and for certain proteins showed a highly cell line-dependent response. Riviciclib and roscovitine inhibited HIF-1 transcriptional activity and HIF-1α accumulation in hypoxic HCT116 cells. Both of these drugs also abrogated migration of HCT116 and in vitro angiogenesis in HUVECs. CONCLUSION Anticancer activity of multitarget CDK inhibitors can be certainly attributed to their off-target effects and should be analyzed while assessing their therapeutic utility against CRC.
Collapse
Affiliation(s)
- Sonal M Manohar
- Department of Biological Sciences, Sunandan Divatia of School of Science, NMIMS (Deemed-to-be) University, Vile Parle (West), Mumbai, India
| | - Kalpana S Joshi
- Discovery Engine, Cipla R and D, Cipla Ltd. Vikhroli (West), Mumbai, India
| |
Collapse
|
3
|
Corona SP, Walker F, Weinstock J, Lessene G, Faux M, Burgess AW. Dual drug targeting to kill colon cancers. Cancer Med 2022; 11:2612-2626. [PMID: 35301819 PMCID: PMC9249985 DOI: 10.1002/cam4.4641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/06/2022] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Colorectal cancer (CRC) is driven by a small set of oncogenic and tumour suppressor mutations. However, different combinations of mutations often lead to poor tumour responses to individual anticancer drugs. We have investigated the antiproliferative and in vitro cytotoxic activity of pair‐wise combinations of inhibitors which target specific signalling pathways in colon cancer cells. Objectives To target specific signaling pathways pairwise with inhibitors in order to kill colon cancer cells. Methods The effects of different concentrations of two inhibitors on the proliferation and viability of colon cancer cell lines were measured using cell titre glow and cytotoxic assays in 2D and 3D cell micro‐cultures. One successful drug combination was used to treat a colon cancer cell line growing as a xenograft in nude mice. Results Colon cancer cells in non‐adherent cultures were killed more effectively by combinations of pyrvinium pamoate (a Wnt pathway inhibitor) and ABT263 (a pro‐apoptotic Bcl‐2 family inhibitor) or Ly29004 (a PI3kinase inhibitor). However, in a mouse xenograft model, the formulation and toxicity of the ABT737/PP combination prevent the use of these drugs for treatment of tumours. Fortunately, oral analogues of PP (pyrvinium phosphate, PPh) and ABT737(ABT263) have equivalent activity and can be used for treatment of mice carrying SW620 colorectal cancer xenografts. The PPh/ABT263 induced SW620 tumour cell apoptosis and reduced the rate of SW620 tumour growth. Conclusion By combining a Wnt signaling inhibitor (pyrvinium phosphate) and a pro‐survival inhibitor (ABT263) colon cancer cells can be killed. Combinations of Wnt signalling inhibitors with an inhibitor of the Bcl pro‐survival protein family should be considered for the treatment of patients with precancerous colon adenomas or advanced colorectal cancers with APC mutations.
Collapse
Affiliation(s)
- Silvia Paola Corona
- Structural Biology Division, WEHI, Parkville, Australia.,Personalised Oncology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia
| | - Francesca Walker
- Structural Biology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia
| | - Janet Weinstock
- Structural Biology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia
| | - Guillaume Lessene
- Department of Medical Biology, University of Melbourne, Parkville, Australia.,Chemical Biology Division, WEHI, Parkville, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Australia
| | - Maree Faux
- Structural Biology Division, WEHI, Parkville, Australia.,Personalised Oncology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Antony W Burgess
- Structural Biology Division, WEHI, Parkville, Australia.,Personalised Oncology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia
| |
Collapse
|
4
|
Lv C, Wang H, Tong Y, Yin H, Wang D, Yan Z, Liang Y, Wu D, Su Q. The function of BTG3 in colorectal cancer cells and its possible signaling pathway. J Cancer Res Clin Oncol 2018; 144:295-308. [PMID: 29270670 PMCID: PMC5794823 DOI: 10.1007/s00432-017-2561-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/16/2017] [Indexed: 01/02/2023]
Abstract
PURPOSE B-cell translocation gene 3 (BTG3) has been identified as a candidate driver gene for various cancers, but its specific role in colorectal cancer (CRC) is poorly understood. We aimed to investigate the relationship between expression of BTG3 and clinicopathological features and prognosis, as well as to explore the effects and the role of a possible BTG3 molecular mechanism on aggressive colorectal cancer behavior. METHODS BTG3 expression was assessed by immunohistochemistry (IHC) on specimens from 140 patients with CRC. The association of BTG3 expression with clinicopathological features was examined. To confirm the biological role of BTG3 in CRC, two CRC cell lines expressing BTG3 were used and BTG3 expression was knocked down by shRNA. CCK-8, cell cycle, apoptosis, migration, and invasion assays were performed. The influence of BTG3 knockdown was further investigated by genomic microarray to uncover the potential molecular mechanisms underlying BTG3-mediated CRC development and progression. RESULTS BTG3 was downregulated in colorectal cancer tissues and positively correlated with pathological classification (p = 0.037), depth of invasion (p = 0.016), distant metastasis (p = 0.024), TNM stage (p = 0.007), and overall survival (OS) and disease-free survival (DFS). BTG3 knockdown promoted cell proliferation, migration, invasion, relieved G2 arrest, and inhibited apoptosis in HCT116 and LoVo cells. A genomic microarray analysis showed that numerous tumor-associated signaling pathways and oncogenes were altered by BTG3 knockdown. At the mRNA level, nine genes referred to the extracellular-regulated kinase/mitogen-activated protein kinase pathway were differentially expressed. Western blotting revealed that BTG3 knockdown upregulated PAK2, RPS6KA5, YWHAB, and signal transducer and activator of transcription (STAT)3 protein levels, but downregulated RAP1A, DUSP6, and STAT1 protein expression, which was consistent with the genomic microarray data. CONCLUSIONS BTG3 expression might contribute to CRC carcinogenesis. BTG3 knockdown might strengthen the aggressive colorectal cancer behavior.
Collapse
Affiliation(s)
- Chi Lv
- Department of General Surgery, Shengjing Hospital Affiliated to China Medical University, Shenyang City, Liaoning Province, 110004, People's Republic of China
- Department of General Surgery, General Hospital of Shenyang Military Region, Shenyang, People's Republic of China
| | - Heling Wang
- Department of General Surgery, Shengjing Hospital Affiliated to China Medical University, Shenyang City, Liaoning Province, 110004, People's Republic of China
| | - Yuxin Tong
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Hongzhuan Yin
- Department of General Surgery, Shengjing Hospital Affiliated to China Medical University, Shenyang City, Liaoning Province, 110004, People's Republic of China
| | - Dalu Wang
- Department of General Surgery, Shengjing Hospital Affiliated to China Medical University, Shenyang City, Liaoning Province, 110004, People's Republic of China
| | - Zhaopeng Yan
- Department of General Surgery, Shengjing Hospital Affiliated to China Medical University, Shenyang City, Liaoning Province, 110004, People's Republic of China
| | - Yichao Liang
- Department of General Surgery, Shengjing Hospital Affiliated to China Medical University, Shenyang City, Liaoning Province, 110004, People's Republic of China
| | - Di Wu
- Department of General Surgery, Shengjing Hospital Affiliated to China Medical University, Shenyang City, Liaoning Province, 110004, People's Republic of China
| | - Qi Su
- Department of General Surgery, Shengjing Hospital Affiliated to China Medical University, Shenyang City, Liaoning Province, 110004, People's Republic of China.
| |
Collapse
|
5
|
Vincan E, Schwab RHM, Flanagan DJ, Moselen JM, Tran BM, Barker N, Phesse TJ. The Central Role of Wnt Signaling and Organoid Technology in Personalizing Anticancer Therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 153:299-319. [PMID: 29389521 DOI: 10.1016/bs.pmbts.2017.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Wnt pathway is at the heart of organoid technology, which is set to revolutionize the cancer field. We can now predetermine a patient's response to any given anticancer therapy by exposing tumor organoids established from the patient's own tumor. This cutting-edge biomedical platform translates to patients being treated with the correct drug at the correct dose from the outset, a truly personalized and precise medical approach. A high throughput drug screen on organoids also allows drugs to be tested in limitless combinations. More recently, the tumor cells that are resistant to the therapy given to a patient were selected in culture using the patient's organoids. The resistant tumor organoids were then screened empirically to identify drugs that will kill the resistant cells. This information allows diagnosis in real-time to either prevent tumor recurrence or effectively treat the recurring tumor. Furthermore, the ability to culture stem cell-derived epithelium as organoids has enabled us to begin to understand how a stem cell becomes a cancer cell or to pin-point the genetic alteration that underlies a given genetic syndrome. Here we summarize these advances and the central role of Wnt signaling, and identify the next challenges for organoid technology.
Collapse
Affiliation(s)
- Elizabeth Vincan
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia; Curtin University, Perth, WA, Australia.
| | - Renate H M Schwab
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Dustin J Flanagan
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Jean M Moselen
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Bang M Tran
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Nick Barker
- A*STAR Institute of Medical Biology, Singapore
| | - Toby J Phesse
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia; European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
6
|
Inhibition of Wnt/β-Catenin pathway and Histone acetyltransferase activity by Rimonabant: a therapeutic target for colon cancer. Sci Rep 2017; 7:11678. [PMID: 28916833 PMCID: PMC5601949 DOI: 10.1038/s41598-017-11688-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022] Open
Abstract
In a high percentage (≥85%) of both sporadic and familial adenomatous polyposis forms of colorectal cancer (CRC), the inactivation of the APC tumor suppressor gene initiates tumor formation and modulates the Wnt/β-Catenin transduction pathways involved in the control of cell proliferation, adhesion and metastasis. Increasing evidence showed that the endocannabinoids control tumor growth and progression, both in vitro and in vivo. We evaluated the effect of Rimonabant, a Cannabinoid Receptor 1 (CB1) inverse agonist, on the Wnt/β-Catenin pathway in HCT116 and SW48 cell lines carrying the genetic profile of metastatic CRC poorly responsive to chemotherapies. In these models, Rimonabant inhibited the Wnt/β-Catenin canonical pathway and increased β-Catenin phosphorylation; in HCT116 cells, but not in SW48, the compound also triggered the Wnt/β-Catenin non canonical pathway activation through induction of Wnt5A and activation of CaMKII. The Rimonabant-induced downregulation of Wnt/β-Catenin target genes was partially ascribable to a direct inhibition of p300/KAT3B histone acetyltransferase, a coactivator of β-Catenin dependent gene regulation. Finally, in HCT116 xenografts, Rimonabant significantly reduced tumor growth and destabilized the nuclear localization of β-Catenin. Obtained data heavily supported the rationale for the use of cannabinoids in combined therapies for metastatic CRC harbouring activating mutations of β-Catenin.
Collapse
|
7
|
Jackson H, Granger D, Jones G, Anderson L, Friel S, Rycroft D, Fieles W, Tunstead J, Steward M, Wattam T, Walker A, Griggs J, Al-Hajj M, Shelton C. Novel Bispecific Domain Antibody to LRP6 Inhibits Wnt and R-spondin Ligand-Induced Wnt Signaling and Tumor Growth. Mol Cancer Res 2016; 14:859-68. [DOI: 10.1158/1541-7786.mcr-16-0088] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/22/2016] [Indexed: 11/16/2022]
|
8
|
Jardé T, Kass L, Staples M, Lescesen H, Carne P, Oliva K, McMurrick PJ, Abud HE. ERBB3 Positively Correlates with Intestinal Stem Cell Markers but Marks a Distinct Non Proliferative Cell Population in Colorectal Cancer. PLoS One 2015; 10:e0138336. [PMID: 26367378 PMCID: PMC4569358 DOI: 10.1371/journal.pone.0138336] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 08/28/2015] [Indexed: 01/28/2023] Open
Abstract
Several studies have suggested ERBB3/HER3 may be a useful prognostic marker for colorectal cancer. Tumours with an intestinal stem cell signature have also been shown to be more aggressive. Here, we investigate whether ERBB3 is associated with intestinal stem cell markers in colorectal cancer and if cancer stem cells within tumours are marked by expression of ERBB3. Expression of ERBB3 and intestinal stem cell markers (LGR5, EPHB2, CD44s and CD44v6) was assessed by qRT-PCR in primary colorectal tumours (stages 0 to IV) and matched normal tissues from 53 patients. The localisation of ERBB3, EPHB2 and KI-67 within tumours was investigated using co-immunofluorescence. Expression of ERBB3 and intestinal stem cell markers were significantly elevated in adenomas and colorectal tumours compared to normal tissue. Positive correlations were found between ERBB3 and intestinal stem cell markers. However, co-immunofluorescence analysis showed that ERBB3 and EPHB2 marked specific cell populations that were mutually exclusive within tumours with distinct proliferative potentials, the majority of ERBB3+ve cells being non-proliferative. This pattern resembles cellular organisation within normal colonic epithelium where EPHB2 labelled proliferative cells reside at the crypt base and ERBB3+ve cells mark differentiated cells at the top of crypts. Our results show that ERBB3 and intestinal stem cell markers correlate in colorectal cancers. ERBB3 localises to differentiated cell populations within tumours that are non-proliferative and distinct from cancer stem cells. These data support the concept that tumours contain discrete stem, proliferative and differentiation compartments similar to that present in normal crypts.
Collapse
Affiliation(s)
- Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash University, Wellington Rd., Clayton, Victoria, Australia; Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Lisa Kass
- Department of Anatomy and Developmental Biology, Monash University, Wellington Rd., Clayton, Victoria, Australia
| | | | - Helen Lescesen
- Department of Anatomy and Developmental Biology, Monash University, Wellington Rd., Clayton, Victoria, Australia
| | - Peter Carne
- Department of Surgery, Cabrini Monash University, Malvern, Victoria, Australia
| | - Karen Oliva
- Department of Surgery, Cabrini Monash University, Malvern, Victoria, Australia
| | - Paul J McMurrick
- Department of Surgery, Cabrini Monash University, Malvern, Victoria, Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Wellington Rd., Clayton, Victoria, Australia
| |
Collapse
|
9
|
Quesenberry PJ, Aliotta J, Camussi G, Abdel-Mageed AB, Wen S, Goldberg L, Zhang HG, Tetta C, Franklin J, Coffey RJ, Danielson K, Subramanya V, Ghiran I, Das S, Chen CC, Pusic KM, Pusic AD, Chatterjee D, Kraig RP, Balaj L, Dooner M. Potential functional applications of extracellular vesicles: a report by the NIH Common Fund Extracellular RNA Communication Consortium. J Extracell Vesicles 2015; 4:27575. [PMID: 26320942 PMCID: PMC4553260 DOI: 10.3402/jev.v4.27575] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 07/15/2015] [Accepted: 07/24/2015] [Indexed: 12/13/2022] Open
Abstract
The NIH Extracellular RNA Communication Program's initiative on clinical utility of extracellular RNAs and therapeutic agents and developing scalable technologies is reviewed here. Background information and details of the projects are presented. The work has focused on modulation of target cell fate by extracellular vesicles (EVs) and RNA. Work on plant-derived vesicles is of intense interest, and non-mammalian sources of vesicles may represent a very promising source for different therapeutic approaches. Retro-viral-like particles are intriguing. Clearly, EVs share pathways with the assembly machinery of several other viruses, including human endogenous retrovirals (HERVs), and this convergence may explain the observation of viral-like particles containing viral proteins and nucleic acid in EVs. Dramatic effect on regeneration of damaged bone marrow, renal, pulmonary and cardiovascular tissue is demonstrated and discussed. These studies show restoration of injured cell function and the importance of heterogeneity of different vesicle populations. The potential for neural regeneration is explored, and the capacity to promote and reverse neoplasia by EV exposure is described. The tremendous clinical potential of EVs underlies many of these projects, and the importance of regulatory issues and the necessity of general manufacturing production (GMP) studies for eventual clinical trials are emphasized. Clinical trials are already being pursued and should expand dramatically in the near future.
Collapse
Affiliation(s)
- Peter J Quesenberry
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA;
| | - Jason Aliotta
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Torino, Italy
| | - Asim B Abdel-Mageed
- Department of Urology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sicheng Wen
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Laura Goldberg
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Huang-Ge Zhang
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Ciro Tetta
- Department of Medical Sciences, University of Turin, Torino, Italy
| | - Jeffrey Franklin
- Department of Molecular Biology, Vanderbilt University, Nashville, TN, USA
| | - Robert J Coffey
- Department of Molecular Biology, Vanderbilt University, Nashville, TN, USA
| | - Kirsty Danielson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vinita Subramanya
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ionita Ghiran
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Saumya Das
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Clark C Chen
- Department of Surgery, Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Kae M Pusic
- Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Aya D Pusic
- Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Devasis Chatterjee
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Richard P Kraig
- Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Leonora Balaj
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Mark Dooner
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
10
|
Carson R, Celtikci B, Fenning C, Javadi A, Crawford N, Carbonell LP, Lawler M, Longley DB, Johnston PG, Van Schaeybroeck S. HDAC Inhibition Overcomes Acute Resistance to MEK Inhibition in BRAF-Mutant Colorectal Cancer by Downregulation of c-FLIPL. Clin Cancer Res 2015; 21:3230-3240. [PMID: 25813020 PMCID: PMC4504978 DOI: 10.1158/1078-0432.ccr-14-2701] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/06/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Activating mutations in the BRAF oncogene are found in 8% to 15% of colorectal cancer patients and have been associated with poor survival. In contrast with BRAF-mutant (MT) melanoma, inhibition of the MAPK pathway is ineffective in the majority of BRAFMT colorectal cancer patients. Therefore, identification of novel therapies for BRAFMT colorectal cancer is urgently needed. EXPERIMENTAL DESIGN BRAFMT and wild-type (WT) colorectal cancer models were assessed in vitro and in vivo. Small-molecule inhibitors of MEK1/2, MET, and HDAC were used, overexpression and siRNA approaches were applied, and cell death was assessed by flow cytometry, Western blotting, cell viability, and caspase activity assays. RESULTS Increased c-MET-STAT3 signaling was identified as a novel adaptive resistance mechanism to MEK inhibitors (MEKi) in BRAFMT colorectal cancer models in vitro and in vivo. Moreover, MEKi treatment resulted in acute increases in transcription of the endogenous caspase-8 inhibitor c-FLIPL in BRAFMT cells, but not in BRAFWT cells, and inhibition of STAT3 activity abrogated MEKi-induced c-FLIPL expression. In addition, treatment with c-FLIP-specific siRNA or HDAC inhibitors abrogated MEKi-induced upregulation of c-FLIPL expression and resulted in significant increases in MEKi-induced cell death in BRAFMT colorectal cancer cells. Notably, combined HDAC inhibitor/MEKi treatment resulted in dramatically attenuated tumor growth in BRAFMT xenografts. CONCLUSIONS Our findings indicate that c-MET/STAT3-dependent upregulation of c-FLIPL expression is an important escape mechanism following MEKi treatment in BRAFMT colorectal cancer. Thus, combinations of MEKi with inhibitors of c-MET or c-FLIP (e.g., HDAC inhibitors) could be potential novel treatment strategies for BRAFMT colorectal cancer.
Collapse
Affiliation(s)
- Robbie Carson
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Basak Celtikci
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Cathy Fenning
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Arman Javadi
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Nyree Crawford
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Lucia Perez Carbonell
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Mark Lawler
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Daniel B. Longley
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Patrick G. Johnston
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Sandra Van Schaeybroeck
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| |
Collapse
|
11
|
Sakamori R, Yu S, Zhang X, Hoffman A, Sun J, Das S, Vedula P, Li G, Fu J, Walker F, Yang CS, Yi Z, Hsu W, Yu DH, Shen L, Rodriguez AJ, Taketo MM, Bonder EM, Verzi MP, Gao N. CDC42 inhibition suppresses progression of incipient intestinal tumors. Cancer Res 2014; 74:5480-92. [PMID: 25113996 DOI: 10.1158/0008-5472.can-14-0267] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mutations in the APC or β-catenin genes are well-established initiators of colorectal cancer, yet modifiers that facilitate the survival and progression of nascent tumor cells are not well defined. Using genetic and pharmacologic approaches in mouse colorectal cancer and human colorectal cancer xenograft models, we show that incipient intestinal tumor cells activate CDC42, an APC-interacting small GTPase, as a crucial step in malignant progression. In the mouse, Cdc42 ablation attenuated the tumorigenicity of mutant intestinal cells carrying single APC or β-catenin mutations. Similarly, human colorectal cancer with relatively higher levels of CDC42 activity was particularly sensitive to CDC42 blockade. Mechanistic studies suggested that Cdc42 may be activated at different levels, including at the level of transcriptional activation of the stem cell-enriched Rho family exchange factor Arhgef4. Our results indicate that early-stage mutant intestinal epithelial cells must recruit the pleiotropic functions of Cdc42 for malignant progression, suggesting its relevance as a biomarker and therapeutic target for selective colorectal cancer intervention.
Collapse
Affiliation(s)
- Ryotaro Sakamori
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Xiao Zhang
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Andrew Hoffman
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, New Jersey
| | - Jiaxin Sun
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Soumyashree Das
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Pavan Vedula
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Guangxun Li
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey
| | - Jiang Fu
- Department of Biomedical Genetics, Center for Oral Biology, James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | | | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey. Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Zheng Yi
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, Cincinnati, Ohio
| | - Wei Hsu
- Department of Biomedical Genetics, Center for Oral Biology, James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Da-Hai Yu
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Lanlan Shen
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Alexis J Rodriguez
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Makoto M Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Michael P Verzi
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, New Jersey. Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey. Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
| |
Collapse
|
12
|
RAD21 cohesin overexpression is a prognostic and predictive marker exacerbating poor prognosis in KRAS mutant colorectal carcinomas. Br J Cancer 2014; 110:1606-13. [PMID: 24548858 PMCID: PMC3960611 DOI: 10.1038/bjc.2014.31] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/26/2013] [Accepted: 01/07/2014] [Indexed: 12/27/2022] Open
Abstract
Background: RAD21 is a component of the cohesion complex and is integral to chromosome segregation and error-free DNA repair. RAD21 is functionally important in tumour progression but its role in colorectal carcinoma (CRC) is unclear. We therefore assessed its clinicopathological and prognostic significance in CRC, as well as its effect on chemosensitivity. Methods: A retrospective observation study examined RAD21 expression in 652 CRCs using a tissue microarray approach. Correlation with clinicopathological factors including gender, tumour grade, mucinous subtype, TNM stage, disease-specific survival (DSS), BRAF and KRAS mutation status, tumour p53 immunostaining, tumour microsatellite instability and tumour CpG island methylator phenotype was performed. Colorectal cancer cell clones with stable RAD21 knockdown were generated and tested for cellular sensitivity to conventional chemotherapeutic drugs. Results: RAD21 expression was significantly correlated with male gender (56.7% vs 43.3%, P=0.02), well-differentiated histology (14.4% vs 4.0%, P=0.0001), higher T-stage (36.1% vs 27.0%, P=0.01), presence of metastasis (18.8% vs 12.6%, P=0.03), and shorter DSS (hazard ratio (HR) 1.4, 95% CI 1.1 to 1.9, P=0.01) in both univariate and multivariate analysis. RAD21 expression was associated with shorter DSS in patients with KRAS mutant tumours (HR:2.6, 95% CI:1.4–4.3, P=0.001) and in patients receiving adjuvant chemoradiotherapy (HR:1.9, 95% CI:1.2–3.0, P=0.008). Colorectal cancer cells with RAD21 knockdown exhibited enhanced sensitivity to 5-fluorouracil, either alone or in combination with oxaliplatin. Conclusions: RAD21 expression in CRC is associated with aggressive disease especially in KRAS mutant tumours and resistance to chemoradiotherapy. RAD21 may be an important novel therapeutic target.
Collapse
|
13
|
Zecchin D, Boscaro V, Medico E, Barault L, Martini M, Arena S, Cancelliere C, Bartolini A, Crowley EH, Bardelli A, Gallicchio M, Di Nicolantonio F. BRAF V600E is a determinant of sensitivity to proteasome inhibitors. Mol Cancer Ther 2013; 12:2950-61. [PMID: 24107445 DOI: 10.1158/1535-7163.mct-13-0243] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A critical step toward defining tailored therapy in patients with cancer is the identification of genetic interactions that may impair-or boost-the efficacy of selected therapeutic approaches. Cell models able to recapitulate combinations of genetic aberrations are important to find drug-genotype interactions poorly affected by the heterogeneous genetics of human tumors. In order to identify novel pharmacogenomic relationships, we employed an isogenic cell panel that reconstructs cancer genetic scenarios. We screened a library of 43 compounds in human hTERT-HME1 epithelial cells in which PTEN or RB1 were silenced in combination with the targeted knockin of cancer-associated mutations in EGFR, KRAS, BRAF, or PIK3CA oncogenes. Statistical analysis and clustering algorithms were applied to display similar drug response profiles and mutation-specific patterns of activity. From the screen, we discovered that proteasome inhibitors show selectivity toward BRAF V600E-mutant cells, irrespective of PTEN or RB1 expression. Preferential targeting of BRAF-mutant cells by proteasome inhibitors was corroborated in a second BRAF V600E isogenic model, as well as in a panel of colorectal cancer cell lines by the use of the proteasome inhibitor carfilzomib. Notably, carfilzomib also showed striking in vivo activity in a BRAF-mutant human colorectal cancer xenograft model. Vulnerability to proteasome inhibitors is dependent on persistent BRAF signaling, because BRAF V600E blockade by PLX4720 reversed sensitivity to carfilzomib in BRAF-mutant colorectal cancer cells. Our findings indicated that proteasome inhibition might represent a valuable targeting strategy in BRAF V600E-mutant colorectal tumors.
Collapse
Affiliation(s)
- Davide Zecchin
- Corresponding Authors: Federica Di Nicolantonio, Department of Oncology, University of Torino, Institute for Cancer Research and Treatment at Candiolo, Strada Provinciale 142 Km 3.95, Candiolo, I-10060, Turin, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Greening DW, Kapp EA, Ji H, Speed TP, Simpson RJ. Colon tumour secretopeptidome: insights into endogenous proteolytic cleavage events in the colon tumour microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2396-407. [PMID: 23684732 DOI: 10.1016/j.bbapap.2013.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 04/26/2013] [Accepted: 05/08/2013] [Indexed: 12/20/2022]
Abstract
The secretopeptidome comprises endogenous peptides derived from proteins secreted into the tumour microenvironment through classical and non-classical secretion. This study characterised the low-Mr (<3kDa) component of the human colon tumour (LIM1215, LIM1863) secretopeptidome, as a first step towards gaining insights into extracellular proteolytic cleavage events in the tumour microenvironment. Based on two biological replicates, this secretopeptidome isolation strategy utilised differential centrifugal ultrafiltration in combination with analytical RP-HPLC and nanoLC-MS/MS. Secreted peptides were identified using a combination of Mascot and post-processing analyses including MSPro re-scoring, extended feature sets and Percolator, resulting in 474 protein identifications from 1228 peptides (≤1% q-value, ≤5% PEP) - a 36% increase in peptide identifications when compared with conventional Mascot (homology ionscore thresholding). In both colon tumour models, 122 identified peptides were derived from 41 cell surface protein ectodomains, 23 peptides (12 proteins) from regulated intramembrane proteolysis (RIP), and 12 peptides (9 proteins) generated from intracellular domain proteolysis. Further analyses using the protease/substrate database MEROPS, (http://merops.sanger.ac.uk/), revealed 335 (71%) proteins classified as originating from classical/non-classical secretion, or the cell membrane. Of these, peptides were identified from 42 substrates in MEROPS with defined protease cleavage sites, while peptides generated from a further 205 substrates were fragmented by hitherto unknown proteases. A salient finding was the identification of peptides from 88 classical/non-classical secreted substrates in MEROPS, implicated in tumour progression and angiogenesis (FGFBP1, PLXDC2), cell-cell recognition and signalling (DDR1, GPA33), and tumour invasiveness and metastasis (MACC1, SMAGP); the nature of the proteases responsible for these proteolytic events is unknown. To confirm reproducibility of peptide fragment abundance in this study, we report the identification of a specific cleaved peptide fragment in the secretopeptidome from the colon-specific GPA33 antigen in 4/14 human CRC models. This improved secretopeptidome isolation and characterisation strategy has extended our understanding of endogenous peptides generated through proteolysis of classical/non-classical secreted proteins, extracellular proteolytic processing of cell surface membrane proteins, and peptides generated through RIP. The novel peptide cleavage site information in this study provides a useful first step in detailing proteolytic cleavage associated with tumourigenesis and the extracellular environment. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
Affiliation(s)
- David W Greening
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia.
| | | | | | | | | |
Collapse
|
15
|
Barthel ER, Levin DE, Speer AL, Sala FG, Torashima Y, Hou X, Grikscheit TC. Human tissue-engineered colon forms from postnatal progenitor cells: an in vivo murine model. Regen Med 2013; 7:807-18. [PMID: 23164081 DOI: 10.2217/rme.12.91] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIM Loss of colon reservoir function after colectomy can adversely affect patient outcomes. In previous work, human fetal intestinal cells developed epithelium without mesenchyme following implantation in mice. However, for humans, postnatal tissue would be the preferred donor source. We generated tissue-engineered colon (TEC) from postnatal human organoid units. MATERIALS & METHODS Organoid units were prepared from human colon waste specimens, loaded onto biodegradable scaffolds and implanted into immunocompromised mice. After 4 weeks, human TEC was harvested. Immunofluorescence staining confirmed human origin, identified differentiated epithelial cell types and verified the presence of supporting mesenchyme. RESULTS Human TEC demonstrated a simple columnar epithelium. Immunofluorescence staining demonstrated human origin and the three differentiated cell types of mature colon epithelium. Key mesenchymal components (smooth muscle, intestinal subepithelial myofibroblasts and ganglion cells) were seen. CONCLUSION Colon can form from human progenitor cells on a scaffold in a mouse host. This proof-of-concept experiment is an important step in transitioning TEC to human therapy.
Collapse
Affiliation(s)
- Erik R Barthel
- Children's Hospital Los Angeles, Keck School of Medicine of the University of Southern California, Division of Pediatric Surgery, 4650 Sunset Boulevard Mailstop #35, Los Angeles, CA 90027, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Ludwig K, Tse ES, Wang JYJ. Colon cancer cells adopt an invasive phenotype without mesenchymal transition in 3-D but not 2-D culture upon combined stimulation with EGF and crypt growth factors. BMC Cancer 2013; 13:221. [PMID: 23638973 PMCID: PMC3667045 DOI: 10.1186/1471-2407-13-221] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 04/27/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The intestinal crypt homeostasis is maintained by a combination of growth factors including Wnt, R-Spondin1, Noggin and the epidermal growth factor (EGF). In human colorectal cancer, the Wnt pathway is constitutively activated through genetic and epigenetic alterations in as many as 11 genes encoding components of this crypt stem-cell maintenance mechanism. Although the proliferation of colon cancer cells does not require Wnt, it is possible that colon cancer cells can still respond to the crypt growth factors in the colonic microenvironment. A number of studies have shown that epithelial cells behave differently in 3-D versus 2-D cultures. Because the 3-D conditions more closely mimic the in vivo environment, we examined the effects of Wnt and other crypt growth factors on colon cancer cell growth in 3-D culture. METHODS Colon cancer cells were grown in 3-D matrigel supplemented with different combinations of crypt growth factors and colonies were examined for morphology and pathways. RESULTS When colon cancer cells were cultured in 3-D with EGF, they grew as round spheroid colonies. However, colon cancer cells also grew as flat, disc-like colonies when cultured with EGF plus Wnt, R-Spondin1 and Noggin. Disc colonies were found to have comparable levels of E-cadherin as the spheroid colonies, but showed decreased E-cadherin at the cell-matrix contact sites. Disc colonies also elaborated F-actin rich protrusions (FRP) at the cell-matrix edge, reminiscent of an invasive phenotype but without the expression of vimentin. These E-cadherin and F-actin alterations were not induced by the four growth factors in 2-D culture. Formation of the disc colonies was inhibited by the knockdown of β-catenin and by protein kinase inhibitors such as gefitinib, imatinib and MK-2206. Furthermore, withdrawal of the crypt growth factors was able to revert the disc colonies to spheroid growth, showing that the invasive phenotype was reversible dependent on the availability of growth factors. CONCLUSIONS These findings show that colon cancer cells remain responsive to the growth factors in the crypt microenvironment and can be induced to undergo morphological transformation in the more physiologically relevant 3-D culture.
Collapse
Affiliation(s)
- Kirsten Ludwig
- Moores UCSD Cancer Center, 3855 Health Sciences Drive, La Jolla, CA, 92093-0820, USA
| | - Edison S Tse
- Moores UCSD Cancer Center, 3855 Health Sciences Drive, La Jolla, CA, 92093-0820, USA
| | - Jean YJ Wang
- Moores UCSD Cancer Center, 3855 Health Sciences Drive, La Jolla, CA, 92093-0820, USA
- Department of Medicine, Division of Hematology-Oncology, University of California, La Jolla, CA, 92093, USA
| |
Collapse
|
17
|
Coffee EM, Faber AC, Roper J, Sinnamon MJ, Goel G, Keung L, Wang WV, Vecchione L, de Vriendt V, Weinstein BJ, Bronson RT, Tejpar S, Xavier RJ, Engelman JA, Martin ES, Hung KE. Concomitant BRAF and PI3K/mTOR blockade is required for effective treatment of BRAF(V600E) colorectal cancer. Clin Cancer Res 2013; 19:2688-98. [PMID: 23549875 DOI: 10.1158/1078-0432.ccr-12-2556] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE BRAF(V600E) mutations are associated with poor clinical prognosis in colorectal cancer (CRC). Although selective BRAF inhibitors are effective for treatment of melanoma, comparable efforts in CRC have been disappointing. Here, we investigated potential mechanisms underlying this resistance to BRAF inhibitors in BRAF(V600E) CRC. EXPERIMENTAL DESIGN We examined phosphoinositide 3-kinase (PI3K)/mTOR signaling in BRAF(V600E) CRC cell lines after BRAF inhibition and cell viability and apoptosis after combined BRAF and PI3K/mTOR inhibition. We assessed the efficacy of in vivo combination treatment using a novel genetically engineered mouse model (GEMM) for BRAF(V600E) CRC. RESULTS Western blot analysis revealed sustained PI3K/mTOR signaling upon BRAF inhibition. Our BRAF(V600E) GEMM presented with sessile serrated adenomas/polyps, as seen in humans. Combination treatment in vivo resulted in induction of apoptosis and tumor regression. CONCLUSIONS We have established a novel GEMM to interrogate BRAF(V600E) CRC biology and identify more efficacious treatment strategies. Combination BRAF and PI3K/mTOR inhibitor treatment should be explored in clinical trials.
Collapse
Affiliation(s)
- Erin M Coffee
- Division of Gastroenterology, Tufts Medical Center, Boston, Massachusetts 02111, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Catimel B, Kapp E, Yin MX, Gregory M, Wong LSM, Condron M, Church N, Kershaw N, Holmes AB, Burgess AW. The PI(3)P interactome from a colon cancer cell. J Proteomics 2013; 82:35-51. [DOI: 10.1016/j.jprot.2013.01.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 12/21/2012] [Accepted: 01/24/2013] [Indexed: 02/07/2023]
|
19
|
Wang G, Wang F, Ding W, Wang J, Jing R, Li H, Wang X, Wang Y, Ju S, Wang H. APRIL induces tumorigenesis and metastasis of colorectal cancer cells via activation of the PI3K/Akt pathway. PLoS One 2013; 8:e55298. [PMID: 23383143 DOI: 10.1371/journal.pone.0055298] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 12/20/2012] [Indexed: 12/25/2022] Open
Abstract
A proliferation-inducing ligand (APRIL) is highly expressed in colorectal cancer (CRC) tissues and cell lines. However, the biological functions and precise signals elicited by APRIL in CRC have not been fully understood. Here, we used small interfering RNA to selectively deplete APRIL and to determine its tumorigenic effects in a CRC cell line SW480 both in vitro and in vivo. Knockdown of APRIL in SW480 cells was associated with modulation of cell proliferation as well as reduction of cell migration and invasion in vitro. Moreover APRIL-knockdown SW480 cells displayed markedly inhibited tumor growth and decreased metastasis to the liver in immunodeficient mice upon subcutaneous injection. Importantly, we observed that downregulation of APRIL in SW480 cells resulted in greatly decreased activity of phosphoinositide 3-kinase (PI3K)/Akt pathway. In addition, we observed that recombinant human APRIL mediated activation of the PI3K/Akt pathway in CRC cells resulting in induced expression of important cell cycle proteins and matrix metalloproteinases in a PI3K/Akt dependent manner. This was concurrent with marked cell growth viability as well as increased cell migration and invasion. Together, these compelling data suggest that APRIL-induced tumorigenesis and metastasis of CRC cells may be accomplished through activation of the PI3K/Akt pathway. These findings may lead to a better understanding of the biological effects of APRIL and may provide clues for identifying novel therapeutic and preventive molecular markers for CRC.
Collapse
Affiliation(s)
- Guihua Wang
- Department of Clinical Laboratory Center, Affiliated Hospital of Nantong University, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Han J, Soletti RC, Sadarangani A, Sridevi P, Ramirez ME, Eckmann L, Borges HL, Wang JYJ. Nuclear expression of β-catenin promotes RB stability and resistance to TNF-induced apoptosis in colon cancer cells. Mol Cancer Res 2013; 11:207-18. [PMID: 23339186 DOI: 10.1158/1541-7786.mcr-12-0670] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor necrosis factor (TNF)-α promotes tumor development under chronic inflammation. Because TNF also activates caspase-8, selective inhibition of TNF-induced extrinsic apoptosis would be required for inflammation-associated tumor growth. In a mouse model of inflammation-associated colon carcinogenesis, we found nuclear expression of β-catenin in tumors of wild-type, but not mutant, mice that were made resistant to TNF-induced apoptosis by a germline mutation blocking caspase cleavage of the retinoblastoma (RB) protein, despite similar frequencies of β-catenin exon-3 mutations in these two genetic backgrounds. TNF-induced apoptosis was also attenuated in human colon cancer cell lines with genetically activated β-catenin. However, we found that HCT116 cells, which contain an activated allele of β-catenin but do not express nuclear β-catenin, were sensitive to TNF-induced apoptosis. In HCT116 cells, TNF stimulated efficient RB cleavage that preceded chromatin condensation. In contrast, TNF did not induce RB cleavage in colon cancer cells expressing nuclear β-catenin and these cells could be sensitized to basal and/or TNF-induced apoptosis by the knockdown of β-catenin or RB. In the apoptosis-resistant colon cancer cells, knockdown of β-catenin led to a reduction in the RB protein without affecting RB mRNA. Furthermore, ectopic expression of the caspase-resistant, but not the wild-type, RB re-established resistance to TNF-induced caspase activation in colon cancer cells without β-catenin. Together, these results suggest that nuclear β-catenin-dependent RB stabilization suppresses TNF-induced apoptosis in caspase-8-positive colon cancer cells.
Collapse
Affiliation(s)
- Jinbo Han
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Han J, Sridevi P, Ramirez M, Ludwig KJ, Wang JYJ. β-Catenin-dependent lysosomal targeting of internalized tumor necrosis factor-α suppresses caspase-8 activation in apoptosis-resistant colon cancer cells. Mol Biol Cell 2012; 24:465-73. [PMID: 23264463 PMCID: PMC3571869 DOI: 10.1091/mbc.e12-09-0662] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Tumor necrosis factor-α (TNF)-induced apoptotic activation of caspase-8 requires internalization of its receptor. This study shows that constitutively activated β-catenin is required to facilitate the lysosomal delivery of internalized TNF, the inhibition of caspase-8 activation, and the suppression of apoptosis in colon cancer cells. The Wnt/β-catenin pathway is constitutively activated in more than 90% of human colorectal cancer. Activated β-catenin stimulates cell proliferation and survival, however, its antiapoptotic mechanisms are not fully understood. We show here that activated β-catenin is required to suppress caspase-8 activation, but only in colon cancer cells that are resistant to tumor necrosis factor-α (TNF)-induced apoptosis. We found that lysosomal delivery of internalized TNF occurred at a faster pace in apoptosis-resistant than in apoptosis-sensitive colon cancer cells. Retardation of endosomal trafficking through vacuolar ATPase (V-ATPase) inhibition enhanced caspase-8 activation in apoptosis-resistant but not apoptosis-sensitive cells. Interestingly, knockdown of β-catenin also prolonged TNF association with the early endosome and enhanced caspase-8 activation in apoptosis-resistant but not apoptosis-sensitive colon cancer cells. In a mouse model of inflammation-associated colon tumors, we found nuclear expression of β-catenin, resistance to TNF-induced apoptosis, and reactivation of apoptosis in vivo after cotreatment of TNF with a V-ATPase inhibitor. Together these results suggest that activated β-catenin can facilitate endosomal trafficking of internalized TNF to suppress caspase-8 activation in colon cancer cells.
Collapse
Affiliation(s)
- Jinbo Han
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093-0820, USA
| | | | | | | | | |
Collapse
|
22
|
Fanayan S, Smith JT, Sethi MK, Cantor D, Goode R, Simpson RJ, Baker MS, Hancock WS, Nice E. Chromosome 7-Centric Analysis of Proteomics Data from a Panel of Human Colon Carcinoma Cell Lines. J Proteome Res 2012; 12:89-96. [DOI: 10.1021/pr300906y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Susan Fanayan
- Department of Chemistry and
Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Joshua T. Smith
- Barnett Institute and Department
of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Manveen K. Sethi
- Department of Chemistry and
Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - David Cantor
- Department of Chemistry and
Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Robert Goode
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Richard J. Simpson
- La
Trobe Institute for Molecular
Science, La Trobe University, Bundoora,
Victoria 3086, Australia
| | - Mark S. Baker
- Department of Chemistry and
Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - William S. Hancock
- Department of Chemistry and
Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Barnett Institute and Department
of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Edouard Nice
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
23
|
Mathivanan S, Ji H, Tauro BJ, Chen YS, Simpson RJ. Identifying mutated proteins secreted by colon cancer cell lines using mass spectrometry. J Proteomics 2012; 76 Spec No.:141-9. [PMID: 22796352 DOI: 10.1016/j.jprot.2012.06.031] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 06/05/2012] [Accepted: 06/21/2012] [Indexed: 01/15/2023]
Abstract
Secreted proteins encoded by mutated genes (mutant proteins) are a particularly rich source of biomarkers being not only components of the cancer secretome but also actually implicated in tumorigenesis. One of the challenges of proteomics-driven biomarker discovery research is that the bulk of secreted mutant proteins cannot be identified directly and quantified by mass spectrometry due to the lack of mutated peptide information in extant proteomics databases. Here we identify, using an integrated genomics and proteomics strategy (referred to iMASp - identification of Mutated And Secreted proteins), 112 putative mutated tryptic peptides (corresponding to 57 proteins) in the collective secretomes derived from a panel of 18 human colorectal cancer (CRC) cell lines. Central to this iMASp was the creation of Human Protein Mutant Database (HPMD), against which experimentally-derived secretome peptide spectra were searched. Eight of the identified mutated tryptic peptides were confirmed by RT-PCR and cDNA sequencing of RNA extracted from those CRC cells from which the mutation was identified by mass spectrometry. The iMASp technology promises to improve the link between proteomics and genomic mutation data thereby providing an effective tool for targeting tryptic peptides with mutated amino acids as potential cancer biomarker candidates. This article is part of a Special Issue entitled: Integrated omics.
Collapse
Affiliation(s)
- Suresh Mathivanan
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | | | | | | | | |
Collapse
|
24
|
Abstract
The past decade has seen rapid advancement in the dissection of the molecular events and players in the development and homeostasis of mineralized tissues, that is, teeth and bones. Much of this is due to research efforts toward the regeneration of these organs and also to develop treatments for pathologies of bone, especially osteoporosis. Of late, great interest has been focused on the Wnt family of proteins and their involvement in tooth and bone development and in the regulation of postnatal bone mass. The purpose of this review is to summarize these findings and to explore new areas of Wnt research such as Wnt?bone morphogenetic protein interactions and the exciting revelation of systemic serotonin being involved in bone mass regulation.
Collapse
Affiliation(s)
- Kevin A Tompkins
- Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
25
|
Burgess AW, Faux MC, Layton MJ, Ramsay RG. Wnt signaling and colon tumorigenesis--a view from the periphery. Exp Cell Res 2011; 317:2748-58. [PMID: 21884696 DOI: 10.1016/j.yexcr.2011.08.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 08/10/2011] [Accepted: 08/13/2011] [Indexed: 02/06/2023]
Abstract
In this brief overview we discuss the association between Wnt signaling and colon cell biology and tumorigenesis. Our current understanding of the role of Apc in the β-catenin destruction complex is compared with potential roles for Apc in cell adhesion and migration. The requirement for phosphorylation in the proteasomal-mediated degradation of β-catenin is contrasted with roles for phospho-β-catenin in the activation of transcription, cell adhesion and migration. The synergy between Myb and β-catenin regulation of transcription in crypt stem cells during Wnt signaling is discussed. Finally, potential effects of growth factor regulatory systems, Apc or truncated-Apc on crypt morphogenesis, stem cell localization and crypt fission are considered.
Collapse
Affiliation(s)
- Antony W Burgess
- Parkville Branch, Ludwig Institute for Cancer Research, Melbourne, 3050, Australia.
| | | | | | | |
Collapse
|
26
|
Walker F, Zhang HH, Odorizzi A, Burgess AW. LGR5 is a negative regulator of tumourigenicity, antagonizes Wnt signalling and regulates cell adhesion in colorectal cancer cell lines. PLoS One 2011; 6:e22733. [PMID: 21829496 PMCID: PMC3145754 DOI: 10.1371/journal.pone.0022733] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 07/04/2011] [Indexed: 02/07/2023] Open
Abstract
Background LGR5 (Leucine-rich repeat-containing G-protein coupled receptor 5) is the most established marker for intestinal stem cells. Mouse models show that LGR5+ cells are the cells of origin of intestinal cancer, and LGR5 expression is elevated in human colorectal cancers, however very little is known about LGR5 function or its contribution to the stem cell phenotype and to colorectal cancer. Principal Findings We have modulated the expression of LGR5 by RNAi (inhibitory RNAs) or overexpression in colorectal cancer cell lines. Paradoxically, ablation of LGR5 induces increased invasion and anchorage-independent growth, and enhances tumourigenicity in xenografts experiments. Conversely, overexpression of LGR5 augments cell adhesion, reduces clonogenicity and attenuates tumourigenicity. Expression profiling revealed enhanced wnt signalling and upregulation of EMT genes upon knockdown of LGR5, with opposite changes in LGR5 overexpressing cells. These findings suggest that LGR5 is important in restricting stem cells to their niche, and that loss of LGR5 concomitant with activated wnt signalling may contribute to the invasive phenotype of colorectal carcinomas.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cell Adhesion/physiology
- Cell Movement
- Cell Proliferation
- Colony-Forming Units Assay
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Fluorescent Antibody Technique
- Gene Expression Profiling
- Humans
- Mice
- Mice, Nude
- Oligonucleotide Array Sequence Analysis
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- Wnt Proteins/genetics
- Wnt Proteins/metabolism
- Wound Healing
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Francesca Walker
- Epithelial Biochemistry Laboratory, Ludwig Institute for Cancer Research, Parkville, Victoria, Australia.
| | | | | | | |
Collapse
|
27
|
Recent advances in understanding the role of diet and obesity in the development of colorectal cancer. Proc Nutr Soc 2011; 70:194-204. [PMID: 21385524 DOI: 10.1017/s0029665111000073] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is a major cause of premature death in the UK and many developed countries. However, the risk of developing CRC is well recognised to be associated not only with diet but also with obesity and lack of exercise. While epidemiological evidence shows an association with factors such as high red meat intake and low intake of vegetables, fibre and fish, the mechanisms underlying these effects are only now being elucidated. CRC develops over many years and is typically characterised by an accumulation of mutations, which may arise as a consequence of inherited polymorphisms in key genes, but more commonly as a result of spontaneously arising mutations affecting genes controlling cell proliferation, differentiation, apoptosis and DNA repair. Epigenetic changes are observed throughout the progress from normal morphology through formation of adenoma, and the subsequent development of carcinoma. The reasons why this accumulation of loss of homoeostatic controls arises are unclear but chronic inflammation has been proposed to play a central role. Obesity is associated with increased plasma levels of chemokines and adipokines characteristic of chronic systemic inflammation, and dietary factors such as fish oils and phytochemicals have been shown to have anti-inflammatory properties as well as modulating established risk factors such as apoptosis and cell proliferation. There is also some evidence that diet can modify epigenetic changes. This paper briefly reviews the current state of knowledge in relation to CRC development and considers evidence for potential mechanisms by which diet may modify risk.
Collapse
|
28
|
Van Schaeybroeck S, Kyula JN, Fenton A, Fenning CS, Sasazuki T, Shirasawa S, Longley DB, Johnston PG. Oncogenic Kras promotes chemotherapy-induced growth factor shedding via ADAM17. Cancer Res 2011; 71:1071-80. [PMID: 21148749 PMCID: PMC3073126 DOI: 10.1158/0008-5472.can-10-0714] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Oncogenic mutations in Kras occur in 40% to 45% of patients with advanced colorectal cancer (CRC). We have previously shown that chemotherapy acutely activates ADAM17, resulting in growth factor shedding, growth factor receptor activation, and drug resistance in CRC tumors. In this study, we examined the role of mutant Kras in regulating growth factor shedding and ADAM17 activity, using isogenic Kras mutant (MT) and wild-type (WT) HCT116 CRC cells. Significantly higher levels of TGF-α and VEGF were shed from KrasMT HCT116 cells, both basally and following chemotherapy treatment, and this correlated with increased pErk (phosphorylated extracellular signal regulated kinase)1/2 levels and ADAM17 activity. Inhibition of Kras, MEK (MAP/ERK kinase)1/2, or Erk1/2 inhibition abrogated chemotherapy-induced ADAM17 activity and TGF-α shedding. Moreover, we found that these effects were not drug or cell line specific. In addition, MEK1/2 inhibition in KrasMT xenografts resulted in significant decreases in ADAM17 activity and growth factor shedding in vivo, which correlated with dramatically attenuated tumor growth. Furthermore, we found that MEK1/2 inhibition significantly induced apoptosis both alone and when combined with chemotherapy in KrasMT cells. Importantly, we found that sensitivity to MEK1/2 inhibition was ADAM17 dependent in vitro and in vivo. Collectively, our findings indicate that oncogenic Kras regulates ADAM17 activity and thereby growth factor ligand shedding in a MEK1/2/Erk1/2-dependent manner and that KrasMT CRC tumors are vulnerable to MEK1/2 inhibitors, at least in part, due to their dependency on ADAM17 activity.
Collapse
Affiliation(s)
- Sandra Van Schaeybroeck
- Drug Resistance Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, N. Ireland
| | - Joan N. Kyula
- Drug Resistance Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, N. Ireland
| | - Audrey Fenton
- Drug Resistance Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, N. Ireland
| | - Catherine S. Fenning
- Drug Resistance Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, N. Ireland
| | - Takehiko Sasazuki
- Institute for Advanced Study, Kyushu University, Higashi-ku, Fukuoka, Japan
| | - Senji Shirasawa
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Jonan-Ku, Fukuoka, Japan
| | - Daniel B. Longley
- Drug Resistance Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, N. Ireland
| | - Patrick G. Johnston
- Drug Resistance Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, N. Ireland
| |
Collapse
|
29
|
Williams DS, Bird MJ, Jorissen RN, Yu YL, Walker F, Zhang HH, Nice EC, Burgess AW. Nonsense mediated decay resistant mutations are a source of expressed mutant proteins in colon cancer cell lines with microsatellite instability. PLoS One 2010; 5:e16012. [PMID: 21209843 PMCID: PMC3013145 DOI: 10.1371/journal.pone.0016012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 12/03/2010] [Indexed: 12/16/2022] Open
Abstract
Background Frameshift mutations in microsatellite instability high (MSI-High) colorectal cancers are a potential source of targetable neo-antigens. Many nonsense transcripts are subject to rapid degradation due to nonsense-mediated decay (NMD), but nonsense transcripts with a cMS in the last exon or near the last exon-exon junction have intrinsic resistance to nonsense-mediated decay (NMD). NMD-resistant transcripts are therefore a likely source of expressed mutant proteins in MSI-High tumours. Methods Using antibodies to the conserved N-termini of predicted mutant proteins, we analysed MSI-High colorectal cancer cell lines for examples of naturally expressed mutant proteins arising from frameshift mutations in coding microsatellites (cMS) by immunoprecipitation and Western Blot experiments. Detected mutant protein bands from NMD-resistant transcripts were further validated by gene-specific short-interfering RNA (siRNA) knockdown. A genome-wide search was performed to identify cMS-containing genes likely to generate NMD-resistant transcripts that could encode for antigenic expressed mutant proteins in MSI-High colon cancers. These genes were screened for cMS mutations in the MSI-High colon cancer cell lines. Results Mutant protein bands of expected molecular weight were detected in mutated MSI-High cell lines for NMD-resistant transcripts (CREBBP, EP300, TTK), but not NMD-sensitive transcripts (BAX, CASP5, MSH3). Expression of the mutant CREBBP and EP300 proteins was confirmed by siRNA knockdown. Five cMS-bearing genes identified from the genome-wide search and without existing mutation data (SFRS12IP1, MED8, ASXL1, FBXL3 and RGS12) were found to be mutated in at least 5 of 11 (45%) of the MSI-High cell lines tested. Conclusion NMD-resistant transcripts can give rise to expressed mutant proteins in MSI-High colon cancer cells. If commonly expressed in primary MSI-High colon cancers, MSI-derived mutant proteins could be useful as cancer specific immunological targets in a vaccine targeting MSI-High colonic tumours.
Collapse
Affiliation(s)
- David S. Williams
- Epithelial Biochemistry Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Parkville, Victoria, Australia
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
- Department of Anatomical Pathology, Melbourne Health, Parkville, Victoria, Australia
- * E-mail: (DSW); (AWB)
| | - Matthew J. Bird
- Epithelial Biochemistry Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Parkville, Victoria, Australia
| | - Robert N. Jorissen
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Parkville, Victoria, Australia
| | - Yen Lin Yu
- Epithelial Biochemistry Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Parkville, Victoria, Australia
| | - Franscesa Walker
- Epithelial Biochemistry Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Parkville, Victoria, Australia
| | - Hui Hua Zhang
- Epithelial Biochemistry Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Parkville, Victoria, Australia
| | - Edouard C. Nice
- Epithelial Biochemistry Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Parkville, Victoria, Australia
| | - Antony W. Burgess
- Epithelial Biochemistry Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Parkville, Victoria, Australia
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Parkville, Victoria, Australia
- * E-mail: (DSW); (AWB)
| |
Collapse
|
30
|
Faux MC, Coates JL, Kershaw NJ, Layton MJ, Burgess AW. Independent interactions of phosphorylated β-catenin with E-cadherin at cell-cell contacts and APC at cell protrusions. PLoS One 2010; 5:e14127. [PMID: 21152425 PMCID: PMC2994709 DOI: 10.1371/journal.pone.0014127] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 10/23/2010] [Indexed: 01/17/2023] Open
Abstract
Background The APC tumour suppressor functions in several cellular processes including the regulation of β-catenin in Wnt signalling and in cell adhesion and migration. Findings In this study, we establish that in epithelial cells N-terminally phosphorylated β-catenin specifically localises to several subcellular sites including cell-cell contacts and the ends of cell protrusions. N-terminally phosphorylated β-catenin associates with E-cadherin at adherens junctions and with APC in cell protrusions. We isolated APC-rich protrusions from stimulated cells and detected β-catenin, GSK3β and CK1α, but not axin. The APC/phospho-β-catenin complex in cell protrusions appears to be distinct from the APC/axin/β-catenin destruction complex. GSK3β phosphorylates the APC-associated population of β-catenin, but not the cell junction population. β-catenin associated with APC is rapidly phosphorylated and dephosphorylated. HGF and wound-induced cell migration promote the localised accumulation of APC and phosphorylated β-catenin at the leading edge of migrating cells. APC siRNA and analysis of colon cancer cell lines show that functional APC is required for localised phospho-β-catenin accumulation in cell protrusions. Conclusions We conclude that N-terminal phosphorylation of β-catenin does not necessarily lead to its degradation but instead marks distinct functions, such as cell migration and/or adhesion processes. Localised regulation of APC-phospho-β-catenin complexes may contribute to the tumour suppressor activity of APC.
Collapse
Affiliation(s)
- Maree C Faux
- Ludwig Institute for Cancer Research, Melbourne, Victoria, Australia.
| | | | | | | | | |
Collapse
|
31
|
Palmieri M, Nowell CJ, Condron M, Gardiner J, Holmes AB, Desai J, Burgess AW, Catimel B. Analysis of cellular phosphatidylinositol (3,4,5)-trisphosphate levels and distribution using confocal fluorescent microscopy. Anal Biochem 2010; 406:41-50. [PMID: 20599646 DOI: 10.1016/j.ab.2010.06.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 06/18/2010] [Accepted: 06/22/2010] [Indexed: 11/29/2022]
Abstract
We have developed an immunocytochemistry method for the semiquantitative detection of phosphatidylinositol (3,4,5)-trisphosphate (PI(3,4,5)P3) at the cell plasma membrane. This protocol combines the use of a glutathione S-transferase-tagged pleckstrin homology (PH) domain of the general phosphoinositides-1 receptor (GST-GRP1PH) with fluorescence confocal microscopy and image segmentation using cell mask software analysis. This methodology allows the analysis of PI(3,4,5)P3 subcellular distribution in resting and epidermal growth factor (EGF)-stimulated HEK293T cells and in LIM1215 (wild-type phosphoinositide 3-kinase (PI3K)) and LIM2550 (H1047R mutation in PI3K catalytic domain) colonic carcinoma cells. Formation of PI(3,4,5)P3 was observed 5min following EGF stimulation and resulted in an increase of the membrane/cytoplasm fluorescence ratio from 1.03 to 1.53 for HEK293T cells and from 2.2 to 3.3 for LIM1215 cells. Resting LIM2550 cells stained with GST-GRP1PH had an elevated membrane/cytoplasm fluorescence ratio of 9.8, suggesting constitutive PI3K activation. The increase in the membrane/cytoplasm fluorescent ratio was inhibited in a concentration-dependent manner by the PI3K inhibitor LY294002. This cellular confocal imaging assay can be used to directly assess the effects of PI3K mutations in cancer cell lines and to determine the potential specificity and effectiveness of PI3K inhibitors in cancer cells.
Collapse
Affiliation(s)
- Michelle Palmieri
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victsoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Koch S, Capaldo CT, Samarin S, Nava P, Neumaier I, Skerra A, Sacks DB, Parkos CA, Nusrat A. Dkk-1 inhibits intestinal epithelial cell migration by attenuating directional polarization of leading edge cells. Mol Biol Cell 2009; 20:4816-25. [PMID: 19776352 DOI: 10.1091/mbc.e09-05-0415] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Wnt signaling pathways regulate proliferation, motility, and survival in a variety of human cell types. Dickkopf-1 (Dkk-1) is a secreted Wnt antagonist that has been proposed to regulate tissue homeostasis in the intestine. In this report, we show that Dkk-1 is secreted by intestinal epithelial cells after wounding and that it inhibits cell migration by attenuating the directional orientation of migrating epithelial cells. Dkk-1 exposure induced mislocalized activation of Cdc42 in migrating cells, which coincided with a displacement of the polarity protein Par6 from the leading edge. Consequently, the relocation of the microtubule organizing center and the Golgi apparatus in the direction of migration was significantly and persistently inhibited in the presence of Dkk-1. Small interfering RNA-induced down-regulation of Dkk-1 confirmed that extracellular exposure to Dkk-1 was required for this effect. Together, these data demonstrate a novel role of Dkk-1 in the regulation of directional polarization of migrating intestinal epithelial cells, which contributes to the effect of Dkk-1 on wound closure in vivo.
Collapse
Affiliation(s)
- Stefan Koch
- Epithelial Pathobiology Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Liu JP. From molecules and cells to diseases: West meets East for medical research in Tianjin. Cell Res 2009; 19:924-8. [PMID: 19597534 DOI: 10.1038/cr.2009.85] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Jun-Ping Liu
- Department of Immunology, Monash University, Commercial Road, Melbourne, Victoria 3004, Australia.
| |
Collapse
|