1
|
Mehra L, Bhowmik S, Makharia GK, Das P. Intestinal stem cell niche: An upcoming area of immense importance in gastrointestinal disorders. Indian J Gastroenterol 2024:10.1007/s12664-024-01699-8. [PMID: 39514159 DOI: 10.1007/s12664-024-01699-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/29/2024] [Indexed: 11/16/2024]
Abstract
The intestinal stem cell (ISC) niche is vital for maintaining the integrity and function of the intestinal epithelium. ISC populations, characterized by their high proliferation and multipotency, reside within a specialized microenvironment at the base of crypts. Crypt base columnar (CBC) cells at the deepest part of crypts serve as replicating ISCs, while position 4 label-retaining cells (LRCs) located higher up in the crypts are also important for ISC maintenance during experiments. The interplay between CBCs, position 4 LRCs, transient amplifying (TA) cells and other niche components, including the pericrypt stromal cells, ensures a continuous supply of differentiated epithelial cells. Recent advancements in ISC biomarker studies have provided valuable insights into their molecular signatures, regulatory pathways and roles in the pathogenesis of intestinal disorders. Understanding the ISC niche has significant therapeutic implications, as manipulating ISC behaviors and regenerating damaged or diseased intestinal tissue show promise for novel therapeutic approaches. ISC organoids have also provided a platform for studying intestinal diseases and testing personalized therapies. This comprehensive review covers the anatomical composition, physiological regulation, ISC biomarker studies, contribution to intestinal disorder pathogenesis and potential therapeutic implications of the ISC niche.
Collapse
Affiliation(s)
- Lalita Mehra
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110 029, India
| | - Subham Bhowmik
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110 029, India
| | - Govind K Makharia
- Department of Gastroenterology and Human Nutritions, All India Institute of Medical Sciences, New Delhi, 110 029, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110 029, India.
| |
Collapse
|
2
|
Morin A, Chu ECP, Sharma A, Adrian-Hamazaki A, Pavlidis P. Characterizing the targets of transcription regulators by aggregating ChIP-seq and perturbation expression data sets. Genome Res 2023; 33:763-778. [PMID: 37308292 PMCID: PMC10317128 DOI: 10.1101/gr.277273.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 04/26/2023] [Indexed: 06/14/2023]
Abstract
Mapping the gene targets of chromatin-associated transcription regulators (TRs) is a major goal of genomics research. ChIP-seq of TRs and experiments that perturb a TR and measure the differential abundance of gene transcripts are a primary means by which direct relationships are tested on a genomic scale. It has been reported that there is a poor overlap in the evidence across gene regulation strategies, emphasizing the need for integrating results from multiple experiments. Although research consortia interested in gene regulation have produced a valuable trove of high-quality data, there is an even greater volume of TR-specific data throughout the literature. In this study, we show a workflow for the identification, uniform processing, and aggregation of ChIP-seq and TR perturbation experiments for the ultimate purpose of ranking human and mouse TR-target interactions. Focusing on an initial set of eight regulators (ASCL1, HES1, MECP2, MEF2C, NEUROD1, PAX6, RUNX1, and TCF4), we identified 497 experiments suitable for analysis. We used this corpus to examine data concordance, to identify systematic patterns of the two data types, and to identify putative orthologous interactions between human and mouse. We build upon commonly used strategies to forward a procedure for aggregating and combining these two genomic methodologies, assessing these rankings against independent literature-curated evidence. Beyond a framework extensible to other TRs, our work also provides empirically ranked TR-target listings, as well as transparent experiment-level gene summaries for community use.
Collapse
Affiliation(s)
- Alexander Morin
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Eric Ching-Pan Chu
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Aman Sharma
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Alex Adrian-Hamazaki
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Paul Pavlidis
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada;
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
3
|
Sajadi M, Fazilti M, Nazem H, Mahdevar M, Ghaedi K. The expression changes of transcription factors including ANKZF1, LEF1, CASZ1, and ATOH1 as a predictor of survival rate in colorectal cancer: a large-scale analysis. Cancer Cell Int 2022; 22:339. [PMID: 36344988 PMCID: PMC9641931 DOI: 10.1186/s12935-022-02751-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION Transcription factors (TFs) are essential for many biological processes and regulate the expression of several genes. This study's objective was to analyze the abnormalities in TF expression, their impact on patient prognosis, and related pathways in colorectal cancer (CRC). METHOD The expression alterations of all TFs were investigated using the cancer genome atlas and GSE39582 data. Clinical data were also used to study the association between TFs expression and patient prognosis through the Cox regression test, and a predictive model of CRC patient survival was constructed based on TFs expression. Co-expression network was used to discover TF-related pathways. To validate the findings, the RT-qPCR method was applied to CRC samples and adjacent normal tissue. RESULTS The findings revealed that ANKZF1, SALL4, SNAI1, TIGD1, LEF1, FOXS1, SIX4, and ETV5 expression levels increased in both cohorts and were linked to the poor prognosis. NR3C2, KLF4, CASZ1, FOXD2, ATOH1, SALL1, and RORC expression, on the other hand, exhibited a significant decrease, and their increase was related to the good prognosis of patients. The patient mortality risk model based on expression of mentioned TFs revealed that, independent of clinical characteristics, the expression of ANKZF1, LEF1, CASZ1, and ATOH1 could accurately predict patient survival rates. According to the co-expression network, increased transcription factors were linked to metastatic pathways, while decreasing TFs were involved to apoptotic pathways. RT-qPCR findings showed that FOXS1 expression was markedly overexpressed in CRC samples. However, in CRC samples, the expression of CASZ1 decreased. CONCLUSION In CRC, TFs expression of ANKZF1, LEF1, CASZ1 and ATOH1 are deregulated, which are associated with prognosis in patients. According to our findings, changes in the expression of the mentioned TFs have the potential to be considered diagnostic and prognostic biomarkers for CRC patients.
Collapse
Affiliation(s)
- Manizheh Sajadi
- Department of Biochemistry, Faculty of Science, Payame Noor University, Isfahan, Iran
| | - Mohammad Fazilti
- Department of Biochemistry, Payame Noor University, P.O.Box 19395-4697, Tehran, Iran.
| | - Habibollah Nazem
- Department of Biochemistry, Faculty of Science, Payame Noor University, Isfahan, Iran
| | | | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
4
|
Elzinga J, van der Lugt B, Belzer C, Steegenga WT. Characterization of increased mucus production of HT29-MTX-E12 cells grown under Semi-Wet interface with Mechanical Stimulation. PLoS One 2021; 16:e0261191. [PMID: 34928974 PMCID: PMC8687553 DOI: 10.1371/journal.pone.0261191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 11/24/2021] [Indexed: 12/29/2022] Open
Abstract
The intestinal mucus layer plays a crucial role in human health. To study intestinal mucus function and structure in vitro, the mucus-producing intestinal cell line HT29-MTX-E12 has been commonly used. However, this cell line produces only low amounts of the intestine-specific MUC2. It has been shown previously that HT29-MTX-E12 cells cultured under Semi-Wet interface with Mechanical Stimulation (SWMS) produced higher amounts of MUC2, concomitant with a thicker mucus layer, compared to cells cultured conventionally. However, it remains unknown which underlying pathways are involved. Therefore, we aimed to further explore the cellular processes underlying the increased MUC2 production by HT29-MTX-E12 cells grown under SWMS conditions. Cells grown on Transwell membranes for 14 days under static and SWMS conditions (after cell seeding and attachment) were subjected to transcriptome analysis to investigate underlying molecular pathways at gene expression level. Caco-2 and LS174T cell lines were included as references. We characterized how SWMS conditions affected HT29-MTX-E12 cells in terms of epithelial barrier integrity, by measuring transepithelial electrical resistance, and cell metabolism, by monitoring pH and lactate production per molecule glucose of the conditioned medium. We confirmed higher MUC2 production under SWMS conditions at gene and protein level and demonstrated that this culturing method primarily stimulated cell growth. In addition, we also found evidence for a more aerobic cell metabolism under SWMS, as shown previously for similar models. In summary, we suggest different mechanisms by which MUC2 production is enhanced under SWMS and propose potential applications of this model in future studies.
Collapse
Affiliation(s)
- Janneke Elzinga
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Benthe van der Lugt
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Wilma T Steegenga
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
5
|
Sun J, Xu Z, Mao Y, Zhang T, Qin Y, Hua D. Prognostic role of oxytocin receptor in colon adenocarcinoma. Open Med (Wars) 2021; 16:1762-1776. [PMID: 34877409 PMCID: PMC8610102 DOI: 10.1515/med-2021-0387] [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: 04/13/2021] [Revised: 09/14/2021] [Accepted: 10/13/2021] [Indexed: 11/15/2022] Open
Abstract
The oxytocin receptor (OXTR) is directly involved in the pathological mechanisms of multiple cancers, including breast cancer, prostate cancer, and ovarian cancer; however, the role of OXTR in the modulation of colon adenocarcinoma (COAD) growth, metastasis, and clinical prognosis remains to be elucidated. This study used systematic bioinformatics analysis to explore the effects of OXTR on modulating COAD growth and prognosis in patients with COAD. Compared with normal tissues, OXTR mRNA level was higher in COAD tissues, which was associated with tumor progression. Elevated mRNA level of OXTR also indicated a poor prognosis in COAD patients. Furthermore, high mRNA level of OXTR was significantly associated with pathways involved in cell cycle regulation and signal transduction pathways, including the hedgehog, mTOR, TGF-β, and Wnt signaling pathways. OXTR expression was significantly correlated with the infiltration level of type 2T helper cell, central memory CD8 T cell, CD56 bright natural killer cell, activated CD8 T cell, activated B cell, and Type 1T helper cell. Moreover, silencing OXTR inhibited cell proliferation, migration, and invasion, and arrested the cell cycle. In conclusion, high mRNA level of OXTR indicates poor prognosis.
Collapse
Affiliation(s)
- Junjie Sun
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, P. R. China.,Department of Oncology, The Affiliated Hospital of Jiangnan University, No. 200, Huihe Road, Wuxi, 214000, Jiangsu, P. R. China
| | - Zhenyu Xu
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, P. R. China.,Department of Oncology, The Affiliated Hospital of Jiangnan University, No. 200, Huihe Road, Wuxi, 214000, Jiangsu, P. R. China
| | - Yong Mao
- Department of Oncology, The Affiliated Hospital of Jiangnan University, No. 200, Huihe Road, Wuxi, 214000, Jiangsu, P. R. China
| | - Ting Zhang
- Department of Oncology, The Affiliated Hospital of Jiangnan University, No. 200, Huihe Road, Wuxi, 214000, Jiangsu, P. R. China
| | - Yan Qin
- Department of Pathology, The Affiliated Hospital of Jiangnan University, No. 200, Huihe Road, Wuxi, 214000, Jiangsu, P. R. China
| | - Dong Hua
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, P. R. China.,Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214000, Jiangsu, P. R. China
| |
Collapse
|
6
|
Helm ET, Burrough ER, Leite FL, Gabler NK. Lawsonia intracellularis infected enterocytes lack sucrase-isomaltase which contributes to reduced pig digestive capacity. Vet Res 2021; 52:90. [PMID: 34147126 PMCID: PMC8214296 DOI: 10.1186/s13567-021-00958-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Lawsonia intracellularis is endemic to swine herds worldwide, however much is still unknown regarding its impact on intestinal function. Thus, this study aimed to characterize the impact of L. intracellularis on digestive function, and how vaccination mitigates these impacts. Thirty-six L. intracellularis negative barrows were assigned to treatment groups (n = 12/trt): (1) nonvaccinated, L. intracellularis negative (NC); (2) nonvaccinated, L intracellularis challenged (PC); and (3) L. intracellularis challenged, vaccinated (Enterisol® Ileitis, Boehringer Ingelheim) 7 weeks pre-challenge (VAC). On days post-inoculation (dpi) 0 PC and VAC pigs were inoculated with L. intracellularis. From dpi 19–21 fecal samples were collected for apparent total tract digestibility (ATTD) and at dpi 21, pigs were euthanized for sample collection. Post-inoculation, ADG was reduced in PC pigs compared with NC (41%, P < 0.001) and VAC (25%, P < 0.001) pigs. Ileal gross lesion severity was greater in PC pigs compared with NC (P = 0.003) and VAC (P = 0.018) pigs. Dry matter, organic matter, nitrogen, and energy ATTD were reduced in PC pigs compared with NC pigs (P ≤ 0.001 for all). RNAscope in situ hybridization revealed abolition of sucrase-isomaltase transcript in the ileum of PC pigs compared with NC and VAC pigs (P < 0.01). Conversely, abundance of stem cell signaling markers Wnt3, Hes1, and p27Kip1 were increased in PC pigs compared with NC pigs (P ≤ 0.085). Taken together, these data demonstrate that reduced digestibility during L. intracellularis challenge is partially driven by abolition of digestive machinery in lesioned tissue. Further, vaccination mitigated several of these effects, likely from lower bacterial burden and reduced disease severity.
Collapse
Affiliation(s)
- Emma T Helm
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Eric R Burrough
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, 50011, USA
| | - Fernando L Leite
- Boehringer Ingelheim Animal Health USA Inc, Duluth, GA, 30096, USA
| | - Nicholas K Gabler
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
7
|
Plotnikov A, Kozer N, Cohen G, Carvalho S, Duberstein S, Almog O, Solmesky LJ, Shurrush KA, Babaev I, Benjamin S, Gilad S, Kupervaser M, Levin Y, Gershovits M, Ben-Avraham D, Barr HM. PRMT1 inhibition induces differentiation of colon cancer cells. Sci Rep 2020; 10:20030. [PMID: 33208761 PMCID: PMC7676271 DOI: 10.1038/s41598-020-77028-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
Differentiation therapy has been recently revisited as a prospective approach in cancer therapy by targeting the aberrant growth, and repairing the differentiation and cell death programs of cancer cells. However, differentiation therapy of solid tumors is a challenging issue and progress in this field is limited. We performed High Throughput Screening (HTS) using a novel dual multiplex assay to discover compounds, which induce differentiation of human colon cancer cells. Here we show that the protein arginine methyl transferase (PRMT) type 1 inhibitor, MS023, is a potent inducer of colon cancer cell differentiation with a large therapeutic window. Differentiation changes in the highly aggressive human colon cancer cell line (HT-29) were proved by proteomic and genomic approaches. Growth of HT-29 xenograft in nude mice was significantly delayed upon MS023 treatment and immunohistochemistry of tumor indicated differentiation changes. These findings may lead to development of clinically effective anti-cancer drugs based on the mechanism of cancer cell differentiation.
Collapse
Affiliation(s)
- Alexander Plotnikov
- Wohl Institute for Drug Discovery, High Throughput Screening Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel.
| | - Noga Kozer
- Wohl Institute for Drug Discovery, High Throughput Screening Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Galit Cohen
- Wohl Institute for Drug Discovery, High Throughput Screening Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Silvia Carvalho
- Wohl Institute for Drug Discovery, High Throughput Screening Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Shirly Duberstein
- Wohl Institute for Drug Discovery, High Throughput Screening Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Ofir Almog
- Wohl Institute for Drug Discovery, High Throughput Screening Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Leonardo Javier Solmesky
- Wohl Institute for Drug Discovery, High Throughput Screening Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Khriesto A Shurrush
- Wohl Institute for Drug Discovery, Medicinal Chemistry Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Ilana Babaev
- Wohl Institute for Drug Discovery, Medicinal Chemistry Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Sima Benjamin
- Crown Institute for Genomics, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Shlomit Gilad
- Crown Institute for Genomics, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Meital Kupervaser
- de Botton Institute for Proteomics, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- de Botton Institute for Proteomics, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Gershovits
- Mantoux Institute for Bioinformatics, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Danny Ben-Avraham
- Mantoux Institute for Bioinformatics, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Haim Michael Barr
- Wohl Institute for Drug Discovery, High Throughput Screening Unit, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
8
|
Abstract
Head and neck cancer is a group of neoplastic diseases affecting the facial, oral, and neck region. It is one of the most common cancers worldwide with an aggressive, invasive evolution. Due to the heterogeneity of the tissues affected, it is particularly challenging to study the molecular mechanisms at the basis of these tumors, and to date we are still lacking accurate targets for prevention and therapy. The Notch signaling is involved in a variety of tumorigenic mechanisms, such as regulation of the tumor microenvironment, aberrant intercellular communication, and altered metabolism. Here, we provide an up-to-date review of the role of Notch in head and neck cancer and draw parallels with other types of solid tumors where the Notch pathway plays a crucial role in emergence, maintenance, and progression of the disease. We therefore give a perspective view on the importance of the pathway in neoplastic development in order to define future lines of research and novel therapeutic approaches.
Collapse
|
9
|
Improved detection of tumor suppressor events in single-cell RNA-Seq data. NPJ Genom Med 2020; 5:43. [PMID: 33083012 PMCID: PMC7541488 DOI: 10.1038/s41525-020-00151-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
Tissue-specific transcription factors are frequently inactivated in cancer. To fully dissect the heterogeneity of such tumor suppressor events requires single-cell resolution, yet this is challenging because of the high dropout rate. Here we propose a simple yet effective computational strategy called SCIRA to infer regulatory activity of tissue-specific transcription factors at single-cell resolution and use this tool to identify tumor suppressor events in single-cell RNA-Seq cancer studies. We demonstrate that tissue-specific transcription factors are preferentially inactivated in the corresponding cancer cells, suggesting that these are driver events. For many known or suspected tumor suppressors, SCIRA predicts inactivation in single cancer cells where differential expression does not, indicating that SCIRA improves the sensitivity to detect changes in regulatory activity. We identify NKX2-1 and TBX4 inactivation as early tumor suppressor events in normal non-ciliated lung epithelial cells from smokers. In summary, SCIRA can help chart the heterogeneity of tumor suppressor events at single-cell resolution.
Collapse
|
10
|
Seetha A, Devaraj H, Sudhandiran G. Indomethacin and juglone inhibit inflammatory molecules to induce apoptosis in colon cancer cells. J Biochem Mol Toxicol 2020; 34:e22433. [PMID: 31916655 DOI: 10.1002/jbt.22433] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/13/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is the third most common fatal cancer. Indomethacin, a nonsteroidal anti-inflammatory drug, is known to reduce the occurrence of CRC. This study evaluated the potential anticolon cancer effects of juglone (5-hydroxy-1,4-naphthoquinone) in combination with indomethacin. Human colon adenocarcinoma cells (HT29) were subjected to treatment with indomethacin, juglone, and a combination of both. Morphological analysis, cell cycle regulation, and dual staining using acridine orange and ethidium bromide in control and treated cells revealed the apoptotic potential of these compounds. Bcl2 and inflammatory molecules (tumor necrosis factor-α, nuclear factor kappa B, and Cox-2) were found to be decreased with a concomitant increase in the expression of proapoptotic molecules (Bad, Bax, cytochrome c, and PUMA) as a result of the molecular regulation of Wnt, Notch, and peroxisome proliferator-activated receptor-γ signaling. Treatment with juglone was not as effective as with indomethacin; however, a combination of both was shown to be more effective, suggesting that juglone may be considered for therapeutic intervention of colon cancer.
Collapse
Affiliation(s)
- Alagesan Seetha
- Department of Biochemistry, University of Madras, Guindy Campus, Chennai, India
| | - Halagowder Devaraj
- Department of Zoology, University of Madras, Guindy Campus, Chennai, India
| | | |
Collapse
|
11
|
Das B, Okamoto K, Rabalais J, Kozan PA, Marchelletta RR, McGeough MD, Durali N, Go M, Barrett KE, Das S, Sivagnanam M. Enteroids expressing a disease-associated mutant of EpCAM are a model for congenital tufting enteropathy. Am J Physiol Gastrointest Liver Physiol 2019; 317:G580-G591. [PMID: 31433211 PMCID: PMC6879886 DOI: 10.1152/ajpgi.00098.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Congenital tufting enteropathy (CTE) is an autosomal recessive disease characterized by severe intestinal failure in infancy and mutations in the epithelial cell adhesion molecule (EPCAM) gene. Previous studies of CTE in mice expressing mutant EpCAM show neonatal lethality. Hence, to study the cellular, molecular, and physiological alterations that result from EpCAM mutation, a tamoxifen-inducible mutant EpCAM enteroid model has been generated. The presence of mutant EpCAM in the model was confirmed at both mRNA and protein levels. Immunofluorescence microscopy demonstrated the reduced expression of mutant EpCAM. Mutant enteroids had reduced budding potential as well as significantly decreased mRNA expression for epithelial lineage markers (Mucin 2, lysozyme, sucrase-isomaltase), proliferation marker Ki67, and secretory pathway transcription factors (Atoh1, Hnf1b). Significantly decreased numbers of Paneth and goblet cells were confirmed by staining. These findings were correlated with intestinal tissue from CTE patients and the mutant mice model that had significantly fewer Paneth and goblet cells than in healthy counterparts. FITC-dextran studies demonstrated significantly impaired barrier function in monolayers derived from mutant enteroids compared with control monolayers. In conclusion, we have established an ex vivo CTE model. The role of EpCAM in the budding potential, differentiation, and barrier function of enteroids is noted. Our study establishes new facets of EpCAM biology that will aid in understanding the pathophysiology of CTE and role of EpCAM in health and disease.NEW & NOTEWORTHY Here, we develop a novel ex vivo enteroid model for congenital tufting enteropathy (CTE) based on epithelial cell adhesion molecule (EPCAM) gene mutations found in patients. With this model we demonstrate the role of EpCAM in maintaining the functional homeostasis of the intestinal epithelium, including differentiation, proliferation, and barrier integrity. This study further establishes a new direction in EpCAM biology that will help in understanding the detailed pathophysiology of CTE and role of EpCAM.
Collapse
Affiliation(s)
- Barun Das
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kevin Okamoto
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Rabalais
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Philip A. Kozan
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | | | - Matthew D. McGeough
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Nassim Durali
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Maria Go
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kim E. Barrett
- 2Department of Medicine, University of California, San Diego, La Jolla, California
| | - Soumita Das
- 3Department of Pathology, University of California, San Diego, La Jolla, California
| | - Mamata Sivagnanam
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California,4Rady Children’s Hospital, San Diego, California
| |
Collapse
|
12
|
Ballweg R, Lee S, Han X, Maini PK, Byrne H, Hong CI, Zhang T. Unraveling the Control of Cell Cycle Periods during Intestinal Stem Cell Differentiation. Biophys J 2018; 115:2250-2258. [PMID: 30467024 DOI: 10.1016/j.bpj.2018.10.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 10/01/2018] [Accepted: 10/22/2018] [Indexed: 01/20/2023] Open
Abstract
During differentiation, intestinal stem cells (ISCs), a prototypical adult stem cell pool, become either secretory transit-amplifying cells, which give rise to all secretory cell types, or absorptive transit-amplifying cells, which give rise to enterocytes. These cells exhibit distinct cell cycle dynamics: ISCs cycle with a period of 24 h and absorptive transit-amplifying cells cycle with a period of ∼12 h, whereas secretory transit-amplifying cells arrest their cycle. The cell cycle dynamics of ISCs and their progeny are a systems-level property that emerges from interactions between the cell cycle control machinery and multiple regulatory pathways. Although many mathematical models have been developed to study the details of the cell cycle and related regulatory pathways, few models have been constructed to unravel the dynamic consequences of their interactions. To fill this gap, we present a simplified model focusing on the interaction between four key regulatory pathways (STAT, Wnt, Notch, and MAPK) and cell cycle control. After experimentally validating a model prediction, which showed that the Notch pathway can fine-tune the cell cycle period, we perform further model analysis that reveals that the change of cell cycle period accompanying ISC differentiation may be controlled by a design principle that has been well studied in dynamical systems theory-a saddle node on invariant circle bifurcation. Given that the mechanisms that control the cell cycle are conserved in most eukaryotic cell types, this general principle potentially controls the interplay between proliferation and differentiation for a broad range of stem cells.
Collapse
Affiliation(s)
- Richard Ballweg
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Suengwon Lee
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Xiaonan Han
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital, Cincinnati, Ohio; Key Laboratory of Human Disease Comparative Medicine, Health Ministry, Beijing, China; Institute of Laboratory Animal Science, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Helen Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Christian I Hong
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, Ohio; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Tongli Zhang
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
13
|
Dunkin D, Iuga AC, Mimouna S, Harris CL, Haure-Mirande JV, Bozec D, Yeretssian G, Dahan S. Intestinal epithelial Notch-1 protects from colorectal mucinous adenocarcinoma. Oncotarget 2018; 9:33536-33548. [PMID: 30323897 PMCID: PMC6173356 DOI: 10.18632/oncotarget.26086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 08/23/2018] [Indexed: 12/30/2022] Open
Abstract
Increasing evidence links Notch-1 signaling with the maintenance of intestinal architecture and homeostasis. Dysfunction in the common Notch-1 pathway transcription factor recombinant binding protein suppressor of hairless (RBP-J) is associated with loss of epithelial barrier integrity and aberrant conversion of proliferative crypt cells into goblet cells. Furthermore, we have recently discovered that epithelial Notch-1 is indispensable in bridging innate and adaptive immunity in the gut and is required for supporting protective epithelial pro-inflammatory responses. Yet, the epithelial specific function of Notch-1 in intestinal tumorigenesis remains unknown. We generated Villin-Cre/Notch-1fl/fl (VN-/-) mice that are selectively deficient in Notch-1 in intestinal epithelial cells. Intestinal epithelial Notch-1 preserved barrier function and integrity, whereas lack of epithelial Notch-1 induced goblet cell hyperplasia, spontaneous serrated lesions, multifocal low- and high-grade dysplasia and colonic mucinous neoplasms in mice. Over time, VN-/- mice displayed high occurrence of colorectal mucinous adenocarcinomas, which correlated with increased levels of mitogenic, angiogenic and pro-tumorigenic gene expression. Finally, we found that the expression of Notch-1 is significantly reduced in human colorectal mucinous adenocarcinoma when compared to colorectal adenocarcinoma. Taken together, our findings reveal a novel and critical protective role for Notch-1 in controlling intestinal tumorigenesis.
Collapse
Affiliation(s)
- David Dunkin
- Department of Pediatric Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alina C Iuga
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Sanda Mimouna
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Immunology and Autoimmunity Research Department, Hospital for Special Surgery Research Institute, New York, NY 10021, USA.,Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carolyn L Harris
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jean-Vianney Haure-Mirande
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dominique Bozec
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Garabet Yeretssian
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Leona M. and Harry B. Helmsley Charitable Trust, New York, NY 10169, USA
| | - Stephanie Dahan
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Sobi, Inc. Waltham, MA 02452, USA
| |
Collapse
|
14
|
Baliou S, Adamaki M, Kyriakopoulos AM, Spandidos DA, Panayiotidis M, Christodoulou I, Zoumpourlis V. CRISPR therapeutic tools for complex genetic disorders and cancer (Review). Int J Oncol 2018; 53:443-468. [PMID: 29901119 PMCID: PMC6017271 DOI: 10.3892/ijo.2018.4434] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
One of the fundamental discoveries in the field of biology is the ability to modulate the genome and to monitor the functional outputs derived from genomic alterations. In order to unravel new therapeutic options, scientists had initially focused on inducing genetic alterations in primary cells, in established cancer cell lines and mouse models using either RNA interference or cDNA overexpression or various programmable nucleases [zinc finger nucleases (ZNF), transcription activator-like effector nucleases (TALEN)]. Even though a huge volume of data was produced, its use was neither cheap nor accurate. Therefore, the clustered regularly interspaced short palindromic repeats (CRISPR) system was evidenced to be the next step in genome engineering tools. CRISPR-associated protein 9 (Cas9)-mediated genetic perturbation is simple, precise and highly efficient, empowering researchers to apply this method to immortalized cancerous cell lines, primary cells derived from mouse and human origins, xenografts, induced pluripotent stem cells, organoid cultures, as well as the generation of genetically engineered animal models. In this review, we assess the development of the CRISPR system and its therapeutic applications to a wide range of complex diseases (particularly distinct tumors), aiming at personalized therapy. Special emphasis is given to organoids and CRISPR screens in the design of innovative therapeutic approaches. Overall, the CRISPR system is regarded as an eminent genome engineering tool in therapeutics. We envision a new era in cancer biology during which the CRISPR-based genome engineering toolbox will serve as the fundamental conduit between the bench and the bedside; nonetheless, certain obstacles need to be addressed, such as the eradication of side-effects, maximization of efficiency, the assurance of delivery and the elimination of immunogenicity.
Collapse
Affiliation(s)
- Stella Baliou
- National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Adamaki
- National Hellenic Research Foundation, 11635 Athens, Greece
| | | | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion 71003, Greece
| | - Mihalis Panayiotidis
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, NE1 8ST, UK
| | | | | |
Collapse
|
15
|
Bigas A, Porcheri C. Notch and Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:235-263. [DOI: 10.1007/978-3-319-89512-3_12] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Constitutive Immune Activity Promotes Tumorigenesis in Drosophila Intestinal Progenitor Cells. Cell Rep 2017; 20:1784-1793. [DOI: 10.1016/j.celrep.2017.07.078] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 06/05/2017] [Accepted: 07/27/2017] [Indexed: 02/02/2023] Open
|
17
|
Badenes M, Trindade A, Pissarra H, Lopes-da-Costa L, Duarte A. Delta-like 4/Notch signaling promotes Apc Min/+ tumor initiation through angiogenic and non-angiogenic related mechanisms. BMC Cancer 2017; 17:50. [PMID: 28086833 PMCID: PMC5237288 DOI: 10.1186/s12885-016-3036-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 12/27/2016] [Indexed: 01/27/2023] Open
Abstract
Background Delta like 4 (Dll4)/Notch signaling is a key regulator of tumor angiogenesis. Additionally, the role of Dll4 has been studied on tumor stem cells. However, as these cells are implicated in tumor angiogenesis, it is conceivable that the effect of Dll4 on these cells may be a consequence of its angiogenic function. Our aim was to evaluate the expression and dissect the functions of Dll4 in the ApcMin/+ model of colorectal cancer. Methods We evaluated the protein expression pattern of Dll4 and other Notch members in the ApcMin/+ tumors relatively to the normal gut and compared endothelial-specific with ubiquitous Dll4 knockout mice on an ApcMin/+ background. Results All Notch pathway members were present in the normal small and large intestine and in the adenomas of the same regions. Dll4, all Notch receptors and Hes1 expression seemed upregulated in the tumors, with some regional differences. The same members and Hes5, instead of Hes1, presented ectopic expression in the tumor parenchyma. Dll4 expression was most pronounced in the tumor cells but it was also present in the tumor blood vessels and in other stromal cells. Ubiquitous and endothelial-specific Dll4 deletion led to an equivalent reduction of tumor growth because of a similarly marked tumoral angiogenic phenotype promoting non-productive vasculature and consequently hypoxia and apoptosis. The ubiquitous Dll4 inhibition led to a stronger decrease of tumor multiplicity than the endothelial-specific deletion by further reducing tumor proliferation and tumor stem cell density through upregulation of the cyclin-dependent kinase inhibitors 1C and 1B and downregulation of Myc, Cyclin D1 and D2 independently of β-catenin activation. This phenotype was associated to the observed increased epithelial differentiation deviated towards the secretory lineages by Atoh1 and Klf4 upregulation only in the ubiquitous Dll4 mutants. Conclusions Dll4 seems to promote ApcMin/+ tumorigenesis through both angiogenic and non-angiogenic related mechanisms. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-3036-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marina Badenes
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Alexandre Trindade
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Hugo Pissarra
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Luís Lopes-da-Costa
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - António Duarte
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
18
|
Liu F, Huang J, Ning B, Liu Z, Chen S, Zhao W. Drug Discovery via Human-Derived Stem Cell Organoids. Front Pharmacol 2016; 7:334. [PMID: 27713700 PMCID: PMC5032635 DOI: 10.3389/fphar.2016.00334] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/09/2016] [Indexed: 12/18/2022] Open
Abstract
Patient-derived cell lines and animal models have proven invaluable for the understanding of human intestinal diseases and for drug development although both inherently comprise disadvantages and caveats. Many genetically determined intestinal diseases occur in specific tissue microenvironments that are not adequately modeled by monolayer cell culture. Likewise, animal models incompletely recapitulate the complex pathologies of intestinal diseases of humans and fall short in predicting the effects of candidate drugs. Patient-derived stem cell organoids are new and effective models for the development of novel targeted therapies. With the use of intestinal organoids from patients with inherited diseases, the potency and toxicity of drug candidates can be evaluated better. Moreover, owing to the novel clustered regularly interspaced short palindromic repeats/CRISPR-associated protein-9 genome-editing technologies, researchers can use organoids to precisely modulate human genetic status and identify pathogenesis-related genes of intestinal diseases. Therefore, here we discuss how patient-derived organoids should be grown and how advanced genome-editing tools may be applied to research on modeling of cancer and infectious diseases. We also highlight practical applications of organoids ranging from basic studies to drug screening and precision medicine.
Collapse
Affiliation(s)
- Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South UniversityChangsha, China; Center for Inflammation and Epigenetics, Houston Methodist Research Institute, HoustonTX, USA
| | - Jing Huang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, HoustonTX, USA; Department of Psychiatry, The Second Xiangya Hospital, Central South University, ChangshaHunan, China; Mental Health Institute of the Second Xiangya Hospital, Central South University, ChangshaHunan, China; Chinese National Clinical Research Center on Mental Disorders, ChangshaHunan, China; Chinese National Technology Institute on Mental Disorders, ChangshaHunan, China; Hunan Key Laboratory of Psychiatry and Mental Health, ChangshaHunan, China
| | - Bo Ning
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston TX, USA
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University Changsha, China
| | - Shen Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Wei Zhao
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
19
|
Prognostic relevance of high atonal homolog-1 expression in Merkel cell carcinoma. J Cancer Res Clin Oncol 2016; 143:43-49. [PMID: 27624714 DOI: 10.1007/s00432-016-2257-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND It has recently been reported that atonal homolog 1 (ATOH1) gene is down-regulated in Merkel cell carcinoma (MCC) and thus may represent a tumor suppressor gene. OBJECTIVES We aimed to test for ATOH1 gene mutations and expression levels in MCC tissues and cell lines. METHODS Genomic DNA isolation and amplification via PCR was successfully performed in 33 MCCs on formalin-fixed paraffin-embedded tissue and three MCC cell lines, followed by Sanger sequencing of the whole ATOH1 gene to detect genomic aberrations. ATOH1 mRNA levels were determined by RT-PCR. Immunohistochemistry of ATOH1 was performed to quantify protein expression in tumor samples and cell lines. RESULTS Neither in any of the 33 MCC tissue samples nor in the three cell lines ATOH1 mutations were present. ATOH1 was expressed in all lesions, albeit at different expression levels. Univariate analysis revealed that the total immunohistology score significantly correlated with the occurrence of tumor relapse (r = 0.57; P = 0.0008). This notion was confirmed in multivariate analysis suggesting that ATOH1 expression is a potential independent predictor for tumor relapse in MCC patients (P = 0.028). MCC-related death also correlated with ATOH1 expression (r = 0.4; P = 0.025); however, ATOH1 expression did not retain its predictive value in the regression model. CONCLUSIONS In contrast to anecdotal reports ATOH1 expression is not lost by genetic alterations in MCC. However, protein expression of ATOH1 is increased in advanced MCC indicating that ATOH1 is involved in MCC progression.
Collapse
|
20
|
MUC4 is negatively regulated through the Wnt/β-catenin pathway via the Notch effector Hath1 in colorectal cancer. Genes Cancer 2016; 7:154-168. [PMID: 27551331 PMCID: PMC4979589 DOI: 10.18632/genesandcancer.108] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
MUC4 is a transmembrane mucin lining the normal colonic epithelium. The aberrant/de novo over-expression of MUC4 is well documented in malignancies of the pancreas, ovary and breast. However, studies have reported the loss of MUC4 expression in the majority of colorectal cancers (CRCs). A MUC4 promoter analysis showed the presence of three putative TCF/LEF sites, implying a possible regulation by the Wnt/β-catenin pathway, which has been shown to drive CRC progression. Thus, the objective of our study was to determine whether MUC4 is regulated by β-catenin in CRC. We first knocked down (KD) β-catenin in three CRC cell lines; LS180, HCT-8 and HCT116, which resulted in increased MUC4 transcript and MUC4 protein. Additionally, the overexpression of stabilized mutant β-catenin in LS180 and HCT-8 resulted in a decrease in MUC4 expression. Immunohistochemistry (IHC) of mouse colon tissue harboring tubular adenomas and high grade dysplasia showed dramatically reduced Muc4 in lesions relative to adjacent normal tissue, with increased cytosolic/nuclear β-catenin. Luciferase assays with the complete MUC4 promoter construct p3778 showed increased MUC4 promoter luciferase activity in the absence of β-catenin (KD). Mutation of all three putative TCF/LEF sites showed that MUC4 promoter luciferase activity was increased relative to the un-mutated promoter. Interestingly, it was observed that MUC4 expressing CRC cell lines also expressed high levels of Hath1, a transcription factor repressed by both active Wnt/β-catenin and Notch signaling. The KD of β-catenin and/or treatment with a Notch γ-secretase inhibitor, Dibenzazepine (DBZ) resulted in increased Hath1 and MUC4 in LS180, HCT-8 and HCT116. Furthermore, overexpression of Hath1 in HCT-8 and LS180 caused increased MUC4 transcript and MUC4 protein. Taken together, our results indicate that the Wnt/β-catenin pathway suppresses the Notch pathway effector Hath1, resulting in reduced MUC4 in CRC.
Collapse
|
21
|
Demitrack ES, Samuelson LC. Notch regulation of gastrointestinal stem cells. J Physiol 2016; 594:4791-803. [PMID: 26848053 DOI: 10.1113/jp271667] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/19/2016] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract epithelium is continuously replenished by actively cycling stem and progenitor cells. These cell compartments are regulated to balance proliferation and stem cell renewal with differentiation into the various mature cell types to maintain tissue homeostasis. In this topical review we focus on the role of the Notch signalling pathway to regulate GI stem cell function in adult small intestine and stomach. We first present the current view of stem and progenitor cell populations in these tissues and then summarize the studies that have established the Notch pathway as a key regulator of gastric and intestinal stem cell function. Notch signalling has been shown to be a niche factor required for maintenance of GI stem cells in both tissues. In addition, Notch has been described to regulate epithelial cell differentiation. Recent studies have revealed key similarities and differences in how Notch regulates stem cell function in the stomach compared to intestine. We summarize the literature regarding Notch regulation of GI stem cell proliferation and differentiation, highlighting tissue-specific functions to compare and contrast Notch in the stomach and intestine.
Collapse
Affiliation(s)
- Elise S Demitrack
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Linda C Samuelson
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| |
Collapse
|
22
|
Weng MT, Tsao PN, Lin HL, Tung CC, Change MC, Chang YT, Wong JM, Wei SC. Hes1 Increases the Invasion Ability of Colorectal Cancer Cells via the STAT3-MMP14 Pathway. PLoS One 2015; 10:e0144322. [PMID: 26650241 PMCID: PMC4674118 DOI: 10.1371/journal.pone.0144322] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/15/2015] [Indexed: 12/21/2022] Open
Abstract
The Notch pathway contributes to self-renewal of tumor-initiating cell and inhibition of normal colonic epithelial cell differentiation. Deregulated expression of Notch1 and Jagged1 is observed in colorectal cancer. Hairy/enhancer of split (HES) family, the most characterized targets of Notch, involved in the development of many cancers. In this study, we explored the role of Hes1 in the tumorigenesis of colorectal cancer. Knocking down Hes1 induced CRC cell senescence and decreased the invasion ability, whereas over-expression of Hes1 increased STAT3 phosphorylation activity and up-regulated MMP14 protein level. We further explored the expression of Hes1 in human colorectal cancer and found high Hes1 mRNA expression is associated with poor prognosis in CRC patients. These findings suggest that Hes1 regulates the invasion ability through the STAT3-MMP14 pathway in CRC cells and high Hes1 expression is a predictor of poor prognosis of CRC.
Collapse
Affiliation(s)
- MT Weng
- Department of Internal Medicine, Far-Eastern Memorial Hospital, New Taipei, Taiwan
- Department of Chemical Engineering & Materials Science, Yuan-Ze University, Taoyuan, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - PN Tsao
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - HL Lin
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - CC Tung
- Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - MC Change
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - YT Chang
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - JM Wong
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - SC Wei
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
23
|
Luo Y, Jin H, Wen GR, Tuo BG. Advances in differentiation therapy of hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2015; 23:4665-4672. [DOI: 10.11569/wcjd.v23.i29.4665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignancy of the digestive system, which shows obvious disorders of differentiation. Currently, its pathogenesis is still elusive. Therefore, to figure out the mechanism of disturbed differentiation in HCC is the key to finding out precise therapeutic targets for differentiation therapy. Differentiation therapy of hepatocellular carcinoma refers to making hepatocellular carcinoma cells differentiate into mature hepatocytes, and rebuilding their normal phenotype and normal biological function through promoting HCC cell apoptosis and inhibiting their proliferation in the presence of specific differentiation-inducing agents or signals. In recent years, increasing research results about HCC differentiation therapy have been reported. This article reviews differentiation-inducing agents or targets, differentiation-related signaling pathways and microRNAs, with an aim to provide some clues for HCC differentiation therapy.
Collapse
|
24
|
Schall KA, Holoyda KA, Grant CN, Levin DE, Torres ER, Maxwell A, Pollack HA, Moats RA, Frey MR, Darehzereshki A, Al Alam D, Lien C, Grikscheit TC. Adult zebrafish intestine resection: a novel model of short bowel syndrome, adaptation, and intestinal stem cell regeneration. Am J Physiol Gastrointest Liver Physiol 2015; 309:G135-45. [PMID: 26089336 PMCID: PMC4525108 DOI: 10.1152/ajpgi.00311.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 05/28/2015] [Indexed: 01/31/2023]
Abstract
Loss of significant intestinal length from congenital anomaly or disease may lead to short bowel syndrome (SBS); intestinal failure may be partially offset by a gain in epithelial surface area, termed adaptation. Current in vivo models of SBS are costly and technically challenging. Operative times and survival rates have slowed extension to transgenic models. We created a new reproducible in vivo model of SBS in zebrafish, a tractable vertebrate model, to facilitate investigation of the mechanisms of intestinal adaptation. Proximal intestinal diversion at segment 1 (S1, equivalent to jejunum) was performed in adult male zebrafish. SBS fish emptied distal intestinal contents via stoma as in the human disease. After 2 wk, S1 was dilated compared with controls and villus ridges had increased complexity, contributing to greater villus epithelial perimeter. The number of intervillus pockets, the intestinal stem cell zone of the zebrafish increased and contained a higher number of bromodeoxyuridine (BrdU)-labeled cells after 2 wk of SBS. Egf receptor and a subset of its ligands, also drivers of adaptation, were upregulated in SBS fish. Igf has been reported as a driver of intestinal adaptation in other animal models, and SBS fish exposed to a pharmacological inhibitor of the Igf receptor failed to demonstrate signs of intestinal adaptation, such as increased inner epithelial perimeter and BrdU incorporation. We describe a technically feasible model of human SBS in the zebrafish, a faster and less expensive tool to investigate intestinal stem cell plasticity as well as the mechanisms that drive intestinal adaptation.
Collapse
Affiliation(s)
- K. A. Schall
- 1Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - K. A. Holoyda
- 1Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - C. N. Grant
- 1Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - D. E. Levin
- 1Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - E. R. Torres
- 1Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - A. Maxwell
- 2Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - H. A. Pollack
- 3Department of Radiology, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - R. A. Moats
- 3Department of Radiology, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - M. R. Frey
- 2Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California; ,4Department of Pediatrics and Department of Biochemistry and Molecular Biology, Keck School of Medicine at University of Southern California, Los Angeles, California; and
| | - A. Darehzereshki
- 2Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - D. Al Alam
- 1Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| | - C. Lien
- 2Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California; ,5Department of Cardiothoracic Surgery, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California
| | - T. C. Grikscheit
- 1Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California; ,2Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine at University of Southern California, Los Angeles, California;
| |
Collapse
|
25
|
Cramer JM, Thompson T, Geskin A, LaFramboise W, Lagasse E. Distinct human stem cell populations in small and large intestine. PLoS One 2015; 10:e0118792. [PMID: 25751518 PMCID: PMC4353627 DOI: 10.1371/journal.pone.0118792] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 01/06/2015] [Indexed: 12/22/2022] Open
Abstract
The intestine is composed of an epithelial layer containing rapidly proliferating cells that mature into two regions, the small and the large intestine. Although previous studies have identified stem cells as the cell-of-origin for intestinal epithelial cells, no studies have directly compared stem cells derived from these anatomically distinct regions. Here, we examine intrinsic differences between primary epithelial cells isolated from human fetal small and large intestine, after in vitro expansion, using the Wnt agonist R-spondin 2. We utilized flow cytometry, fluorescence-activated cell sorting, gene expression analysis and a three-dimensional in vitro differentiation assay to characterize their stem cell properties. We identified stem cell markers that separate subpopulations of colony-forming cells in the small and large intestine and revealed important differences in differentiation, proliferation and disease pathways using gene expression analysis. Single cells from small and large intestine cultures formed organoids that reflect the distinct cellular hierarchy found in vivo and respond differently to identical exogenous cues. Our characterization identified numerous differences between small and large intestine epithelial stem cells suggesting possible connections to intestinal disease.
Collapse
Affiliation(s)
- Julie M. Cramer
- Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA, 15261, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, United States of America
| | - Timothy Thompson
- Department of Bioengineering, University of Pittsburgh, 3700 O'Hara St, Pittsburgh, PA, 15261, United States of America
| | - Albert Geskin
- Department of Pathology, University of Pittsburgh, Shadyside Hospital, West Wing, WG 02.11, 5230 Center Avenue, Pittsburgh, PA 15232, United States of America
| | - William LaFramboise
- Department of Pathology, University of Pittsburgh, Shadyside Hospital, West Wing, WG 02.11, 5230 Center Avenue, Pittsburgh, PA 15232, United States of America
| | - Eric Lagasse
- Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA, 15261, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, United States of America
- * E-mail:
| |
Collapse
|
26
|
Mulvaney JF, Amemiya Y, Freeman SD, Ladher RK, Dabdoub A. Molecular cloning and functional characterisation of chicken Atonal homologue 1: a comparison with human Atoh1. Biol Cell 2015; 107:41-60. [PMID: 25412697 DOI: 10.1111/boc.201400078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/20/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND INFORMATION The vertebrate basic helix-loop-helix transcription factor Atoh1 is essential for maturation and survival of mechanosensory hair cells of the inner ear, neurogenesis, differentiation of the intestine, homeostasis of the colon and is implicated in cancer progression. Given that mutations in Atoh1 are detected in malignant tumours, study of functionally different Atoh1 alleles and homologues might yield useful avenues for investigation. The predicted sequence of chicken Atoh1 (cAtoh1) has large regions of dissimilarity to that of mammalian Atoh1 homologues. We hypothesise that cAtoh1 might have intrinsic functional differences to mammalian Atoh1. RESULTS In this study, we cloned and sequenced the full open reading frame of cAtoh1. In overexpression experiments, we show that this sequence is sufficient to generate a cAtoh1 protein capable of inducing hair cell markers when expressed in nonsensory regions of the developing inner ear, and that morpholino-mediated knock-down using a section of the sequence 5' to the start codon inhibits differentiation of hair cells in the chicken basilar papilla. Furthermore, we compare the behaviour of cAtoh1 and human Atoh1 (hAtoh1) in embryonic mouse cochlear explants, showing that cAtoh1 is a potent inducer of hair cell differentiation and that it can overcome Sox2-mediated repression of hair cell differentiation more effectively than hAtoh1. CONCLUSIONS cAtoh1 is both necessary and sufficient for avian mechanosensory hair cell differentiation. The non-conserved regions of the cAtoh1 coding region have functional consequences on its behaviour.
Collapse
Affiliation(s)
- Joanna F Mulvaney
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, M4N 3M5, Canada
| | | | | | | | | |
Collapse
|
27
|
Philpott A, Winton DJ. Lineage selection and plasticity in the intestinal crypt. Curr Opin Cell Biol 2014; 31:39-45. [PMID: 25083805 PMCID: PMC4238899 DOI: 10.1016/j.ceb.2014.07.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 07/06/2014] [Accepted: 07/11/2014] [Indexed: 12/21/2022]
Abstract
We know more about the repertoire of cellular behaviours that define the stem and progenitor cells maintaining the intestinal epithelium than any other renewing tissue. Highly dynamic and stochastic processes define cell renewal. Historically the commitment step in differentiation is viewed as a ratchet, irreversibly promoting a given fate and corresponding to a programme imposed at the point of cell division. However, the emerging view of intestinal self-renewal is one of plasticity in which a stem cell state is easily reacquired. The pathway mediators of lineage selection are largely known but how they interface within highly dynamic populations to promote different lineages and yet permit plasticity is not. Advances in understanding gene regulation in the nervous system suggest possible mechanisms.
Collapse
Affiliation(s)
- Anna Philpott
- Department of Oncology, University of Cambridge, Hutchison/Medical Research Council (MRC) Research Centre, Cambridge CB2 0XZ, UK
| | - Douglas J Winton
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK.
| |
Collapse
|
28
|
Giammanco A, Blanc V, Montenegro G, Klos C, Xie Y, Kennedy S, Luo J, Chang SH, Hla T, Nalbantoglu I, Dharmarajan S, Davidson NO. Intestinal epithelial HuR modulates distinct pathways of proliferation and apoptosis and attenuates small intestinal and colonic tumor development. Cancer Res 2014; 74:5322-35. [PMID: 25085247 DOI: 10.1158/0008-5472.can-14-0726] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
HuR is a ubiquitous nucleocytoplasmic RNA-binding protein that exerts pleiotropic effects on cell growth and tumorigenesis. In this study, we explored the impact of conditional, tissue-specific genetic deletion of HuR on intestinal growth and tumorigenesis in mice. Mice lacking intestinal expression of HuR (Hur (IKO) mice) displayed reduced levels of cell proliferation in the small intestine and increased sensitivity to doxorubicin-induced acute intestinal injury, as evidenced by decreased villus height and a compensatory shift in proliferating cells. In the context of Apc(min/+) mice, a transgenic model of intestinal tumorigenesis, intestinal deletion of the HuR gene caused a three-fold decrease in tumor burden characterized by reduced proliferation, increased apoptosis, and decreased expression of transcripts encoding antiapoptotic HuR target RNAs. Similarly, Hur(IKO) mice subjected to an inflammatory colon carcinogenesis protocol [azoxymethane and dextran sodium sulfate (AOM-DSS) administration] exhibited a two-fold decrease in tumor burden. Hur(IKO) mice showed no change in ileal Asbt expression, fecal bile acid excretion, or enterohepatic pool size that might explain the phenotype. Moreover, none of the HuR targets identified in Apc(min/+)Hur(IKO) were altered in AOM-DSS-treated Hur(IKO) mice, the latter of which exhibited increased apoptosis of colonic epithelial cells, where elevation of a unique set of HuR-targeted proapoptotic factors was documented. Taken together, our results promote the concept of epithelial HuR as a contextual modifier of proapoptotic gene expression in intestinal cancers, acting independently of bile acid metabolism to promote cancer. In the small intestine, epithelial HuR promotes expression of prosurvival transcripts that support Wnt-dependent tumorigenesis, whereas in the large intestine epithelial HuR indirectly downregulates certain proapoptotic RNAs to attenuate colitis-associated cancer. Cancer Res; 74(18); 5322-35. ©2014 AACR.
Collapse
Affiliation(s)
- Antonina Giammanco
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Valerie Blanc
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Grace Montenegro
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | - Coen Klos
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | - Yan Xie
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Susan Kennedy
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Jianyang Luo
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Sung-Hee Chang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, New York
| | - Timothy Hla
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, New York
| | - Ilke Nalbantoglu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri
| | - Sekhar Dharmarajan
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | - Nicholas O Davidson
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri.
| |
Collapse
|
29
|
Abstract
Differentiation therapy for tumors refers to treating malignant tumors via the induction of cell differentiation. The best characterized clinical application of differentiation therapy is the use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia (APL), which markedly improved the outcome of this disease. Unlike the situation with APL, the development of differentiation therapy for solid tumors is far from satisfactory. To date, no differentiation-inducing agents have been demonstrated to exert a curative effect on solid tumors. However, over the past decade progress in understanding the differentiation pathways and the development of differentiation-inducing agents might shed new light on the differentiation therapy for solid tumors.
Collapse
Affiliation(s)
- Wen Ping Xu
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | | | | |
Collapse
|
30
|
The acquisition of malignant potential in colon cancer is regulated by the stabilization of Atonal homolog 1 protein. Biochem Biophys Res Commun 2013; 432:175-81. [PMID: 23333391 DOI: 10.1016/j.bbrc.2013.01.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 01/09/2013] [Indexed: 11/23/2022]
Abstract
The transcription factor Atonal homolog 1 (Atoh1) plays crucial roles in the differentiation of intestinal epithelium cells. Although we have reported that the Atoh1 protein was degraded in colon cancer by aberrant Wnt signaling, a recent study has indicated that the Atoh1 protein is expressed in mucinous colon cancer (MC) and signet ring cell carcinoma (SRCC). However, the roles of the Atoh1 protein in MC are unknown. To mimic MC, a mutated Atoh1 protein was stably expressed in undifferentiated colon cancer cells. Microarray analysis revealed the acquisition of not only the differentiated cell form, but also malignant potential by Atoh1 protein stabilization. In particular, Atoh1 enhanced Wnt signaling, resulting in the induction of Lgr5 as a representative stem cell marker with the enrichment of cancer stem cells. Moreover, the fluorescent ubiquitination-based cell cycle indicator system with time-lapse live imaging demonstrated cell cycle arrest in the G0/G1 phase by Atoh1 protein stabilization. In conclusion, the Atoh1 protein regulates malignant potential rather than the differentiation phenotype of MC, suggesting the mechanism by which MC and SRCC are more malignant than non-mucinous adenocarcinoma.
Collapse
|
31
|
Abstract
The small and large intestines are tubular organs composed of several tissue types. The columnar epithelium that lines the inner surface of the intestines distinguishes the digestive physiology of each region of the intestine and consists of several distinct cell types that are rapidly and continually renewed by intestinal stem cells that reside near the base of the crypts of Lieberkühn. Notch signaling controls the fate of intestinal stem cells by regulating the expression of Hes genes and by repressing Atoh1. Alternate models of Notch pathway control of cell fate determination are presented. Roles for Notch signaling in development of the intestine, including mesenchymal and neural cells, are discussed. The oncogenic activities of Notch in colorectal cancer, as well as the tumor suppressive activities of Atoh1, are reviewed. Therapeutic targeting of the Notch pathway in colorectal cancers is discussed, along with potential caveats.
Collapse
Affiliation(s)
- Taeko K Noah
- Division of Gastroenterology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | | |
Collapse
|