151
|
Musashi 2 is a regulator of the HSC compartment identified by a retroviral insertion screen and knockout mice. Blood 2011; 118:554-64. [DOI: 10.1182/blood-2010-12-322081] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
We used a retroviral integration screen to search for novel genes that regulate HSC function. One of the genes that conferred HSC dominance when overexpressed due to an adjacent retroviral insertion was Musashi 2 (Msi2), an RNA-binding protein that can act as a translational inhibitor. A gene-trap mouse model that inactivates the gene shows that Msi2 is more highly expressed in long-term (LT) and short-term (ST) HSCs, as well as in lymphoid myeloid primed progenitors (LMPPs), but much less in intermediate progenitors and mature cells. Mice lacking Msi2 are fully viable for up to a year or more, but exhibit severe defects in primitive precursors, most significantly a reduction in the number of ST-HSCs and LMPPs and a decrease in leukocyte numbers, effects that are exacerbated with age. Cell-cycle and gene-expression analyses suggest that the main hematopoietic defect in Msi2-defective mice is the decreased proliferation capacity of ST-HSCs and LMPPs. In addition, HSCs lacking Msi2 are severely impaired in competitive repopulation experiments, being overgrown by wild-type cells even when mutant cells were provided in excess. Our data indicate that Msi2 maintains the stem cell compartment mainly by regulating the proliferation of primitive progenitors downstream of LT-HSCs.
Collapse
|
152
|
Gunter KM, McLaughlin EA. Translational control in germ cell development: A role for the RNA-binding proteins Musashi-1 and Musashi-2. IUBMB Life 2011; 63:678-85. [PMID: 21766416 DOI: 10.1002/iub.499] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 04/13/2011] [Indexed: 12/26/2022]
Abstract
Mammalian gametogenesis is a complex process involving specialised cell cycle progression and differentiation. As part of their differentiation, germ cells experience periods of transcriptional inactivation and chromatin inaccessibility whilst continuing to coordinate the correct temporal and spatial expression of genes required for continued development. To overcome these obstacles, mammalian germ cells express a wide variety of sequence-specific RNA-binding proteins, which assist in the translational control of many mRNA transcripts which are produced and stored during periods of high mRNA synthesis. In this review we focus on the Musashi family of RNA-binding proteins, a highly conserved family of translational regulatory proteins whose recent identification in germ cells of Drosophila and Xenopus, as well as their well described role in processes such as cell cycle progression and stem cell identity, has led us to investigate the role of these proteins in mammalian germ cell development.
Collapse
Affiliation(s)
- Kara M Gunter
- Reproductive Science Group, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia
| | | |
Collapse
|
153
|
Changes in Musashi-1 subcellular localization correlate with cell cycle exit during postnatal retinal development. Exp Eye Res 2011; 92:344-52. [DOI: 10.1016/j.exer.2011.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/19/2011] [Accepted: 02/04/2011] [Indexed: 12/30/2022]
|
154
|
Colon Cancer Stem Cells: Bench-to-Bedside-New Therapeutical Approaches in Clinical Oncology for Disease Breakdown. Cancers (Basel) 2011; 3:1957-74. [PMID: 24212791 PMCID: PMC3757399 DOI: 10.3390/cancers3021957] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 04/11/2011] [Accepted: 04/12/2011] [Indexed: 12/18/2022] Open
Abstract
It is widely accepted by the scientific community that cancer, including colon cancer, is a “stem cell disease”. Until a few years ago, common opinion was that all neoplastic cells within a tumor contained tumorigenic growth capacity, but recent evidences hint to the possibility that such a feature is confined to a small subset of cancer-initiating cells, also called cancer stem cells (CSCs). Thus, malignant tumors are organized in a hierarchical fashion in which CSCs give rise to more differentiated tumor cells. CSCs possess high levels of ATP-binding cassette (ABC) transporters and anti-apoptotic molecules, active DNA-repair, slow replication capacities and they produce growth factors that confer refractoriness to antineoplastic treatments. The inefficacy of conventional therapies towards the stem cell population might explain cancer chemoresistance and the high frequency of relapse shown by the majority of tumors. Nowadays, in fact all the therapies available are not sufficient to cure patients with advanced forms of colon cancer since they target differentiated cancer cells which constitute most of the tumor mass and spare CSCs. Since CSCs are the entities responsible for the development of the tumor and represent the only cell population able to sustain tumor growth and progression, these cells represent the elective target for innovative therapies.
Collapse
|
155
|
MacNicol MC, Cragle CE, MacNicol AM. Context-dependent regulation of Musashi-mediated mRNA translation and cell cycle regulation. Cell Cycle 2011; 10:39-44. [PMID: 21191181 DOI: 10.4161/cc.10.1.14388] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Musashi-mediated mRNA translational control has been implicated in the promotion of physiological and pathological stem cell proliferation. During self-renewal of mammalian stem cells, Musashi has been proposed to act to repress the translation of mRNAs encoding inhibitors of cell cycle progression. By contrast, in maturing Xenopus oocytes Musashi activates translation of target mRNAs that encode proteins promoting cell cycle progression. The mechanisms directing Musashi to differentially control mRNA translation in mammalian stem cells and Xenopus oocytes is unknown. In this study, we demonstrate that the mechanisms defining Musashi function lie within the cellular context. Specifically, we show that murine Musashi acts as an activator of translation in maturing Xenopus oocytes while Xenopus Musashi functions as a repressor of target mRNA translation in mammalian cells. We further demonstrate that within the context of a primary mammalian neural stem/progenitor cell, Musashi can be converted from a repressor of mRNA translation to an activator of translation in response to extracellular stimuli. We present current models of Musashi-mediated mRNA translational control and discuss possible mechanisms for regulating Musashi function. An understanding of these mechanisms presents exciting possibilities for development of therapeutic targets to control physiological and pathological stem cell proliferation.
Collapse
Affiliation(s)
- Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | | |
Collapse
|
156
|
Sharma P, Cline HT. Visual activity regulates neural progenitor cells in developing xenopus CNS through musashi1. Neuron 2010; 68:442-55. [PMID: 21040846 DOI: 10.1016/j.neuron.2010.09.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2010] [Indexed: 11/30/2022]
Abstract
Regulation of progenitor cell fate determines the numbers of neurons in the developing brain. While proliferation of neural progenitors predominates during early central nervous system (CNS) development, progenitor cell fate shifts toward differentiation as CNS circuits develop, suggesting that signals from developing circuits may regulate proliferation and differentiation. We tested whether activity regulates neurogenesis in vivo in the developing visual system of Xenopus tadpoles. Both cell proliferation and the number of musashi1-immunoreactive progenitors in the optic tectum decrease as visual system connections become stronger. Visual deprivation for 2 days increased proliferation of musashi1-immunoreactive radial glial progenitors, while visual experience increased neuronal differentiation. Morpholino-mediated knockdown and overexpression of musashi1 indicate that musashi1 is necessary and sufficient for neural progenitor proliferation in the CNS. These data demonstrate a mechanism by which increased brain activity in developing circuits decreases cell proliferation and increases neuronal differentiation through the downregulation of musashi1 in response to circuit activity.
Collapse
Affiliation(s)
- Pranav Sharma
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
157
|
Lan SY, Yu T, Xia ZS, Yuan YH, Shi L, Lin Y, Huang KH, Chen QK. Musashi 1-positive cells derived from mouse embryonic stem cells can differentiate into neural and intestinal epithelial-like cells in vivo. Cell Biol Int 2010; 34:1171-1180. [PMID: 20670215 DOI: 10.1042/cbi20100108] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Msi1 (Musashi 1) is regarded as a marker for neural and intestinal epithelial stem cells. However, it is still unclear whether Msi1-positive cells derived from mouse embryonic stem cells have the ability to differentiate into neural or intestinal epithelial cells. A pMsi1-GFP (green fluorescent protein) reporter plasmid was constructed in order to sort Msi1-positive cells out of the differentiated cell population. The GFP-positive cells (i.e. Msi1-positive cells) were sorted by FACS and were hypodermically engrafted into the backs of NOD/SCID (non-obese diabetic/severe combined immunodeficient) mice. The presence of neural and intestinal epithelial cells in the grafts was detected. Msi1 was highly expressed in the GFP-positive cells, but not in the GFP-negative cells. The markers for neural cells (Nestin and Tubulin β III) and intestinal epithelial cells [FABP2 (fatty acid binding protein 2), Lyz (lysozyme) and ChA (chromogranin A)] were more highly expressed in the grafts from Msi1-positive cells than those from Msi1-negative cells (P<0.05). The grafts from the Msi1-negative cells contained more mesodermal-like tissues than those from the Msi1-positive cells. The pMsi1-GFP vector can be used to sort Msi1-positive cells from a cell population derived from mouse embryonic stem cells. The Msi1-positive cells can differentiate into neural and intestinal epithelial-like cells in vivo.
Collapse
Affiliation(s)
- Shao-Yang Lan
- Department of Gastroenterology, the Second Affiliated Hospital, Sun YatSen University, Guangzhou, Guangdong, Peoples Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
158
|
Chia LA, Kuo CJ. The intestinal stem cell. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010. [PMID: 21075344 DOI: 10.1016/b978‐0‐12‐381280‐3.00007‐5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The intestinal epithelium is one of the most rapidly proliferating organs in the body. A complete turnover of the epithelium occurs every 3-5 days in the mouse, a process that is maintained by a small population of intestinal stem cells (ISCs) that reside in the crypt bases. The signals that regulate the behavior of these ISCs are still unknown. This has been due, until recently, to the singular lack of definitive ISC markers. The recent identification of genes that mark functional stem cells has yielded insights into how ISCs are regulated and maintained by their surrounding niche. Herein, we examine the body of literature regarding the precise identity and location of the ISCs, the role of the surrounding niche in ISC maintenance and regulation, as well as the hypothesis that the ISCs are the cells of origin in colorectal cancer.
Collapse
Affiliation(s)
- Luis A Chia
- Cancer Biology Program, Stanford University School of Medicine, Stanford, California, USA
| | | |
Collapse
|
159
|
|
160
|
Konuma N, Wakabayashi K, Matsumoto T, Kusumi Y, Masuko T, Iribe Y, Mitsumata M, Okano H, Kusafuka T, Mugishima H. Mouse embryonic stem cells give rise to gut-like morphogenesis, including intestinal stem cells, in the embryoid body model. Stem Cells Dev 2010; 18:113-26. [PMID: 18680392 DOI: 10.1089/scd.2008.0045] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Embryonic stem (ES) cells have been proposed as candidates for cell replacement therapy in patients with intestinal failure because these cells can be expanded indefinitely without losing their pluripotent phenotype. We investigated the differentiation capacity of mouse ES cells into gut-like structures, including intestinal stem cells, and defined culture conditions for efficient induction of formation of these structures. ES cell-derived gut-like structures (ES-guts) were reproducibly induced in developing embryoid bodies (EBs) by day 21 of differentiation culture. ES-guts contained an endodermal epithelium, a smooth muscle layer, interstitial cells of Cajal, and enteric neurons and showed spontaneous contraction. Transplantation of ES-guts under the kidney capsules of immunodeficient mice induced formation of highly differentiated epithelium composed of absorptive cells and goblet cells in the grafts. Immunoreactivity for Musashi-1 (Msi-1), a marker of intestinal stem cells, was detected in 1.9% of the columnar epithelial cells in the graft. Culture with 0.1% dimethyl sulfoxide increased the numbers of ES-guts in EBs, and serum-replacement (SR) culture, in comparison to standard ES culture containing 15% serum, increased the area ratio of ES-guts to EBs. SR culture also promoted maturation of epithelium to form a single layer of columnar epithelial cells, including absorptive cells and goblet cells. Expression of Msi-1 mRNA and protein was significantly enhanced when EBs were cultured under SR conditions. In conclusion, SR conditions efficiently induce formation of ES-guts and promote differentiation of epithelium, including intestinal stem cells. These results suggest the feasibility of cell-based therapy for intestinal failure based on ES cell culture systems.
Collapse
Affiliation(s)
- Noriyoshi Konuma
- Division of Cell Regeneration and Transplantation, Advanced Medical Research Center, Nihon University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
An RNAi screen identifies Msi2 and Prox1 as having opposite roles in the regulation of hematopoietic stem cell activity. Cell Stem Cell 2010; 7:101-13. [PMID: 20621054 DOI: 10.1016/j.stem.2010.06.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 03/26/2010] [Accepted: 06/09/2010] [Indexed: 12/18/2022]
Abstract
In this study, we describe an in vivo RNA interference functional genetics approach to evaluate the role of 20 different conserved polarity factors and fate determinants in mouse hematopoietic stem cell (HSC) activity. In total, this screen revealed three enhancers and one suppressor of HSC-derived reconstitution. Pard6a, Prkcz, and Msi2 shRNA-mediated depletion significantly impaired HSC repopulation. An in vitro promotion of differentiation was observed after the silencing of these genes, consistent with their function in regulating HSC self-renewal. Conversely, Prox1 knockdown led to in vivo accumulation of primitive and differentiated cells. HSC activity was also enhanced in vitro when Prox1 levels were experimentally reduced, identifying it as a potential antagonist of self-renewal. HSC engineered to overexpress Msi2 or Prox1 showed the reverse phenotype to those transduced with corresponding shRNA vectors. Gene expression profiling studies identified a number of known HSC and cell cycle regulators as potential downstream targets to Msi2 and Prox1.
Collapse
|
162
|
Nishimoto Y, Okano H. New insight into cancer therapeutics: induction of differentiation by regulating the Musashi/Numb/Notch pathway. Cell Res 2010; 20:1083-5. [PMID: 20805843 DOI: 10.1038/cr.2010.122] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Yoshinori Nishimoto
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | | |
Collapse
|
163
|
Rezza A, Skah S, Roche C, Nadjar J, Samarut J, Plateroti M. The overexpression of the putative gut stem cell marker Musashi-1 induces tumorigenesis through Wnt and Notch activation. J Cell Sci 2010; 123:3256-65. [PMID: 20826465 DOI: 10.1242/jcs.065284] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The RNA-binding protein Musashi-1 (Msi1) has been proposed as a marker of intestinal epithelial stem cells. These cells are responsible for the continuous renewal of the intestinal epithelium. Although the function of Msi1 has been studied in several organs from different species and in mammalian cell lines, its function and molecular regulation in mouse intestinal epithelium progenitor cells are still undefined. We describe here that, in these cells, the expression of Msi1 is regulated by the canonical Wnt pathway, through a mechanism involving a functional Tcf/Lef binding site on its promoter. An in vitro study in intestinal epithelium primary cultures showed that Msi1 overexpression promotes progenitor proliferation and activates Wnt and Notch pathways. Moreover, Msi1-overexpressing cells exhibit tumorigenic properties in xenograft experiments. These data point to a positive feedback loop between Msi1 and Wnt in intestinal epithelial progenitors. They also suggest that Msi1 has oncogenic properties in these cells, probably through induction of both the Wnt and Notch pathways.
Collapse
Affiliation(s)
- Amelie Rezza
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | | | | | | | | | | |
Collapse
|
164
|
Abstract
Although the notion that cancer is a disease caused by genetic and epigenetic alterations is now widely accepted, perhaps more emphasis has been given to the fact that cancer is a genetic disease. It should be noted that in the post-genome sequencing project period of the 21st century, the underlined phenomenon nevertheless could not be discarded towards the complete control of cancer disaster as the whole strategy, and in depth investigation of the factors associated with tumorigenesis is required for achieving it. Otto Warburg has won a Nobel Prize in 1931 for the discovery of tumor bioenergetics, which is now commonly used as the basis of positron emission tomography (PET), a highly sensitive noninvasive technique used in cancer diagnosis. Furthermore, the importance of the cancer stem cell (CSC) hypothesis in therapy-related resistance and metastasis has been recognized during the past 2 decades. Accumulating evidence suggests that tumor bioenergetics plays a critical role in CSC regulation; this finding has opened up a new era of cancer medicine, which goes beyond cancer genomics.
Collapse
Affiliation(s)
- Hideshi Ishii
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
165
|
Wang XY, Penalva LO, Yuan H, Linnoila RI, Lu J, Okano H, Glazer RI. Musashi1 regulates breast tumor cell proliferation and is a prognostic indicator of poor survival. Mol Cancer 2010; 9:221. [PMID: 20727204 PMCID: PMC2939568 DOI: 10.1186/1476-4598-9-221] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 08/21/2010] [Indexed: 12/23/2022] Open
Abstract
Background Musashi1 (Msi1) is a conserved RNA-binding protein that regulates the Notch and Wnt pathways, and serves as a stem cell marker in the breast and other tissues. It is unknown how Msi1 relates to other breast cancer markers, whether it denotes tumor initiating cells (TICs), and how it affects gene expression and tumor cell survival in breast cancer cells. Results Msi1 expression was analyzed in 20 breast cancer cell lines and in 140 primary breast tumors by western blotting and immunohistochemistry, respectively. Lentivirus RNA interference was used to reduce Msi1 expression in breast cancer cell lines MCF-7 and T47D grown as spheroid cultures and to assess stem cell gene expression and the growth of these cell lines as xenografts. In normal human breast tissue, Msi1 was expressed in 10.6% of myoepithelum and 1.2% of ductal epithelium in the terminal ductal lobular unit (TDLU), whereas, less than 0.05% of ductal epithelium and myoepithelium in large ducts outside the TDLU expressed Msi1. Msi1 was expressed in 55% of the breast cancer cell lines and correlated with ErbB2 expression in 50% of the cell lines. Msi1 was expressed in 68% of primary tumors and in 100% of lymph node metastases, and correlated with 5 year survival. Msi1 was enriched in CD133+ MCF-7 and T47D cells and in spheroid cultures of these cells, and Msi1 'knockdown' (KD) with a lentivirus-expressed shRNA decreased the number and size of spheroid colonies. Msi1 KD reduced Notch1, c-Myc, ErbB2 and pERK1/2 expression, and increased p21CIP1 expression, which is consistent with known Msi1 target mRNAs. Msi1 KD also reduced the expression of the somatic and embryonic stem cell markers, CD133, Bmi1, Sox2, Nanog and Oct4. Xenografts of MCF-7 and T47D Msi1 KD cells resulted in a marked reduction of tumor growth, reduced Msi1 and Notch1 expression and increased p21CIP1 expression. Conclusion Msi1 is a negative prognostic indicator of breast cancer patient survival, and is indicative of tumor cells with stem cell-like characteristics. Msi1 KD reduces tumor cell survival and tumor xenograft growth, suggesting that it may represent a novel target for drug discovery.
Collapse
Affiliation(s)
- Xiao-Yang Wang
- Department of Oncology, Georgetown University, and Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA.
| | | | | | | | | | | | | |
Collapse
|
166
|
Ito T, Kwon HY, Zimdahl B, Congdon KL, Blum J, Lento WE, Zhao C, Lagoo A, Gerrard G, Foroni L, Goldman J, Goh H, Kim SH, Kim DW, Chuah C, Oehler VG, Radich JP, Jordan CT, Reya T. Regulation of myeloid leukaemia by the cell-fate determinant Musashi. Nature 2010; 466:765-8. [PMID: 20639863 PMCID: PMC2918284 DOI: 10.1038/nature09171] [Citation(s) in RCA: 277] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 05/13/2010] [Indexed: 12/25/2022]
Abstract
Chronic myelogenous leukemia (CML) can progress from an indolent chronic phase to an aggressive blast crisis phase1 but the molecular basis of this transition remains poorly understood. Here we have used mouse models of CML2,3 to show that disease progression is regulated by the Musashi-Numb signaling axis4,5. Specifically, we find that chronic phase is marked by high and blast crisis phase by low levels of Numb expression, and that ectopic expression of Numb promotes differentiation and impairs advanced phase disease in vivo. As a possible explanation for the decreased levels of Numb in blast crisis, we show that NUP98-HOXA9, an oncogene associated with blast crisis CML6,7, can trigger expression of the RNA binding protein Musashi2 (Msi2) which in turn represses Numb. Importantly, loss of Msi2 restores Numb expression and significantly impairs the development and propagation of blast crisis CML in vitro and in vivo. Finally, we show that Msi2 expression is not only highly upregulated during human CML progression but is also an early indicator of poorer prognosis. These data show that the Musashi-Numb pathway can control the differentiation of CML cells, and raise the possibility that targeting this pathway may provide a new strategy for therapy of aggressive leukemias.
Collapse
Affiliation(s)
- Takahiro Ito
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Abstract
Carcinomas may arise as a disorder of regeneration, so that a malignant cell may represent a failure to fully attain the characteristics of differentiated tissue. We hypothesized that there is a differential distribution of progenitor cell markers among different histological types of lung cancers, with poorly differentiated tumors being more likely to express progenitor stem cell markers. The study was limited to paraffin-embedded archival material of resected untreated pulmonary carcinomas, including adenocarcinoma, squamous cell carcinoma, large cell carcinoma, and small cell carcinoma. The sections were stained for putative stem cells markers (Musashi-1, Musashi-2, CD34, CD21, KIT, CD133, p63, and OCT-4). Positivity was read as isolated, focal, or diffuse staining. Stem cell markers were detected in all histological types of pulmonary carcinomas. There was a difference in the expression of markers among the histological types. Small cell carcinoma showed diffuse positivity for most of the markers; in contrast to focal or negative staining in other histological groups. An inverse relationship between CD21 and Musashi-1 was observed. No staining for OCT-4 and CD34 was seen in any of the tumor types. Hierarchical clustering based on marker expression separated tumors into two groups, with one group marked by high expression of Musashi-1 and KIT, contained most of the poorly differentiated adenocarcinomas and small cell carcinomas. Therefore, stem cell markers are expressed in lung cancers with different patterns seen for different histological types and degrees of differentiation.
Collapse
|
168
|
Todaro M, Francipane MG, Medema JP, Stassi G. Colon cancer stem cells: promise of targeted therapy. Gastroenterology 2010; 138:2151-62. [PMID: 20420952 DOI: 10.1053/j.gastro.2009.12.063] [Citation(s) in RCA: 346] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 11/28/2009] [Accepted: 12/15/2009] [Indexed: 12/12/2022]
Abstract
First developed for hematologic disorders, the concept of cancer stem cells (CSCs) was expanded to solid tumors, including colorectal cancer (CRC). The traditional model of colon carcinogenesis includes several steps that occur via mutational activation of oncogenes and inactivation of tumor suppressor genes. Intestinal epithelial cells exist for a shorter amount of time than that required to accumulate tumor-inducing genetic changes, so researchers have investigated the concept that CRC arises from the long-lived stem cells, rather than from the differentiated epithelial cells. Colon CSCs were originally identified through the expression of the CD133 glycoprotein using an antibody directed to its epitope AC133. It is not clear if CD133 is a marker of colon CSCs-other cell surface markers, such as epithelial-specific antigen, CD44, CD166, Musashi-1, CD29, CD24, leucine-rich repeat-containing G-protein-coupled receptor 5, and aldehyde dehydrogenase 1, have been proposed. In addition to initiating and sustaining tumor growth, CSCs are believed to mediate cancer relapse after chemotherapy. How can we identify and analyze colon CSCs and what agents are being designed to kill this chemotherapy-refractory population?
Collapse
Affiliation(s)
- Matilde Todaro
- Department of Surgical and Oncological Sciences, Cellular and Molecular Pathophysiology Laboratory, Palermo, Italy
| | | | | | | |
Collapse
|
169
|
Musashi1 as a marker of reactive astrocytes after transient focal brain ischemia. Neurosci Res 2010; 66:390-5. [DOI: 10.1016/j.neures.2009.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2009] [Revised: 12/09/2009] [Accepted: 12/15/2009] [Indexed: 11/22/2022]
|
170
|
Hattori E, Shu HJ, Saito T, Okumoto K, Haga H, Yokozawa J, Ito J, Watanabe H, Saito K, Togashi H, Kawata S. Expression of the RNA-binding protein Musashi1 in adult liver stem-like cells. Hepatol Res 2010; 40:432-7. [PMID: 20394675 DOI: 10.1111/j.1872-034x.2009.00612.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM Musashi1 is an RNA-binding protein that regulates the Notch signaling pathway in stem cells. Our previous study revealed that Musashi1 is expressed in early hepatocytes during liver development in the mouse. However, whether this unique protein is expressed with Notch signaling markers in adult liver stem-like cells remains unknown. METHODS Established hepatic stem-like cells (HSLC), which were derived from adult Sprague-Dawley rats, were used for experiments in vitro. HSLC were differentiated into mature cells in terms of producing albumin when co-cultured with epidermal growth factor (EGF). The mRNA expression of Musashi1, Notch family (Notch1 and Notch2), Jagged1 and Hes1 was examined in HSLC before and after cell differentiation using polymerase chain reaction-based techniques. Protein expression of Musashi1 was examined in the HSLC and normal mature hepatocytes by immunofluorescence staining. RESULTS The mRNA expression of Musashi1, Notch1, Jagged1 and Hes1 was detected in the original HSLC before culturing with EGF but not in primary cultured mature hepatocytes. The mRNA expression of Musashi1 and Hes1 was found to be downregulated in differentiated HSLC that produce albumin. Protein expression of Musashi1 was detectable in the original HSLC but not in both differentiated HSLC and mature hepatocytes. CONCLUSION These findings demonstrate that the RNA-binding protein Musashi1 is expressed with Notch signaling markers in adult liver stem-like cells.
Collapse
Affiliation(s)
- Etsuko Hattori
- Department of Gastroenterology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata, Yamagata 990-9585, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Anant S, Houchen CW, Pawar V, Ramalingam S. Role of RNA-Binding Proteins in Colorectal Carcinogenesis. CURRENT COLORECTAL CANCER REPORTS 2010; 6:68-73. [PMID: 20401169 DOI: 10.1007/s11888-010-0048-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
RNA-binding proteins (RBPs) play key roles in the posttranscriptional regulation of gene expression. RBPs control various posttranscriptional events, including splicing, polyadenylation, mRNA stability, transport, and translation. It is becoming apparent that RBPs play a significant role in pathophysiologic conditions such as inflammation and cancer. More importantly, we and others have begun dissecting the role of mRNA stability and translation in regulating gene expression, dysregulation of which has serious consequences for the fate of the cell. In this article, we discuss this emerging area of posttranscriptional gene regulation and the role of RBPs in the aberrant expression of proteins in tumorigenesis.
Collapse
Affiliation(s)
- Shrikant Anant
- Section of Digestive Diseases and Nutrition, Department of Internal Medicine, University of Oklahoma Health Sciences Center, 920 Stanton L. Young Boulevard WP1345, Oklahoma City, OK 73104, USA; Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | |
Collapse
|
172
|
Chopra DP, Dombkowski AA, Stemmer PM, Parker GC. Intestinal epithelial cells in vitro. Stem Cells Dev 2010; 19:131-42. [PMID: 19580443 PMCID: PMC3136723 DOI: 10.1089/scd.2009.0109] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 07/06/2009] [Indexed: 02/06/2023] Open
Abstract
Recent advances in the biology of stem cells has resulted in significant interest in the development of normal epithelial cell lines from the intestinal mucosa, both to exploit the therapeutic potential of stem cells in tissue regeneration and to develop treatment models of degenerative disorders of the digestive tract. However, the difficulty of propagating cell lines of normal intestinal epithelium has impeded research into the molecular mechanisms underlying differentiation of stem/progenitor cells into the various intestinal lineages. Several short-term organ/organoid and epithelial cell culture models have been described. There is a dearth of long-term epithelial and/or stem cell cultures of intestine. With an expanding role of stem cells in the treatment of degenerative disorders, there is a critical need for additional efforts to develop in vitro models of stem/progenitor epithelial cells of intestine. The objective of this review is to recapitulate the current status of technologies and knowledge for in vitro propagation of intestinal epithelial cells, markers of the intestinal stem cells, and gene and protein expression profiles of the intestinal cellular differentiation.
Collapse
Affiliation(s)
- Dharam P. Chopra
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan
| | - Alan A. Dombkowski
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan
| | - Paul M. Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan
| | - Graham C. Parker
- Carman and Ann Adams Department of Pediatrics, Wayne State University, Detroit, Michigan
| |
Collapse
|
173
|
Potten CS, Gandara R, Mahida YR, Loeffler M, Wright NA. The stem cells of small intestinal crypts: where are they? Cell Prolif 2009; 42:731-50. [PMID: 19788585 PMCID: PMC6496740 DOI: 10.1111/j.1365-2184.2009.00642.x] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 05/27/2009] [Indexed: 12/11/2022] Open
Abstract
Recently, there has been resurgence of interest in the question of small intestinal stem cells, their precise location and numbers in the crypts. In this article, we attempt to re-assess the data, including historical information often omitted in recent studies on the subject. The conclusion we draw is that the evidence supports the concept that active murine small intestinal stem cells in steady state are few in number and are proliferative. There are two evolving, but divergent views on their location (which may be more related to scope of capability and reversibility than to location) several lineage labelling and stem cell self-renewing studies (based on Lgr5 expression) suggest a location intercalated between the Paneth cells (crypt base columnar cells (CBCCs)), or classical cell kinetic, label-retention and radiobiological evidence plus other recent studies, pointing to a location four cell positions luminally from the base of the crypt The latter is supported by recent lineage labelling of Bmi-1-expressing cells and by studies on expression of Wip-1 phosphatase. The situation in the human small intestine remains unclear, but recent mtDNA mutation studies suggest that the stem cells in humans are also located above the Paneth cell zone. There could be a distinct and as yet undiscovered relationship between these observed traits, with stem cell properties both in cells of the crypt base and those at cell position 4.
Collapse
|
174
|
Ricci-Vitiani L, Fabrizi E, Palio E, De Maria R. Colon cancer stem cells. J Mol Med (Berl) 2009; 87:1097-104. [PMID: 19727638 DOI: 10.1007/s00109-009-0518-4] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 08/10/2009] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is the third most common form of cancer and the second cause of cancer-related death in the Western world, leading to 655,000 deaths worldwide per year (Jemal et al. in CA Cancer J Clin 56:106-130, 2006). Despite the emergence of new targeted agents and the use of various therapeutic combinations, none of the treatment options available is curative in patients with advanced cancer. A growing body of evidence is increasingly supporting the idea that human cancers can be considered as a stem cell disease. According to the cancer stem cell model, malignancies originate from a small fraction of cancer cells that show self-renewal and pluripotency and are capable of initiating and sustaining tumor growth (Boman and Wicha in J Clin Oncol 26:2795-2799, 2008). The cancer-initiating cells or "cancer stem cells" were first identified in hematologic malignancies and most recently in several solid tumors, including CRC. The hypothesis of stem cell-driven tumorigenesis in colon cancer raises questions as to whether current treatments are able to efficiently target the tumorigenic cell population that is responsible for tumor growth and maintenance. This review will focus on the different aspects of stem cell biology in the context of CRC, which might help to understand the mechanisms that give rise to tumor development and resistance to therapy. First, we will briefly revise the knowledge available on normal intestinal stem cells and recent advances in understanding crypt biology, which have led to new theory on the origins of colon adenomas and cancers. Then, we will summarize the evidence and current status on colon cancer stem cells, focusing on their relevance and promises for the treatment of colorectal carcinoma.
Collapse
Affiliation(s)
- Lucia Ricci-Vitiani
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | | | | | | |
Collapse
|
175
|
McDonald SAC, Graham TA, Schier S, Wright NA, Alison MR. Stem cells and solid cancers. Virchows Arch 2009; 455:1-13. [PMID: 19499244 DOI: 10.1007/s00428-009-0783-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 04/22/2009] [Accepted: 04/28/2009] [Indexed: 02/06/2023]
Abstract
Recently, there have been significant advances in our knowledge of stem cells found in tissues that can develop solid tumours. In particular, novel stem cell markers have been identified for the first time identifying multipotential cells: a required characteristic of a stem cell. The scarcity of cancer stem cells has been questioned. Current dogma states that they are rare, but novel research has suggested that this may not be the case. Here, we review the latest literature on stem cells, particularly cancer stem cells within solid tumours. We discuss current thinking on how stem cells develop into cancer stem cells and how they protect themselves from doing so and do they express unique markers that can be used to detect stem cells. We attempt to put into perspective these latest advances in stem cell biology and their potential for cancer therapy.
Collapse
Affiliation(s)
- Stuart A C McDonald
- Centre for Gastroenterology, Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Blizard Building, 4 Newark Street, Whitechapel, London, E1 2AT, UK.
| | | | | | | | | |
Collapse
|
176
|
Willis ND, Przyborski SA, Hutchison CJ, Wilson RG. Colonic and colorectal cancer stem cells: progress in the search for putative biomarkers. J Anat 2008; 213:59-65. [PMID: 18638071 DOI: 10.1111/j.1469-7580.2008.00917.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The maintenance of healthy colonic crypts is dependent on the integrity of the adult epithelial stem cells located within them. Perturbations in stem cell dynamics are generally believed to represent the first step towards colorectal tumorigenesis. Experimental manipulation of intestinal stem cells has greatly increased our understanding of them, but further progress has been slowed due to the absence of a reliable stem cell biomarker. In this review we discuss the candidate colonic stem cell biomarkers which have been proposed. Furthermore, we investigate the putative biomarkers for so-called colorectal cancer stem cells, a highly aggressive subpopulation of cells considered to drive tumour development.
Collapse
Affiliation(s)
- Naomi D Willis
- School of Biological and Biomedical Sciences, Durham University, Durham, UK.
| | | | | | | |
Collapse
|
177
|
Kaneko J, Chiba C. Immunohistochemical analysis of Musashi-1 expression during retinal regeneration of adult newt. Neurosci Lett 2008; 450:252-7. [PMID: 19028551 DOI: 10.1016/j.neulet.2008.11.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 11/05/2008] [Accepted: 11/12/2008] [Indexed: 11/29/2022]
Abstract
The adult newt retinal regeneration is an ideal model for studying retinal regeneration by transdifferentiation of the retinal pigment epithelium (RPE) cells. Accumulated evidence suggests that the RNA-binding protein Musashi-1 (Msi1) is expressed in mature photoreceptors and RPE cells as well as in retinal stem/progenitor cells, being essential for vision. We have been investigating whether Msi1 is also essential for retinal regeneration. In the last paper [K. Susaki, J. Kaneko, Y. Yamano, K. Nakamura, W. Inami, T. Yoshikawa, Y. Ozawa, S. Shibata, O. Matsuzaki, H. Okano, C. Chiba, Musashi-1, an RNA-binding protein, is indispensable for survival of photoreceptors. Exp. Eye Res. (in press)], we showed that the expression profiles of Msi1 transcripts and protein isoforms change during retinal regeneration. In the current report, we show by immunohistochemistry that Msi1 is expressed in transdifferentiating cells or cells of regenerating retinal tissues. Upon retinectomy, Msi1 protein, which is expressed in the nuclei of intact (stage E-0) RPE cells, changed its subcellular localization, being expressed in both the nucleus and cytoplasm of the RPE-derived stem-like cells at stage E-1. As the retinal rudiment/regenerating retina (rR) and renewing RPE (rRPE) are specified from the stem-like cell population (stage E-2), Msi1 expression was maintained or up-regulated in the rR, while down-regulated in the rRPE. During further retinal regeneration, Msi1 expression was decreased in association with cell differentiation, except for photoreceptors and RPE cells whose Msi1 expression increased as they differentiate. Thus, Msi1 is likely to be regulated at various cellular events during retinal regeneration, implying that Msi1 may have multi-functions in retinal regeneration. All together, it is probable that Msi1 is one of the essential factors that need to be regulated in retinal regeneration.
Collapse
Affiliation(s)
- Jun Kaneko
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8572, Japan
| | | |
Collapse
|
178
|
Function and regulation of the mammalian Musashi mRNA translational regulator. Biochem Soc Trans 2008; 36:528-30. [PMID: 18481998 DOI: 10.1042/bst0360528] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The evolutionarily conserved RNA-binding protein, Musashi, regulates neural stem cell self-renewal. Musashi expression is also indicative of stem cell populations in breast and intestinal tissues and is linked to cell overproliferation in cancers of these tissues. Musashi has been primarily implicated as a repressor of target mRNAs in stem cell populations. However, little is known about the mechanism by which Musashi exerts mRNA translational control or how Musashi function is regulated. Recent findings in oocytes of the frog, Xenopus, indicate an unexpected role for Musashi as an activator of a number of maternal mRNAs during meiotic cell cycle progression. Given the importance of Musashi function in stem cell biology and the implications of aberrant Musashi expression in cancer, it is critical that we understand the molecular processes that regulate Musashi function.
Collapse
|
179
|
Markel TA, Crisostomo PR, Lahm T, Novotny NM, Rescorla FJ, Tector AJ, Meldrum DR. Stem cells as a potential future treatment of pediatric intestinal disorders. J Pediatr Surg 2008; 43:1953-63. [PMID: 18970924 PMCID: PMC2584666 DOI: 10.1016/j.jpedsurg.2008.06.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Revised: 05/18/2008] [Accepted: 06/22/2008] [Indexed: 12/27/2022]
Abstract
All surgical disciplines encounter planned and unplanned ischemic events that may ultimately lead to cellular dysfunction and death. Stem cell therapy has shown promise for the treatment of a variety of ischemic and inflammatory disorders where tissue damage has occurred. As stem cells have proven beneficial in many disease processes, important opportunities in the future treatment of gastrointestinal disorders may exist. Therefore, this article will serve to review the different types of stem cells that may be applicable to the treatment of gastrointestinal disorders, review the mechanisms suggesting that stem cells may work for these conditions, discuss current practices for harvesting and purifying stem cells, and provide a concise summary of a few of the pediatric intestinal disorders that could be treated with cellular therapy.
Collapse
Affiliation(s)
- Troy A. Markel
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Paul R. Crisostomo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Tim Lahm
- Department of Pulmonary and Critical Care Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Nathan M. Novotny
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - A. Joseph Tector
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Daniel R. Meldrum
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana,Center for Immunobiology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
180
|
Kinomura M, Kitamura S, Tanabe K, Ichinose K, Hirokoshi K, Takazawa Y, Kitayama H, Nasu T, Sugiyama H, Yamasaki Y, Sugaya T, Maeshima Y, Makino H. Amelioration of cisplatin-induced acute renal injury by renal progenitor-like cells derived from the adult rat kidney. Cell Transplant 2008; 17:143-58. [PMID: 18468244 DOI: 10.3727/000000008783907008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The replacement of a necrotic tubular epithelium with functional tubular epithelial cells is required for recovery from acute renal failure (ARF). A rat renal progenitor-like (rKS56) cell line was recently established derived from the S3 segment of renal proximal tubules. The therapeutic efficacy of rKS56 cells was examined in a rat model of cisplatin-induced ARF. rKS56-lacZ cells expressing 3-galactosidase were injected into SD rats either at the subcapsule of the left kidney (rKS-SC) or via the left renal artery (rKS-IA) 2 days after the injection of cisplatin. Bluo-gal(+) rKS56-lacZ cells were observed in the subcapsule in the rKS-SC group on day 5, and were further increased in number on day 9, accompanied by partial distribution in the corticomedullary junction, but not in the rKS-IA group. A portion of Bluo-gal(+) cells coexpressed Ki-67, aquaporin-1, hepatocyte growth factor (HGF), and c-Met. rKS-SC treatment significantly improved the tubular injury scores, ameliorated tubular cell apoptosis, and induced cell proliferation. The renal function also significantly improved in the rKS-SC group on day 5. These results demonstrate that locally implanted rKS56 cells could differentiate into tubular epithelial cells, thereby accelerating the recovery from tubular injury, most likely by producing tubular trophic factors. These results suggest the therapeutic potential of this novel approach for patients with end-stage renal failure.
Collapse
Affiliation(s)
- Masaru Kinomura
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Ishii H, Iwatsuki M, Ieta K, Ohta D, Haraguchi N, Mimori K, Mori M. Cancer stem cells and chemoradiation resistance. Cancer Sci 2008; 99:1871-7. [PMID: 19016744 PMCID: PMC11159283 DOI: 10.1111/j.1349-7006.2008.00914.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 06/03/2008] [Accepted: 06/03/2008] [Indexed: 12/30/2022] Open
Abstract
Cancer is a disease of genetic and epigenetic alterations, which are emphasized as the central mechanisms of tumor progression in the multistepwise model. Discovery of rare subpopulations of cancer stem cells (CSCs) has created a new focus in cancer research. The heterogeneity of tumors can be explained with the help of CSCs supported by antiapoptotic signaling. CSCs mimic normal adult stem cells by demonstrating resistance to toxic injuries and chemoradiation therapy. Moreover, they might be responsible for tumor relapse following apparent beneficial treatments. Compared with hematopoietic malignancies, conventional therapy regimes in solid tumors have improved the overall survival marginally, illustrating the profound impact of treatment resistance. This implies that the present therapies, which follow total elimination of rapidly dividing and differentiated tumor cells, need to be modified to target CSCs that repopulate the tumor. In this review article, we report on recent findings regarding the involvement of CSCs in chemoradiation resistance and provide new insights into their therapeutic implications in cancer.
Collapse
Affiliation(s)
- Hideshi Ishii
- Department of Gastroenterological Surgery, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
182
|
Glazer RI, Wang XY, Yuan H, Yin Y. Musashi1: a stem cell marker no longer in search of a function. Cell Cycle 2008; 7:2635-9. [PMID: 18719393 DOI: 10.4161/cc.7.17.6522] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
One of the earliest genes identified with stem and early progenitor cells is the RNA-binding protein, Musashi1 (Msi1). Through gene profiling of mammary epithelial cells transduced with Msi1, a unique autocrine signaling pathway was identified that activates both the Wnt and Notch pathways. This process was associated with increased secretion of the growth factor, PLF1 and inhibition of the secreted Wnt pathway inhibitor, DKK3. Identification of PLF1 as an effector of these pathways in the absence of the DKK3 tumor suppressor provides a new avenue for investigating differences between normal and malignant tissues, and potentially targeting tumor stem cells.
Collapse
Affiliation(s)
- Robert I Glazer
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA.
| | | | | | | |
Collapse
|
183
|
The expression of Msi-1 and its significance in small intestinal mucosa severely damaged by high-dose 5-FU. Dig Dis Sci 2008; 53:2436-42. [PMID: 18270838 DOI: 10.1007/s10620-007-0155-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2007] [Accepted: 11/26/2007] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The purpose was to investigate the expression of musashi-1 (msi-1) and its significances in small intestinal mucosa that was severely damaged by high-dose 5-FU. METHODS A total of 40 adult C57BL/6J mice were divided into two groups: the control group (n = 8, group A) and experimental group (n = 32). The mice in the control group were treated with PBS by intraperitoneal injection, and the other mice were treated with high-dose 5-FU (150 mg/kg body weight for 5 consecutive days) by intraperitoneal injection. At the 1st (group B), 3rd (group C) and 5th (group D) day after treatment with high-dose 5-FU, the dying mice were killed, HE staining and immunohistochemical techniques were used to detect the expression of the putative marker of intestinal epithelial stem cells, msi-1, in samples of the middle intestine from these mice, and the percentage of the msi-1-positive cells from the intestinal mucosal cells of the mice in group B was detected by FACS. RESULTS After treatment with high-dose 5-FU, the intestinal mucosa suffered severe damage: the villi and crypts disappeared, the number of msi-1-positive cells increased greatly, the intestinal epithelial cells could be divided into two fractions by FACS, and the percentage of msi-1-positive cells was up to 67.75% in the fraction in which the value of FSC was higher. CONCLUSIONS After treatment with high-dose 5-FU, the percentage of intestinal stem cells had increased significantly, which was useful for the further isolation and enrichment of intestinal epithelial stem cells.
Collapse
|
184
|
Kanaya T, Miyazawa K, Takakura I, Itani W, Watanabe K, Ohwada S, Kitazawa H, Rose MT, McConochie HR, Okano H, Yamaguchi T, Aso H. Differentiation of a murine intestinal epithelial cell line (MIE) toward the M cell lineage. Am J Physiol Gastrointest Liver Physiol 2008; 295:G273-84. [PMID: 18556421 DOI: 10.1152/ajpgi.00378.2007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
M cells are a kind of intestinal epithelial cell in the follicle-associated epithelium of Peyer's patches. These cells can transport antigens and microorganisms into underlying lymphoid tissues. Despite the important role of M cells in mucosal immune responses, the origin and mechanisms of differentiation as well as cell death of M cells remain unclear. To clarify the mechanism of M cell differentiation, we established a novel murine intestinal epithelial cell line (MIE) from the C57BL/6 mouse. MIE cells grow rapidly and have a cobblestone morphology, which is a typical feature of intestinal epithelial cells. Additionally, they express cytokeratin, villin, cell-cell junctional proteins, and alkaline phosphatase activity and can form microvilli. Their expression of Musashi-1 antigen indicates that they may be close to intestinal stem cells or transit-amplifying cells. MIE cells are able to differentiate into the M cell lineage following coculture with intestinal lymphocytes, but not with Peyer's patch lymphocytes (PPL). However, PPL costimulated with anti-CD3/CD28 MAbs caused MIE cells to display typical features of M cells, such as transcytosis activity, the disorganization of microvilli, and the expression of M cell markers. This transcytosis activity of MIE cells was not induced by T cells isolated from PPL costimulated with the same MAbs and was reduced by the depletion of the T cell population from PPL. A mixture of T cells treated with MAbs and B cells both from PPL led MIE cells to differentiate into M cells. We report here that MIE cells have the potential ability to differentiate into M cells and that this differentiation required activated T cells and B cells.
Collapse
Affiliation(s)
- Takashi Kanaya
- Cellular Biology Laboratory, Graduate School of Agricultural Science, Tohoku Univ., 1-1 Tsutsumidori Amamiyamachi, Aoba-ku, 981-8555 Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Asai R, Okano H, Yasugi S. Correlation between Musashi-1 and c-hairy-1 expression and cell proliferation activity in the developing intestine and stomach of both chicken and mouse. Dev Growth Differ 2008; 47:501-10. [PMID: 16287482 DOI: 10.1111/j.1440-169x.2005.00825.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Musashi-1 (Msi-1) is an RNA-binding protein that plays key roles in the maintenance of neural stem cell states and in their differentiation into neural cells. Msi-1 has also been proposed as a candidate marker gene of mammalian intestinal stem cells and their immediate lineages. In this study, we examined Msi-1 expression in the small intestine and the stomach of both chicken and mouse during embryonic, fetal and postnatal development. In addition, we analyzed the expression of c-hairy-1, a chicken homologue of mouse Hes1, and assessed the proliferative activity of the cells expressing both of these factors. Significantly, during the development of these digestive organs in both species Msi-1 expression showed dynamic changes, suggesting that it is important for digestive organ development, particularly for epithelial differentiation. Based on our observations of the expression patterns of Msi-1 and c-hairy-1 in the adult small intestine, we speculate that Msi-1 is also a stem cell marker of the chicken small intestinal epithelium.
Collapse
Affiliation(s)
- Rieko Asai
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, 1-1, Minamiosawa, Hachioji, Tokyo 192-0397, Japan
| | | | | |
Collapse
|
186
|
Expression and functional analysis of musashi-like genes in planarian CNS regeneration. Mech Dev 2008; 125:631-45. [DOI: 10.1016/j.mod.2008.03.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Revised: 03/01/2008] [Accepted: 03/11/2008] [Indexed: 01/01/2023]
|
187
|
Fabre CCG, Casal J, Lawrence PA. The abdomen of Drosophila: does planar cell polarity orient the neurons of mechanosensory bristles? Neural Dev 2008; 3:12. [PMID: 18668998 PMCID: PMC2409322 DOI: 10.1186/1749-8104-3-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background In the adult abdomen of Drosophila, the shafts of mechanosensory bristles point consistently from anterior to posterior. This is an example of planar cell polarity (PCP); some genes responsible for PCP have been identified. Each adult bristle is made by a clone of four cells, including the neuron that innervates it, but little is known as to how far the formation or positions of these cells depends on PCP. The neurons include a single dendrite and an axon; it is not known whether the orientation of these processes is influenced by PCP. Results We describe the development of the abdominal mechanosensory bristles in detail. The division of the precursor cell gives two daughters, one (pIIa) divides to give rise to the bristle shaft and socket cell and the other (pIIb) generates the neuron, the sheath and the fifth cell. Although the bristles and their associated shaft and socket cells are consistently oriented, the positioning and behaviour of the neuron, the sheath and the fifth cell, as well as the orientation of the axons and the dendritic paths, depend on location. For example, in the anterior zone of the segment, the axons grow posteriorly, while in the posterior zone, they grow anteriorly. Manipulating the PCP genes can reverse bristle orientation, change the path taken by the dendrite and the position of the cell body of the neuron. However, the paths taken by the axon are not affected. Conclusion PCP genes, such as starry night and dachsous orient the bristles and position the neuronal cell body and affect the shape of the dendrites. However, these PCP genes do not appear to change the paths followed by the sensory axons, which must, therefore, be polarised by other factors.
Collapse
Affiliation(s)
- Caroline C G Fabre
- Department of Zoology, Downing St., Cambridge CB2 3EJ, UK, MRC Laboratory of Molecular Biology, Hills Rd., Cambridge CB2 OQH, UK.
| | | | | |
Collapse
|
188
|
Affiliation(s)
- L Ricci-Vitiani
- Department of Hematology and Oncology, Istituto Superiore di Sanitè, Rome, Italy
| | | | | | | | | |
Collapse
|
189
|
Abstract
Stem cells are captivating because they have the potential to make multiple cell types yet maintain their undifferentiated state. Recent studies of Drosophila and mammalian neural stem cells have shed light on how stem cells regulate self-renewal versus differentiation and have revealed the proteins, processes and pathways that all converge to regulate neural progenitor self-renewal. If we can better understand how stem cells balance self-renewal versus differentiation, we will significantly advance our knowledge of embryogenesis, cancer biology and brain evolution, as well as the use of stem cells for therapeutic purposes.
Collapse
Affiliation(s)
- Chris Q Doe
- Howard Hughes Medical Institute, Institute of Neuroscience, Institute of Molecular Biology, University of Oregon, Eugene, OR, USA.
| |
Collapse
|
190
|
Murata H, Tsuji S, Tsujii M, Nakamura T, Fu HY, Eguchi H, Asahi K, Okano H, Kawano S, Hayashi N. Helicobacter pylori infection induces candidate stem cell marker Musashi-1 in the human gastric epithelium. Dig Dis Sci 2008; 53:363-9. [PMID: 17549630 DOI: 10.1007/s10620-007-9858-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Accepted: 04/24/2007] [Indexed: 01/12/2023]
Abstract
Musashi-1 (Msi-1), a mammalian neural RNA-binding protein, has been found to play important roles in the maintenance of stem cell states and differentiation in neural stem cells and mouse intestinal cells. We explored Msi-1 expression and its potential implications in the human stomach. Reverse transcription-PCR revealed that Msi-1 levels were significantly higher in the corpus than in antrum in Helicobacter pylori (Hp)-infected patients (n = 49) (P < 0.00001) in paired biopsy samples, whereas they were low and comparable at these two sites in Hp-negative patients (n = 31). Msi-1 levels were significantly higher in the Hp-infected corpus (n = 107) than in the Hp-negative corpus (n = 69) (P < 0.00000001). Immunohistochemistry and in situ hybridization demonstrated that Msi-1 was expressed at the base and neck/isthmus region of the fundic glands and partly co-expressed in Ki-67-positive cells in the corpus and antrum. Msi-1 levels correlated with Hp density (P < 0.05). Based on these results, we conclude that Hp infection strongly induces Msi-1 in the corpus. Given its expression in dividing cells, Msi-1 may modulate the state of gastric progenitor cells.
Collapse
Affiliation(s)
- Hiroaki Murata
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2, Yamada-Oka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Abstract
Cancer stem cells (CSCs) resemble normal stem cells in several ways. Both cell types are self-renewing and when they divide, one of the daughter cells differentiates while the other retains stem cell properties, including the ability to divide in the same way again. CSCs have been demonstrated to exist in several solid tumors, including colon carcinoma; these cells are able to initiate and sustain tumor growth. There are essentially three different methods to isolate CSCs: establishment culture, the MACS (magnetic cell sorting) technology, and the FACS (fluorescence-activated cell sorting) technology.
Collapse
|
192
|
Yen TH, Wright NA. The gastrointestinal tract stem cell niche. ACTA ACUST UNITED AC 2007; 2:203-12. [PMID: 17625256 DOI: 10.1007/s12015-006-0048-1] [Citation(s) in RCA: 193] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/12/2022]
Abstract
The gastrointestinal epithelium is unique in that cell proliferation, differentiation, and apoptosis occur in an orderly fashion along the crypt-villus axis. The intestinal crypt is mainly a proliferative compartment, is monoclonal and is maintained by stem cells. The villus represents the differentiated compartment, and is polyclonal as it receives cells from multiple crypts. In the small intestine, cell migration begins near the base of the crypt, and cells migrate from here emerging onto the villi. The basal crypt cells at position 5 are candidate stem cells. As the function of stem cells is to maintain the integrity of the intestinal epithelium, it must self-renew, proliferate, and differentiate within a protective niche. This niche is made up of proliferating and differentiating epithelial cells and surrounding mesenchymal cells. These mesenchymal cells promote the epithelial- mesenchymal crosstalk required to maintain the niche. A stochastic model of cell division has been proposed to explain how a single common ancestral stem cell exists from which all stem cells in a niche are descended. Our group has argued that these crypts then clonally expand by crypt fission, forming two daughters' crypts, and that this is the mechanism by which mutated stem cells or even cancer stem cell clones expand in the colon and in the entire gastrointestinal tract. Until recently, the differentiation potential of stem cells into adult tissues has been thought to be limited to cell lineages in the organ from which they were derived. Bone marrow cells are rare among adult stem cells regarding their abundance and role in the continuous, lifelong, physiological replenishment of circulating cells. In human and mice experiments, we have shown that bone marrow can contribute to the regeneration of intestinal myofibroblasts and thereby after epithelium following damage, through replacing the cells, which maintain the stem cells niche. Little is known about the markers characterizing the stem and transit amplifying populations of the gastrointestinal tract, although musashi-1 and hairy and enhancer of split homolog-1 have been proposed. As the mammalian gastrointestinal tract develops from the embryonic gut, it is made up of an endodermally-derived epithelium surrounded by cells of mesoderm origin. Cell signaling between these two tissue layers plays a critical role in coordinating patterning and organogenesis of the gut and its derivatives. Many lines of evidence have revealed that Wnt signaling is the most dominant force in controlling cell proliferation, differentiation, and apoptosis along the crypt-villus axis. We have found Wnt messenger RNAs expression in intestinal subepithelial myofibroblasts and frizzled messenger RNAs expression in both myofibroblasts and crypt epithelium. Moreover, there are many other factors, for example, bone morphogenetic protein, homeobox, forkhead, hedgehog, homeodomain, and platelet-derived growth factor that are also important to stem cell signaling in the gastrointestinal tract.
Collapse
Affiliation(s)
- Tzung-Hai Yen
- Histopathology Unit, Cancer Research UK, London Research Institute, London, UK.
| | | |
Collapse
|
193
|
Chen YT, Li W, Hayashida Y, He H, Chen SY, Tseng DY, Kheirkhah A, Tseng SCG. Human amniotic epithelial cells as novel feeder layers for promoting ex vivo expansion of limbal epithelial progenitor cells. Stem Cells 2007; 25:1995-2005. [PMID: 17495107 PMCID: PMC3197019 DOI: 10.1634/stemcells.2006-0677] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Human amniotic epithelial cells (HAECs) are a unique embryonic cell source that potentially can be used as feeder layers for expanding different types of stem cells. In vivo, HAECs uniformly expressed pan-cytokeratins (pan-CK) and heterogeneously expressed vimentin (Vim). The two phenotypes expressing either pan-CK(+)/Vim(+) or pan-CK(+)/Vim(-) were maintained in serum-free media with high calcium. In contrast, all HAECs became pan-CK(+)/Vim(+) in serum-containing media, which also promoted HAEC proliferation for at least eight passages, especially supplemented with epidermal growth factor and insulin. Mitomycin C-arrested HAEC feeder layers were more effective in promoting clonal growth of human limbal epithelial progenitors than conventional 3T3 murine feeder layers. Cells in HAEC-supported clones were uniformly smaller, sustained more proliferation, and expressed less CK12 and connexin 43 but higher levels of stem cell-associated markers such as p63, Musashi-1, and ATP-binding cassette subfamily G2 than those of 3T3-supported clones. Subculturing of clonally expanded limbal progenitors from HAEC feeder layers, but not from 3T3 feeder layers, gave rise to uniformly p63-positive epithelial progenitor cells as well as nestin-positive neuronal-like progenitors. Collectively, these results indicated that HAECs can be used as a human feeder layer equivalent for more effective ex vivo expansion of adult epithelial stem cells from the human limbus. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Ying Ting Chen
- TissueTech, Inc. and Ocular Surface Center, Miami, Florida, USA
- Department of Ophthalmology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei Li
- TissueTech, Inc. and Ocular Surface Center, Miami, Florida, USA
| | | | - Hua He
- TissueTech, Inc. and Ocular Surface Center, Miami, Florida, USA
| | - Szu Yu Chen
- TissueTech, Inc. and Ocular Surface Center, Miami, Florida, USA
| | - David Y. Tseng
- TissueTech, Inc. and Ocular Surface Center, Miami, Florida, USA
| | - Ahmad Kheirkhah
- TissueTech, Inc. and Ocular Surface Center, Miami, Florida, USA
| | | |
Collapse
|
194
|
Watanabe K, Kondo K, Takeuchi N, Okano H, Yamasoba T. Musashi-1 expression in postnatal mouse olfactory epithelium. Neuroreport 2007; 18:641-4. [PMID: 17426590 DOI: 10.1097/wnr.0b013e3280bef7e2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We investigated the age-related change in the distribution of a molecular marker for neural stem and precursor cells, Musashi-1, in the olfactory epithelium of mice from 1 day up to 16 months of age using immunohistochemistry. We also compared the distribution pattern of Musashi-1 with that of growth-associated protein 43, the olfactory marker protein, and Notch-1. Musashi-1 was expressed in the globose basal cell layer and the lower part of the growth-associated protein 43-positive layer, with immunoreactivity decreasing with aging. Notch-1 was observed only in the early postnatal period. These findings are consistent with the fact that globose basal cells are proliferating olfactory precursor cells and that their ability to generate new neurons decreases with aging.
Collapse
Affiliation(s)
- Kenta Watanabe
- Department of Otolaryngology, Faculty of Medicine, University of Tokyo, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
195
|
Kanai R, Eguchi K, Takahashi M, Goldman S, Okano H, Kawase T, Yazaki T. Enhanced therapeutic efficacy of oncolytic herpes vector G207 against human non-small cell lung cancer--expression of an RNA-binding protein, Musashi1, as a marker for the tailored gene therapy. J Gene Med 2007; 8:1329-40. [PMID: 16955534 DOI: 10.1002/jgm.965] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Oncolytic herpes vectors like G207 have shown considerable promise in the treatment of solid tumors, but their potency must be enhanced for the full achievement of therapeutic efficacy. Deletion of the innate gamma34.5 gene made these vectors extremely safe, but their efficacy was also severely attenuated. Use of tumor-specific promoters is one method to direct toxicity and enhance efficacy against tumors. Recently, Musashi1 has been shown expressed in some tumor tissues. METHODS Eleven human cancer cell lines including five non-small cell lung cancers (NSCLCs) were investigated. Musashi1 mRNA expression was examined by RT-PCR analysis. Western blotting was also performed. Transcriptional activity of P/musashi1 in NSCLCs was assayed by GFP reporter plasmids. Then we constructed a defective amplicon vector containing musashi1 promoter/ICP34.5 with G207 as helper virus (dvM345). In vitro cytotoxicity against NSCLCs and growth characteristics of helper virus were examined. A Lu-99 subcutaneous tumor model was used in an animal study. The tumor volume treated with G207 alone or dvM345 was measured. RESULTS Musashi1 mRNA was detected in four cell lines. Two in five NSCLCs were positive, and P/musashi1 was proved functional within them. Against these cell lines, dvM345 showed enhanced cytotoxicity, and helper viral growth was augmented. A subcutaneous tumor study confirmed the enhanced therapeutic efficacy of G207 by dvM345 without compromising safety. CONCLUSIONS These results suggest that Musashi1 might be involved in the development of several carcinomas including NSCLC. In the context of oncolytic herpes vector strategy, the P/musashi1-ICP34.5 method could be used for the treatment of cancers expressing Musashi1.
Collapse
MESH Headings
- Animals
- Base Sequence
- Biomarkers, Tumor/genetics
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/therapy
- Cell Line, Tumor
- Female
- Genetic Therapy/methods
- Genetic Vectors
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/therapy
- Mice
- Mice, Nude
- Neoplasm Transplantation
- Nerve Tissue Proteins/genetics
- Oncolytic Virotherapy/methods
- Oncolytic Viruses/genetics
- Promoter Regions, Genetic
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- RNA-Binding Proteins/genetics
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Ryuichi Kanai
- Molecular Neurosurgery Laboratory, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
196
|
Nakano A, Kanemura Y, Mori K, Kodama E, Yamamoto A, Sakamoto H, Nakamura Y, Okano H, Yamasaki M, Arita N. Expression of the Neural RNA-binding protein Musashi1 in pediatric brain tumors. Pediatr Neurosurg 2007; 43:279-84. [PMID: 17627143 DOI: 10.1159/000103307] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Accepted: 11/29/2006] [Indexed: 12/29/2022]
Abstract
Musashi1 (MSI1) is an evolutionarily conserved RNA-binding protein, selectively expressed in neural stem cells (NSCs) and considered a versatile marker for normal NSCs and tumor cell diagnosis. Here, we examined MSI1 expression in primary pediatric brain tumors, medulloblastomas and ependymomas, by double immunostaining with lineage phenotypic markers (Lin). These tumors highly express MSI1 and are heterogeneous, containing both MSI1+/Lin- tumor cells in regions of relatively high cellularity and proliferative activity and MSI1+/Lin+ tumor cells in regions of lower cellularity. These findings suggest that MSI1 may be a useful marker for characterizing tumor heterogeneity and for examining in situ the analogy between normal NSCs and MSI1+ cells in pediatric brain tumors. This test could be easily applied to routine clinical diagnosis.
Collapse
Affiliation(s)
- Aya Nakano
- Department of Neurosurgery, Hyogo College of Medicine, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
McDonald SAC, Preston SL, Lovell MJ, Wright NA, Jankowski JAZ. Mechanisms of disease: from stem cells to colorectal cancer. ACTA ACUST UNITED AC 2006; 3:267-74. [PMID: 16673006 DOI: 10.1038/ncpgasthep0473] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Accepted: 02/10/2006] [Indexed: 12/12/2022]
Abstract
Over the past decade, the advances in our understanding of stem cell biology and the role of stem cells in diseases, such as colorectal cancer, have been remarkable. In particular, discoveries related to the control of stem cell proliferation and how dysregulation of proliferation leads to oncogenesis have been foremost. For intestinal stem cells, the WNT family of growth factors, and events such as the regulation of the nuclear localization of beta-catenin, seem to be central to normal homeostasis, and mutations in the components of these pathways seem to lead to the development of colorectal cancer. A paradigm of abnormal stem cell biology is illustrated by patients with familial adenomatous polyposis, who have mutations in the adenomatous polyposis coli gene. The wild-type protein encoded by this gene is important for the prevention of mass beta-catenin accumulation in the nucleus and the subsequent overtranscription of cell cycle proteins. This review discusses the basic mechanisms behind stem cell regulation in the gut and follows their role in the natural history of tumor progression.
Collapse
Affiliation(s)
- Stuart A C McDonald
- Histopathology Unit at the London Research Institute, Cancer Research UK, London, UK.
| | | | | | | | | |
Collapse
|
198
|
Fukui T, Takeda H, Shu HJ, Ishihama K, Otake S, Suzuki Y, Nishise S, Ito N, Sato T, Togashi H, Kawata S. Investigation of Musashi-1 expressing cells in the murine model of dextran sodium sulfate-induced colitis. Dig Dis Sci 2006; 51:1260-8. [PMID: 16944023 DOI: 10.1007/s10620-006-8046-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2005] [Accepted: 07/27/2005] [Indexed: 01/12/2023]
Abstract
Musashi-1 (Msi-1), an RNA-binding protein, had been proposed to be a specific marker for neural stem/precursor cells. Msi-1 expressing cells in the intestinal epithelium are also strongly considered as potential stem/precursor cells. To clarify the behavior of those cells in the injury or regeneration phase, we investigated Msi-1 expressing cells of intestinal mucosa in the murine model of dextran sodium sulfate (DSS)-induced colitis. Immunohistochemically, Msi-1-positive cells were found in the area just along the layer of Paneth's cells in the small intestine and in the bottom layer of crypts in the large intestine. During DSS administration, the number of PCNA-positive cells in the large intestine increased markedly. In contrast, the number of Msi-1-positive cells decreased slightly with DSS but returned to normal after DSS administration was stopped. The level of mRNA for Msi-1 was consistent with the result of immunohistochemical examinations. Conclusively, we could describe the behavior of intestinal stem/precursor cells during inflammation using Msi-1.
Collapse
Affiliation(s)
- Tadahisa Fukui
- Department of Gastroenterology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata, Japan, 990-9585
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Ratti A, Fallini C, Cova L, Fantozzi R, Calzarossa C, Zennaro E, Pascale A, Quattrone A, Silani V. A role for the ELAV RNA-binding proteins in neural stem cells: stabilization of Msi1 mRNA. J Cell Sci 2006; 119:1442-52. [PMID: 16554442 DOI: 10.1242/jcs.02852] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Post-transcriptional regulation exerted by neural-specific RNA-binding proteins plays a pivotal role in the development and maintenance of the nervous system. Neural ELAV proteins are key inducers of neuronal differentiation through the stabilization and/or translational enhancement of target transcripts bearing the AU-rich elements (AREs), whereas Musashi-1 maintains the stem cell proliferation state by acting as a translational repressor. Since the gene encoding Musashi-1 (Msi1) contains a conserved ARE in its 3' untranslated region, we focused on the possibility of a mechanistic relationship between ELAV proteins and Musashi-1 in cell fate commitment. Colocalization of neural ELAV proteins with Musashi-1 clearly shows that ELAV proteins are expressed at early stages of neural commitment, whereas interaction studies demonstrate that neural ELAV proteins exert an ARE-dependent binding activity on the Msi1 mRNA. This binding activity has functional effects, since the ELAV protein family member HuD is able to stabilize the Msi1 ARE-containing mRNA in a sequence-dependent way in a deadenylation/degradation assay. Furthermore activation of the neural ELAV proteins by phorbol esters in human SH-SY5Y cells is associated with an increase of Musashi-1 protein content in the cytoskeleton. We propose that ELAV RNA-binding proteins exert an important post-transcriptional control on Musashi-1 expression in the transition from proliferation to neural differentiation of stem/progenitor cells.
Collapse
Affiliation(s)
- Antonia Ratti
- Department of Neuroscience, Dino Ferrari Centre, University of Milan-IRCCS Istituto Auxologico Italiano, Via Zucchi 18, 20095 Cusano Milanino, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Siddall NA, McLaughlin EA, Marriner NL, Hime GR. The RNA-binding protein Musashi is required intrinsically to maintain stem cell identity. Proc Natl Acad Sci U S A 2006; 103:8402-7. [PMID: 16717192 PMCID: PMC1570104 DOI: 10.1073/pnas.0600906103] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
A key goal of regenerative medicine is an understanding of the genetic factors that define the properties of stem cells. However, stem cell research in mammalian tissue has been hampered by a paucity of stem cell-specific markers. Although increasing evidence suggests that members of the Musashi (Msi) family of RNA-binding proteins play important functions in progenitor cells, it remains unclear whether there is a stem cell-autonomous requirement for Msi because of an inability to distinguish stem cells from early-lineage cells in mammalian tissues. Here, using the Drosophila testis as a model system for the study of stem cell regulation, we show specific evidence for a cell-autonomous requirement for Msi family proteins in regulating stem cell differentiation, leading to the identification of an RNA-binding protein required for spermatogonial stem cell maintenance. We found that loss of Msi function disrupts the balance between germ-line stem cell renewal and differentiation, resulting in the premature differentiation of germ-line stem cells. Moreover, we found that, although Msi is expressed in both somatic and germ cells, Msi function is required intrinsically in stem cells for maintenance of stem cell identity. We also discovered a requirement for Msi function in male meiosis, revealing that Msi has distinct roles at different stages of germ cell differentiation. We describe the complementary expression patterns of the murine Msi paralogues Msi1 and Msi2 during spermatogenesis, which support the idea of distinct, evolutionarily conserved roles of Msi.
Collapse
Affiliation(s)
- Nicole A. Siddall
- *Department of Anatomy and Cell Biology, University of Melbourne, Melbourne VIC 3010, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan NSW 2308, Australia; and
| | - Eileen A. McLaughlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan NSW 2308, Australia; and
- Australian Research Council Centre of Excellence in Biotechnology and Development, Callaghan NSW 2308, Australia
| | - Neisha L. Marriner
- *Department of Anatomy and Cell Biology, University of Melbourne, Melbourne VIC 3010, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan NSW 2308, Australia; and
| | - Gary R. Hime
- *Department of Anatomy and Cell Biology, University of Melbourne, Melbourne VIC 3010, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan NSW 2308, Australia; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|