551
|
Bera TK, Saint Fleur A, Ha D, Yamada M, Lee Y, Lee B, Hahn Y, Kaufman DS, Pera M, Pastan I. Selective POTE paralogs on chromosome 2 are expressed in human embryonic stem cells. Stem Cells Dev 2008; 17:325-32. [PMID: 18447647 DOI: 10.1089/scd.2007.0079] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
POTE is a primate-specific gene family that encodes cancer testis antigens that contain three domains, although the proteins vary greatly in size. The amino-terminal domain is novel and has three cysteine-rich domains of 37 amino acids. The second and third domains are rich in ankyrin repeats and spectrin-like helices respectively. In humans, 13 highly homologous paralogs are dispersed among eight chromosomes. Some members of the POTE gene family have an actin insertion at the carboxyl end of the protein. The expression of the POTE gene in normal adult tissues is restricted, but several POTE paralogs are frequently expressed in many cancers including breast, prostate, and lung cancers. We show here that POTE is expressed in several human embryonic stem (ES) cell lines. We found that UC06, WA01 and ES03 cell lines express mainly a POTE-2gamma transcript but ES02 and ES04 cell lines predominantly express POTE-2alpha. The WA09 cell line expressed both POTE-2gamma and POTE-2alpha. There is no detectable POTE gene expression in fetal tissues (ages 16-36 weeks). The POTE paralogs that are expressed in ES cells may have a specific function during lineage-specific differentiation of ES cells.
Collapse
Affiliation(s)
- Tapan K Bera
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
552
|
Rajan P, Srinivasan R. Targeting cancer stem cells in cancer prevention and therapy. ACTA ACUST UNITED AC 2008; 4:211-6. [PMID: 18663609 DOI: 10.1007/s12015-008-9037-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2008] [Indexed: 12/17/2022]
Abstract
The cancer stem cell hypothesis is an attractive framework within which one may think about cancer initiation, recurrence, and metastasis, and methods to devise treatment strategies for cancers. Although all cancers do not appear to sustain themselves with cancer stem cells, but also through a dominant cell population, creating strategies for cancer treatment which include cancer stem cells as targets seems reasonable. In this perspective we discuss possible strategies for controlling the viability and tumorigenecity of cancer stem cells, and extend our discussion to strategies approaching the prevention of cancer.
Collapse
Affiliation(s)
- Prithi Rajan
- Center for Stem Cell Research, Christian Medical College, Vellore, 632002, India,
| | | |
Collapse
|
553
|
Wu JM, Fackler MJ, Halushka MK, Molavi DW, Taylor ME, Teo WW, Griffin C, Fetting J, Davidson NE, De Marzo AM, Hicks JL, Chitale D, Ladanyi M, Sukumar S, Argani P. Heterogeneity of breast cancer metastases: comparison of therapeutic target expression and promoter methylation between primary tumors and their multifocal metastases. Clin Cancer Res 2008; 14:1938-46. [PMID: 18381931 DOI: 10.1158/1078-0432.ccr-07-4082] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE A comprehensive comparison of biomarker expression between patients' primary breast carcinoma (PBC) and their metastatic breast carcinomas (MBC) has not been done. EXPERIMENTAL DESIGN We did rapid autopsies (postmortem intervals, 1-4 hours) on 10 consenting patients who died of MBC. We constructed single-patient tissue microarrays from the patients' archived PBC and multiple different MBCs harvested at autopsy, which were immunohistochemically labeled for multiple biomarkers. Methylation of multiple gene promoters was assessed quantitatively on dissected PBC and MBC samples. RESULTS Extensive heterogeneity was observed between PBC and their paired MBC, as well as among multiple MBC from the same patient. Estrogen and progesterone receptors tended to be uniformly down-regulated in metastases. E-cadherin was down-regulated in a subset of the MBC of one case. Variable overexpression in MBC compared with the PBC was observed for cyclooxygenase-2 (five cases), epidermal growth factor receptor (EGFR; four cases), MET (four cases), and mesothelin (four cases). No case strongly overexpressed HER-2/neu by immunohistochemistry, but eight cases showed variable protein expression ranging from negative to equivocal (2+) in different MBC. In one case, variable low-level HER-2/neu gene amplification was found. EGFR and MET overexpression were restricted to the four basal-type cancers. EGFR protein overexpression did not correlate with EGFR gene amplification. Multigene promoter hypermethylation of RASSF1a, HIN1, cyclin D2, Twist, estrogen receptor alpha, APC1, and RARbeta was overall very similar in the PBC and all MBCs in all cases. CONCLUSIONS Therapeutic targets identified in the PBC or even some MBC may not reflect targets present in all metastatic sites.
Collapse
Affiliation(s)
- Julie M Wu
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
554
|
Abstract
Appropriate treatment of head and neck squamous cell cancer (HNSCC) remains one of the most difficult challenges in head and neck oncology. Overall survival of patients with HNSCC remains at approximately 50% at 5 years. Surgical therapy can be mutilating and often has significant effects on swallowing, speech, and physical appearance. The addition of chemotherapy to radiation treatment has shown efficacy in organ preservation in some sites in the head and neck, but has resulted in limited improvement in survival rates. HNSCC resistance to chemotherapy has limited the usefulness of chemotherapy in the treatment of this disease. We have recently demonstrated that human head and neck squamous cell cancers contain a tumorigenic, so-called cancer stem cell, subpopulation of cells that can self-renew and produce differentiated cells that form the bulk of the tumor. These tumorigenic HNSCC cells have a distinct phenotype and can be identified by a surface marker. Current treatment for HNSCC regimens may selectively kill the differentiated cancer cells, producing tumor regression while sparing the cancer stem cells, leading to tumor regrowth and relapse. It is important for us to understand why HNSCC does not respond to chemotherapy and to identify new targeted treatments that can overcome resistance and improve patient outcomes. Further study of HNSCC stem cells will increase our knowledge of this devastating disease and allow us to develop novel treatments.
Collapse
Affiliation(s)
- Mark E P Prince
- University of Michigan, Otolaryngology-HNS, 1904 Taubman Center, 1500 East Medical Center Dr, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
555
|
Dylla SJ, Beviglia L, Park IK, Chartier C, Raval J, Ngan L, Pickell K, Aguilar J, Lazetic S, Smith-Berdan S, Clarke MF, Hoey T, Lewicki J, Gurney AL. Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy. PLoS One 2008; 3:e2428. [PMID: 18560594 PMCID: PMC2413402 DOI: 10.1371/journal.pone.0002428] [Citation(s) in RCA: 432] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 05/08/2008] [Indexed: 12/11/2022] Open
Abstract
Background Patients generally die of cancer after the failure of current therapies to eliminate residual disease. A subpopulation of tumor cells, termed cancer stem cells (CSC), appears uniquely able to fuel the growth of phenotypically and histologically diverse tumors. It has been proposed, therefore, that failure to effectively treat cancer may in part be due to preferential resistance of these CSC to chemotherapeutic agents. The subpopulation of human colorectal tumor cells with an ESA+CD44+ phenotype are uniquely responsible for tumorigenesis and have the capacity to generate heterogeneous tumors in a xenograft setting (i.e. CoCSC). We hypothesized that if non-tumorigenic cells are more susceptible to chemotherapeutic agents, then residual tumors might be expected to contain a higher frequency of CoCSC. Methods and Findings Xenogeneic tumors initiated with CoCSC were allowed to reach ∼400 mm3, at which point mice were randomized and chemotherapeutic regimens involving cyclophosphamide or Irinotecan were initiated. Data from individual tumor phenotypic analysis and serial transplants performed in limiting dilution show that residual tumors are enriched for cells with the CoCSC phenotype and have increased tumorigenic cell frequency. Moreover, the inherent ability of residual CoCSC to generate tumors appears preserved. Aldehyde dehydrogenase 1 gene expression and enzymatic activity are elevated in CoCSC and using an in vitro culture system that maintains CoCSC as demonstrated by serial transplants and lentiviral marking of single cell-derived clones, we further show that ALDH1 enzymatic activity is a major mediator of resistance to cyclophosphamide: a classical chemotherapeutic agent. Conclusions CoCSC are enriched in colon tumors following chemotherapy and remain capable of rapidly regenerating tumors from which they originated. By focusing on the biology of CoCSC, major resistance mechanisms to specific chemotherapeutic agents can be attributed to specific genes, thereby suggesting avenues for improving cancer therapy.
Collapse
Affiliation(s)
- Scott J Dylla
- OncoMed Pharmaceuticals Inc., Redwood City, California, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
556
|
Bmi1 is expressed in vivo in intestinal stem cells. Nat Genet 2008; 40:915-20. [PMID: 18536716 DOI: 10.1038/ng.165] [Citation(s) in RCA: 956] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 04/02/2008] [Indexed: 12/17/2022]
Abstract
Bmi1 plays an essential part in the self-renewal of hematopoietic and neural stem cells. To investigate its role in other adult stem cell populations, we generated a mouse expressing a tamoxifen-inducible Cre from the Bmi1 locus. We found that Bmi1 is expressed in discrete cells located near the bottom of crypts in the small intestine, predominantly four cells above the base of the crypt (+4 position). Over time, these cells proliferate, expand, self-renew and give rise to all the differentiated cell lineages of the small intestine epithelium. The induction of a stable form of beta-catenin in these cells was sufficient to rapidly generate adenomas. Moreover, ablation of Bmi1(+) cells using a Rosa26 conditional allele, expressing diphtheria toxin, led to crypt loss. These experiments identify Bmi1 as an intestinal stem cell marker in vivo. Unexpectedly, the distribution of Bmi1-expressing stem cells along the length of the small intestine suggested that mammals use more than one molecularly distinguishable adult stem cell subpopulation to maintain organ homeostasis.
Collapse
|
557
|
Mei-P26 regulates microRNAs and cell growth in the Drosophila ovarian stem cell lineage. Nature 2008; 454:241-5. [PMID: 18528333 DOI: 10.1038/nature07014] [Citation(s) in RCA: 193] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2008] [Accepted: 04/03/2008] [Indexed: 12/11/2022]
Abstract
Drosophila neuroblasts and ovarian stem cells are well characterized models for stem cell biology. In both cell types, one daughter cell self-renews continuously while the other undergoes a limited number of divisions, stops to proliferate mitotically and differentiates. Whereas neuroblasts segregate the Trim-NHL (tripartite motif and Ncl-1, HT2A and Lin-41 domain)-containing protein Brain tumour (Brat) into one of the two daughter cells, ovarian stem cells are regulated by an extracellular signal from the surrounding stem cell niche. After division, one daughter cell looses niche contact. It undergoes 4 transit-amplifying divisions to form a cyst of 16 interconnected cells that reduce their rate of growth and stop to proliferate mitotically. Here we show that the Trim-NHL protein Mei-P26 (refs 7, 8) restricts growth and proliferation in the ovarian stem cell lineage. Mei-P26 expression is low in stem cells but is strongly induced in 16-cell cysts. In mei-P26 mutants, transit-amplifying cells are larger and proliferate indefinitely leading to the formation of an ovarian tumour. Like brat, mei-P26 regulates nucleolar size and can induce differentiation in Drosophila neuroblasts, suggesting that these genes act through the same pathway. We identify Argonaute-1, a component of the RISC complex, as a common binding partner of Brat and Mei-P26, and show that Mei-P26 acts by inhibiting the microRNA pathway. Mei-P26 and Brat have a similar domain composition that is also found in other tumour suppressors and might be a defining property of a new family of microRNA regulators that act specifically in stem cell lineages.
Collapse
|
558
|
Dietrich J, Imitola J, Kesari S. Mechanisms of Disease: the role of stem cells in the biology and treatment of gliomas. ACTA ACUST UNITED AC 2008; 5:393-404. [PMID: 18521117 DOI: 10.1038/ncponc1132] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 11/01/2007] [Indexed: 12/26/2022]
Abstract
The study of neural stem cell and progenitor cell biology has improved our understanding of the biology of brain tumors in a developmental context. Recent work has demonstrated that brain tumors may harbor small subpopulations of cells that share characteristics of neural stem cells. There is still an ongoing debate about the specific role of these stem-like cells in cancer initiation, development and progression. Nonetheless, the concept of cancer stem cells has offered a new paradigm to understand tumor biology and resistance to current treatment modalities. Molecular aberrations in these cancer stem cells might be crucial targets for therapeutic intervention, with the hope of achieving more durable clinical responses. Recent studies have demonstrated that endogenous and transplanted neural stem cells and progenitor cells show a marked tropism to brain tumors. Although the mechanisms that govern these processes are poorly understood, the use of neural stem cells and progenitor cells as delivery vehicles for molecules toxic to tumors offers a promising experimental treatment strategy. This Review summarizes recent advances in the basic understanding of neural stem cell and cancer stem cell biology and the progress towards translating these novel concepts into the clinic.
Collapse
Affiliation(s)
- Jörg Dietrich
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
559
|
Affiliation(s)
- Konrad L Streetz
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | | |
Collapse
|
560
|
|
561
|
Halvorsen OJ. Molecular and prognostic markers in prostate cancer. APMIS 2008. [DOI: 10.1111/j.1600-0463.2008.0s123.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
562
|
Wong DJ, Liu H, Ridky TW, Cassarino D, Segal E, Chang HY. Module map of stem cell genes guides creation of epithelial cancer stem cells. Cell Stem Cell 2008; 2:333-44. [PMID: 18397753 DOI: 10.1016/j.stem.2008.02.009] [Citation(s) in RCA: 569] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Revised: 01/18/2008] [Accepted: 02/12/2008] [Indexed: 10/22/2022]
Abstract
Self-renewal is a hallmark of stem cells and cancer, but existence of a shared stemness program remains controversial. Here, we construct a gene module map to systematically relate transcriptional programs in embryonic stem cells (ESCs), adult tissue stem cells, and human cancers. This map reveals two predominant gene modules that distinguish ESCs and adult tissue stem cells. The ESC-like transcriptional program is activated in diverse human epithelial cancers and strongly predicts metastasis and death. c-Myc, but not other oncogenes, is sufficient to reactivate the ESC-like program in normal and cancer cells. In primary human keratinocytes transformed by Ras and I kappa B alpha, c-Myc increases the fraction of tumor-initiating cells by 150-fold, enabling tumor formation and serial propagation with as few as 500 cells. c-Myc-enhanced tumor initiation is cell-autonomous and independent of genomic instability. Thus, activation of an ESC-like transcriptional program in differentiated adult cells may induce pathologic self-renewal characteristic of cancer stem cells.
Collapse
Affiliation(s)
- David J Wong
- Program in Epithelial Biology, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
563
|
Götz R. Inter-cellular adhesion disruption and the RAS/RAF and beta-catenin signalling in lung cancer progression. Cancer Cell Int 2008; 8:7. [PMID: 18492263 PMCID: PMC2427011 DOI: 10.1186/1475-2867-8-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 05/20/2008] [Indexed: 12/30/2022] Open
Abstract
Cadherin cell adhesion molecules play an essential role in creating tight intercellular association and their loss has been correlated with poor prognosis in human cancer. Mutational activation of protein kinases and loss of cell adhesion occur together in human lung adenocarcinoma but how these two pathways interconnect is only poorly understood. Mouse models of human lung adenocarcinoma with oncogene expression targeted to subtypes of lung epithelial cells led to formation of adenomas or adenocarcinomas that lacked metastatic potential. Conditional genetic abrogation of epithelial tumour cell adhesion in mice with benign lung tumours induced by oncogenic RAF kinase has been demonstrated to induce intratumourous vascularization (angiogenic switch), progression to invasive adenocarcinoma and micrometastasis. Importantly, breaking cell adhesion in benign oncogene-driven lung tumour cells activated beta-catenin signalling and induced the expression of several genes that are normally expressed in intestine rather than the lung. I will discuss potential routes to nuclear beta-catenin signalling in cancer and how nuclear beta-catenin may epigenetically alter the plasticity of tumour cells during malignant progression.
Collapse
Affiliation(s)
- Rudolf Götz
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), Universität Würzburg, Germany.
| |
Collapse
|
564
|
Gilbertson RJ, Ellison DW. The origins of medulloblastoma subtypes. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2008; 3:341-65. [PMID: 18039127 DOI: 10.1146/annurev.pathmechdis.3.121806.151518] [Citation(s) in RCA: 208] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Childhood tumors containing cells that are morphologically and functionally similar to normal progenitor cells provide fertile ground for investigating the links between development and cancer. In this respect, integrated studies of normal cerebellar development and the medulloblastoma, a malignant embryonal tumor of the cerebellum, have proven especially fruitful. Emerging evidence indicates that the different precursor cell populations that form the cerebellum and the cell signaling pathways that regulate its development likely represent distinct compartments from which the various subtypes of medulloblastoma arise. Definitive characterization of each medulloblastoma subtype will undoubtedly improve treatment of this disease and provide important insights to the origins of cancer.
Collapse
Affiliation(s)
- Richard J Gilbertson
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | |
Collapse
|
565
|
Wu X, Gong Y, Yue J, Qiang B, Yuan J, Peng X. Cooperation between EZH2, NSPc1-mediated histone H2A ubiquitination and Dnmt1 in HOX gene silencing. Nucleic Acids Res 2008; 36:3590-9. [PMID: 18460542 PMCID: PMC2441805 DOI: 10.1093/nar/gkn243] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
An intricate interplay between DNA methylation and polycomb-mediated gene silencing has been highlighted recently. Here we provided evidence that Nervous System Polycomb 1 (NSPc1), a BMI1 homologous polycomb protein, plays important roles in promoting H2A ubiquitination and cooperates with DNA methylation in HOX gene silencing. We showed that NSPc1 stimulates H2A ubiquitination in vivo and in vitro through direct interaction with both RING2 and H2A. RT-PCR analysis revealed that loss of NSPc1, EZH2 or DNA methyltransferase 1 (Dnmt1), or inhibition of DNA methylation in HeLa cells de-represses the expression of HOXA7. Chromatin immunoprecipitation (ChIP) assays demonstrated that NSPc1, EZH2 and Dnmt1 bind to the promoter of HOXA7, which is frequently hypermethylated in tumors. Knockdown of NSPc1 results in significant reduction of H2A ubiquitination and DNA demethylation as well as Dnmt1 dissociation in the HOXA7 promoter. Meanwhile Dnmt1 deficiency affects NSPc1 recruitment and H2A ubiquitination, whereas on both cases EZH2-mediated H3K27 trimethylation remains unaffected. When EZH2 was depleted, however, NSPc1 and Dnmt1 enrichment was abolished concomitant with local reduction of H3K27 trimethylation, H2A ubiquitination and DNA methylation. Taken together, our findings indicated that NSPc1-mediated H2A ubiquitination and DNA methylation, both being directed by EZH2, are interdependent in long-term target gene silencing within cancer cells.
Collapse
Affiliation(s)
- Xudong Wu
- School of Basic Medicine, Peking Union Medical College; National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China
| | | | | | | | | | | |
Collapse
|
566
|
Shafee N, Smith CR, Wei S, Kim Y, Mills GB, Hortobagyi GN, Stanbridge EJ, Lee EYHP. Cancer stem cells contribute to cisplatin resistance in Brca1/p53-mediated mouse mammary tumors. Cancer Res 2008; 68:3243-50. [PMID: 18451150 PMCID: PMC2929908 DOI: 10.1158/0008-5472.can-07-5480] [Citation(s) in RCA: 227] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The majority of BRCA1-associated breast cancers are basal cell-like, which is associated with a poor outcome. Using a spontaneous mouse mammary tumor model, we show that platinum compounds, which generate DNA breaks during the repair process, are more effective than doxorubicin in Brca1/p53-mutated tumors. At 0.5 mg/kg of daily cisplatin treatment, 80% primary tumors (n = 8) show complete pathologic response. At greater dosages, 100% show complete response (n = 19). However, after 2 to 3 months of complete remission following platinum treatment, tumors relapse and become refractory to successive rounds of treatment. Approximately 3.8% to 8.0% (mean, 5.9%) of tumor cells express the normal mammary stem cell markers, CD29(hi)24(med), and these cells are tumorigenic, whereas CD29(med)24(-/lo) and CD29(med)24(hi) cells have diminished tumorigenicity or are nontumorigenic, respectively. In partially platinum-responsive primary transplants, 6.6% to 11.0% (mean, 8.8%) tumor cells are CD29(hi)24(med); these populations significantly increase to 16.5% to 29.2% (mean, 22.8%; P < 0.05) in platinum-refractory secondary tumor transplants. Further, refractory tumor cells have greater colony-forming ability than the primary transplant-derived cells in the presence of cisplatin. Expression of a normal stem cell marker, Nanog, is decreased in the CD29(hi)24(med) populations in the secondary transplants. Top2A expression is also down-regulated in secondary drug-resistant tumor populations and, in one case, was accompanied by genomic deletion of Top2A. These studies identify distinct cancer cell populations for therapeutic targeting in breast cancer and implicate clonal evolution and expansion of cancer stem-like cells as a potential cause of chemoresistance.
Collapse
Affiliation(s)
- Norazizah Shafee
- Department of Microbiology and Molecular Genetics, College of Medicine, University of California, Irvine, California
| | - Christopher R. Smith
- Department of Biological Chemistry and Developmental and Cell Biology, College of Medicine, University of California, Irvine, California
| | - Shuanzeng Wei
- Department of Biological Chemistry and Developmental and Cell Biology, College of Medicine, University of California, Irvine, California
| | - Yoon Kim
- Department of Biological Chemistry and Developmental and Cell Biology, College of Medicine, University of California, Irvine, California
| | - Gordon B. Mills
- Department of System Biology, University of Texas M. D. Anderson Cancer Center, Houston, Texas,Department of Breast Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Gabriel N. Hortobagyi
- Department of Breast Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Eric J. Stanbridge
- Department of Microbiology and Molecular Genetics, College of Medicine, University of California, Irvine, California
| | - Eva Y-H. P. Lee
- Department of Biological Chemistry and Developmental and Cell Biology, College of Medicine, University of California, Irvine, California
| |
Collapse
|
567
|
Long-term haematopoietic reconstitution by Trp53-/-p16Ink4a-/-p19Arf-/- multipotent progenitors. Nature 2008; 453:228-32. [DOI: 10.1038/nature06869] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Accepted: 02/25/2008] [Indexed: 01/05/2023]
|
568
|
Fehm T, Müller V, Alix-Panabières C, Pantel K. Micrometastatic spread in breast cancer: detection, molecular characterization and clinical relevance. Breast Cancer Res 2008; 10 Suppl 1:S1. [PMID: 19091005 PMCID: PMC2605098 DOI: 10.1186/bcr1869] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Immunocytochemical or molecular assays allow the detection of single disseminated tumor cells (DTCs) in the bone marrow (BM) or the peripheral blood in 10% to 60% of breast cancer patients without signs of metastasis. Results from recently reported studies suggest that circulating tumor cell (CTC) levels may serve as a prognostic marker and be used for early assessment of therapeutic response in patients with metastatic breast cancer. In early stage breast cancer, however, the impact of CTCs is less well established than that of DTCs in BM, where several clinical studies demonstrated that such cells are an independent prognostic factor at primary diagnosis. The characterization of DTCs/CTCs has already shed new light on the complex process underlying early tumor cell dissemination and metastatic progression in cancer patients. Characterization of DTCs should help to identify novel targets for biological therapies aimed to prevent metastatic relapse. In addition, understanding tumor 'dormancy' and identifying metastatic stem cells might result in the development of new therapeutic concepts.
Collapse
Affiliation(s)
- Tanja Fehm
- Department of Gynecology and Obstetrics, University Tübingen, Tübingen, Germany
| | | | | | | |
Collapse
|
569
|
Integrating the biological characteristics of oncolytic viruses and immune cells can optimize therapeutic benefits of cell-based delivery. Gene Ther 2008; 15:753-8. [PMID: 18356814 DOI: 10.1038/gt.2008.42] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite significant advances in the development of tumor-selective agents, strategies for effective delivery of these agents across biological barriers to cells within the tumor microenvironment has been limiting. One tactical approach to overcoming biological barriers is to use cells as delivery vehicles, and a variety of different cell types have been investigated with a range of agents. In addition to transporting agents with targeted delivery, cells can also produce their own tumoricidal effect, conceal a payload from an immune response, amplify a selective agent at the target site and facilitate an antitumor immune response. We have reported a therapeutic combination consisting of cytokine induced killer cells and an oncolytic vaccinia virus with many of these features that led to therapeutic synergy in animal models of human cancer. The synergy was due to the interaction of the two agents to enhance the antitumor benefits of each individual component. As both of these agents display broad tumor-targeting potential and possess unique tumor killing mechanisms, together they were able to recognize and destroy a far greater number of malignant cells within the heterogeneous tumor than either agent alone. Effective cancer therapy will require recognition and elimination of the root of the disease, the cancer stem cell, and the combination of CIK cells and oncolytic vaccinia viruses has this potential. To create effective tumor-selective agents the viruses are modified to take advantage of the unique biology of the cancer cell. Similarly, if we are to develop targeted therapies that are sufficiently multifaceted to eliminate cancer cells at all stages of disease, we should integrate the virus into the unique biology of the cell delivery vehicle.
Collapse
|
570
|
Cho RW, Clarke MF. Recent advances in cancer stem cells. Curr Opin Genet Dev 2008; 18:48-53. [PMID: 18356041 DOI: 10.1016/j.gde.2008.01.017] [Citation(s) in RCA: 179] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Revised: 01/10/2008] [Accepted: 01/23/2008] [Indexed: 01/08/2023]
Abstract
The theory of cancer stem cells states that a subset of cancer cells within a tumor has the ability to self-renew and differentiate. Only those cells within a tumor that have these two properties are called cancer stem cells. This concept was first demonstrated in the study of leukemia where only cells with specific surface antigen profiles were able to cause leukemia when engrafted into immunodeficient mice. In recent years solid tumors were studied utilizing similar techniques in mice. Human tumors where evidence of cancer stem cells has been published include tumors of the breast, brain, pancreas, head and neck, and colon. If this difference in tumorigenicity of cancer cells also occurs in patients, then the ability to enrich for cancer stem cells lays an important groundwork for future studies where mechanisms involved in cancer stem cells can now be investigated.
Collapse
Affiliation(s)
- Robert W Cho
- Department of Pediatrics Division of Stem Cell Transplantation, Stanford University, Palo Alto, CA 94304-1334, United States
| | | |
Collapse
|
571
|
Abstract
Proliferation in continuously renewing tissues, including the mammary gland, is hierarchically organized with a small number of slowly dividing stem cells and a greater number of more rapidly proliferating 'transit amplifying' cells. Mammary stem cells have been recently identified and purified based on their surface antigen expression. The recognition of mammary epithelial stem cells had led to the hypothesis that these may be at the root of breast cancer. In support of this, a highly tumorigenic subpopulation of cancer cells - cancer stem cells - has recently been identified in primary and metastatic breast cancer samples and in a number of established breast cancer cell lines. The existence of cancer stem cells would explain why only a small minority of cancer cells is capable of extensive proliferation and transferral of the tumour. In this article we aim to review the evidence in support of the existence of both normal mammary stem cells and breast cancer stem cells, and provide further insight into how taking this subpopulation of cells into account may affect the way we treat epithelial cancers in the future.
Collapse
Affiliation(s)
- M Cariati
- Department of Academic Oncology, King's College London, Guy's Hospital, London, UK
| | | |
Collapse
|
572
|
Wu PS, Egger B, Brand AH. Asymmetric stem cell division: lessons from Drosophila. Semin Cell Dev Biol 2008; 19:283-93. [PMID: 18328747 DOI: 10.1016/j.semcdb.2008.01.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Accepted: 01/23/2008] [Indexed: 12/18/2022]
Abstract
Asymmetric cell division is an important and conserved strategy in the generation of cellular diversity during animal development. Many of our insights into the underlying mechanisms of asymmetric cell division have been gained from Drosophila, including the establishment of polarity, orientation of mitotic spindles and segregation of cell fate determinants. Recent studies are also beginning to reveal the connection between the misregulation of asymmetric cell division and cancer. What we are learning from Drosophila as a model system has implication both for stem cell biology and also cancer research.
Collapse
|
573
|
Tam PKH. Towards predictive, preventive, and personalized paediatric surgery. J Pediatr Surg 2008; 43:267-73. [PMID: 18280272 DOI: 10.1016/j.jpedsurg.2007.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 10/09/2007] [Indexed: 10/22/2022]
Abstract
Future medicine will be revolutionized by genomic and stem cell research, becoming predictive, preventive, and personalized. Despite the smallness of the specialty, paediatric surgery is well placed to play a determining role in this exciting development. First, paediatric surgeons are innovators and leaders. Second, paediatric surgery thrives on the multidisciplinary approach. Third, congenital anomalies provide genetic models for studies of complex diseases. Fourth, morphogenesis underpins basic understanding in development, ageing, cancer, and immunology. The next generation paediatric surgeons must seize the opportunities in large-scale biology research to develop the best future treatment for their patients.
Collapse
Affiliation(s)
- Paul K H Tam
- Department of Surgery, University of Hong Kong Medical Centre, Queen Mary Hospital, Hong Kong.
| |
Collapse
|
574
|
Affiliation(s)
- Derrick J Rossi
- Immune Disease Institute, Harvard Stem Cell Institute, and the Department of Pathology, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
575
|
Roukos DH. Innovative genomic-based model for personalized treatment of gastric cancer: integrating current standards and new technologies. Expert Rev Mol Diagn 2008; 8:29-39. [PMID: 18088228 DOI: 10.1586/14737159.8.1.29] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the era of network biology, understanding the complexity of the signaling pathways network in cancer origin, progression and metastasis will dramatically alter and improve treatment strategies. Prognosis of gastric cancer remains poor. Clinical decisions on treatment are based on tumor-node-metastasis (TNM) staging, but are suboptimal. This perspective review, integrating several concepts, including cancer stem cells, provides a novel treatment model for tailoring the best treatment in individual patients with gastric cancer. Biologic metastatic steps (invasion, angiogenesis, intra/extravasation, colonization and microenvironment at distant organs) are orchestrated by mutated genes. Identifying and profiling these key genes and their interactions with environmental factors such as Helicobacter pylori, driver mutations and interacting signaling pathways using high-throughput technologies (including omics, resequencing, genome-wide associations studies and RNAi) in unbiased studies can lead to the development of both novel biomarkers and targeted agents. A comprehensive bench-to-bedside treatment-guided algorithm is provided for optimum preoperative or postoperative combination of cytotoxic and targeted agents. The protocol can be applied with adequate modification for most solid tumors.
Collapse
Affiliation(s)
- Dimitrios H Roukos
- Surgical Oncology Research Unit, Department of Surgery, Ioannina University School of Medicine, GR 451 10 Ioannina, Greece.
| |
Collapse
|
576
|
|
577
|
The 5th International Society for Stem Cell Research (ISSCR) Annual Meeting, June 2007. Stem Cells 2008; 26:292-8. [DOI: 10.1634/stemcells.2007-0647] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
578
|
Martin J, Helm K, Ruegg P, Varella-Garcia M, Burnham E, Majka S. Adult lung side population cells have mesenchymal stem cell potential. Cytotherapy 2008; 10:140-51. [DOI: 10.1080/14653240801895296] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
579
|
What can we learn about breast cancer from stem cells? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 617:17-22. [PMID: 18497027 DOI: 10.1007/978-0-387-69080-3_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
580
|
Abstract
PURPOSE OF REVIEW Cancer is thought to be an evolutionary process. Modern studies of evolution increasingly rely on genome comparisons, and similar molecular phylogeny approaches could be translated to somatic cell genomes to reconstruct colorectal cancer progression. The purpose of this review is to outline how human somatic cell ancestral trees can organize many old and new observations. RECENT FINDINGS A somatic cell tree starts from the zygote and ends with present day normal or neoplastic cells. In between are ancestors and dead ends, which functionally correspond to stem and nonstem cells. Cancer genome projects illustrate that mutations are relatively infrequent, and consistent with normal mutation rates, particularly if mutations begin to accumulate from birth. Therefore, some mutations eventually found in cancers may first occur in normal appearing crypts, which are maintained by niches that allow for stem cell clonal evolution and selection. Although mutations occur too infrequently to function as somatic cell molecular clocks, potentially more labile epigenetic changes in CpG methylation may also record somatic cell ancestry. SUMMARY Somatic cell evolution can occur throughout life, and potentially at least some of this unseen past may be reconstructed by 'reading' the lifetime changes that accumulate within our genomes.
Collapse
|
581
|
|
582
|
Abstract
Cancers originally develop from normal cells that gain the ability to proliferate aberrantly and eventually turn malignant. These cancerous cells then grow clonally into tumors and eventually have the potential to metastasize. A central question in cancer biology is, which cells can be transformed to form tumors? Recent studies elucidated the presence of cancer stem cells that have the exclusive ability to regenerate tumors. These cancer stem cells share many characteristics with normal stem cells, including self-renewal and differentiation. With the growing evidence that cancer stem cells exist in a wide array of tumors, it is becoming increasingly important to understand the molecular mechanisms that regulate self-renewal and differentiation because corruption of genes involved in these pathways likely participates in tumor growth. This new paradigm of oncogenesis has been validated in a growing list of tumors. Studies of normal and cancer stem cells from the same tissue have shed light on the ontogeny of tumors. That signaling pathways such as Bmi1 and Wnt have similar effects in normal and cancer stem cell self-renewal suggests that common molecular pathways regulate both populations. Understanding the biology of cancer stem cells will contribute to the identification of molecular targets important for future therapies.
Collapse
Affiliation(s)
- Neethan A Lobo
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | |
Collapse
|
583
|
Deugnier MA, Petit V, Taddéi-De La Hosseraye I, Faraldo M, Glukhova MA. Vers la caractérisation des cellules souches de la glande mammaire murine adulte. Med Sci (Paris) 2007; 23:1125-31. [DOI: 10.1051/medsci/200723121125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
584
|
Gillis AJM, Stoop HJ, Hersmus R, Oosterhuis JW, Sun Y, Chen C, Guenther S, Sherlock J, Veltman I, Baeten J, van der Spek PJ, de Alarcon P, Looijenga LHJ. High-throughput microRNAome analysis in human germ cell tumours. J Pathol 2007; 213:319-28. [PMID: 17893849 DOI: 10.1002/path.2230] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Testicular germ cell tumours (GCTs) of adolescents and adults can be subdivided into seminomas (referred to as dysgerminomas of the ovary) and non-seminomas, all referred to as type II GCTs. They originate from carcinoma in situ (CIS), being the malignant counterparts of primordial germ cells (PGCs)/gonocytes. The invasive components mimic embryogenesis, including the stem cell component embryonal carcinoma (EC), the somatic lineage teratoma (TE), and the extra-embryonic tissues yolk sac tumour (YST) and choriocarcinoma (CH). The other type is the so-called spermatocytic seminomas (SS, type III GCT), composed of neoplastic primary spermatocytes. We reported previously that the miRNAs hsa-miR 371-373 cluster is involved in overruling cellular senescence induced by oncogenic stress, allowing cells to become malignant. Here we report the first high-throughput screen of 156 microRNAs in a series of type II and III GCTs (n = 69, in duplicate) using a quantitative PCR-based approach. After normalization to allow inter-sample analysis, the technical replicates clustered together, and the previous hsa-miRNA 371-373 cluster finding was confirmed. Unsupervised cluster analysis demonstrated that the cell lines are different from the in vivo samples. The in vivo samples, both normal and malignant, clustered predominantly based on their maturation status. This parallels normal embryogenesis, rather than chromosomal anomalies in the tumours. miRNAs within a single cluster showed a similar expression pattern, implying common regulatory mechanisms. Normal testicular tissue expressed most discriminating miRNAs at a higher level than SE and SS. Moreover, differentiated non-seminomas showed overexpression of discriminating miRNAs. These results support the model that miRNAs are involved in regulating differentiation of stem cells, retained in GCTs.
Collapse
Affiliation(s)
- A J M Gillis
- Department of Pathology, Josephine Nefkens Institute, Erasmus MC-University Medical Center Rotterdam, Daniel den Hoed, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
585
|
Abstract
How a cancer is initiated and established remains elusive despite all the advances in decades of cancer research. Recently the cancer stem cell (CSC) hypothesis has been revived, challenging the long-standing model of "clonal evolution" for cancer development and implicating the dawning of a potential cure for cancer [1]. The recent identification of precancerous stem cells (pCSCs) in cancer, an early stage of CSC development, however, implicates that the "clonal evolution" is not contradictory to the CSC hypothesis, but is rather an aspect of the process of CSC development [2]. The discovery of pCSC has revealed and will continue to reveal the volatile properties of CSC with respects to their phenotype, differentiation and tumorigenic capacity during initiation and progression. Both pCSC and CSC might also serve as precursors of tumor stromal components such as tumor vasculogenic stem/progenitor cells (TVPCs). Thus, the CSC hypothesis covers the developing process of tumor-initiating cells (TIC) --> pCSC --> CSC --> cancer, a cellular process that should parallel the histological process of hyperplasia/metaplasia (TIC) --> precancerous lesions (pCSC) --> malignant lesions (CSC --> cancer). The embryonic stem (ES) cell and germline stem (GS) cell genes are subverted in pCSCs. Especially the GS cell protein piwil2 may play an important role during the development of TIC --> pCSC --> CSC, and this protein may be used as a common biomarker for early detection, prevention, and treatment of cancer. As cancer stem cell research is yet in its infancy, definitive conclusions regarding the role of pCSC can not be made at this time. However this review will discuss what we have learned from pCSC and how this has led to innovative ideas that may eventually have major impacts on the understanding and treatment of cancer.
Collapse
Affiliation(s)
- Jian-Xin Gao
- Department of Pathology and Comprehensive Cancer Center, Medical Center, Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
586
|
Grinstein E, Wernet P. Cellular signaling in normal and cancerous stem cells. Cell Signal 2007; 19:2428-33. [PMID: 17651940 DOI: 10.1016/j.cellsig.2007.06.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Accepted: 06/21/2007] [Indexed: 12/30/2022]
Abstract
Self-renewing divisions of normal and cancerous stem cells are responsible for the initiation and maintenance of normal and certain cancerous tissues, respectively. Recent findings suggest that tumor surveillance mechanisms can reduce regenerative capacity and frequency of normal stem cells, thereby contributing to tissue aging. Signaling pathways promoting self-renewal of stem cells can also drive proliferation in cancer. The BMI-1 proto-oncogene is required for the maintenance of tissue-specific stem cells and is involved in carcinogenesis within the same tissues. BMI-1 promotes self-renewal of stem cells largely by interfering with two central cellular tumor suppressor pathways, p16(Ink4a)/retinoblastoma protein (Rb) and ARF/p53, whose disruption is a hallmark of cancer. Nucleolin, an Rb-associated protein, is abundant in proliferating cancerous cells and likely contributes to the maintenance of human CD34-positive stem/progenitor cells of hematopoiesis. Elucidation of the involvement of proto-oncogenes and tumor suppressors in the maintenance of stem cells might have therapeutic implications.
Collapse
Affiliation(s)
- Edgar Grinstein
- Institute of Transplantation Diagnostics and Cellular Therapeutics, Heinrich Heine University Medical Center, 40225 Düsseldorf, Germany.
| | | |
Collapse
|
587
|
Celis JE, Gromova I, Cabezón T, Gromov P, Shen T, Timmermans-Wielenga V, Rank F, Moreira JMA. Identification of a subset of breast carcinomas characterized by expression of cytokeratin 15: relationship between CK15+ progenitor/amplified cells and pre-malignant lesions and invasive disease. Mol Oncol 2007; 1:321-49. [PMID: 19383306 PMCID: PMC5543867 DOI: 10.1016/j.molonc.2007.09.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 09/12/2007] [Accepted: 09/13/2007] [Indexed: 10/22/2022] Open
Abstract
Recently, we presented evidence--based on the analysis of benign hyperproliferative lesions of the breast--for the presence of cells that express the stem cell marker cytokeratin (CK) 15 in combination with CK19, a protein widely expressed by mammary epithelial cells. Here we report the finding of a subset of breast carcinomas characterized by expression of CK15. CK15 expressing tumors constituted 5% (6 out of 120; 4 of ductal type and 2 of lobular type) of the high-risk breast carcinomas examined by gel-based proteomics and immunohistochemistry. Five out of the six CK15+ carcinomas were CK15+/CK19-. The remaining tumor was mainly composed of cells expressing both CK15 and CK19 (CK15+/CK19+), but it also contained invasive areas with cells expressing only one of these makers (CK15+/CK19- and CK15-/CK19+ cells). To address the relationship between putative luminal progenitor/amplified CK15+ cells and malignant disease, and to determine whether cells/lesions lose expression of CK15 as a result of tumour initiation and/or progression, we searched among our sample set for carcinomas in which invasive tumor areas co-existed with non-malignant cells and hyperproliferative and known pre-malignant lesions. Only one such tumour was found (T71), a CK15-/CK19+/p53+ carcinoma that contained p53 negative non-malignant epithelial cells exhibiting a variety of, CK15/CK19 cellular phenotypes (CK15+/CK19+; CK15+/CK19-; CK15-/CK19+; CK15-/CK19-), often associated with simple columnar cells. Single layers of epithelial cells exhibiting all four CK15/CK19 phenotypes were observed contiguous to areas of atypical ductal hyperplasia that contained p53 positive cells that lost CK15 expression (CK15-/CK19+) and had a very similar phenotype to those of the neighboring ductal carcinoma in situ (DCIS) and invasive cells. The undifferentiated CK15+/CK19+ cells, which had the phenotype CK15+/CK19+/CK14+/CK8+ and -/ER-/PgR-/AR-/CD44+ (weak)/CK17-/p63-/vimentin+/Ki67-/Bcl-2+ (weak)/GATA-3-/p53-, most likely correspond to lineage-restricted luminal progenitor cells able to generate the other more differentiated CK15/CK19 cellular phenotypes, thus giving rise to the daunting intratumour heterogeneity displayed by carcinoma T71. Cells with a very similar phenotype to the CK15+/CK19+ progenitor cells were observed in a juvenile fibroadenoma as well as in the large collecting ducts of the breast. The latter, however, expressed in addition CK14 and had a phenotype (CK15+/CK19+/CK14+/CK8+ (weak)/ER-/PgR-/AR-/CD44+ (weak)/CK17-/p63-/vimentin-/Ki67-/Bcl-2+/GATA-3-/p53-) that resembled that of the putative normal adult breast stem cells as inferred from published data. Further molecular characterization of these progenitor cells as well as unraveling of the signaling pathways that regulate their growth and differentiation may prove invaluable for developing novel therapeutic strategies that target cancer at an early stage.
Collapse
Affiliation(s)
- Julio E Celis
- Danish Centre for Translational Breast Cancer Research, Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
588
|
Hsu HJ, LaFever L, Drummond-Barbosa D. Diet controls normal and tumorous germline stem cells via insulin-dependent and -independent mechanisms in Drosophila. Dev Biol 2007; 313:700-12. [PMID: 18068153 DOI: 10.1016/j.ydbio.2007.11.006] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2007] [Revised: 10/31/2007] [Accepted: 11/02/2007] [Indexed: 01/08/2023]
Abstract
The external environment influences stem cells, but this process is poorly understood. Our previous work showed that germline stem cells (GSCs) respond to diet via neural insulin-like peptides (DILPs) that act directly on the germ line to upregulate stem cell division and cyst growth under a protein-rich diet in Drosophila. Here, we report that DILPs specifically control the G2 phase of the GSC cell cycle via phosphoinositide-3 kinase (PI3K) and dFOXO, and that a separate diet mediator regulates the G1 phase. Furthermore, GSC tumors, which escape the normal stem cell regulatory microenvironment, or niche, still respond to diet via both mechanisms, indicating that niche signals are not required for GSCs to sense or respond to diet. Our results document the effects of diet and insulin-like signals on the cell cycle of stem cells within an intact organism and demonstrate that the response to diet requires multiple signals. Moreover, the retained ability of GSC tumors to respond to diet parallels the long known connections between diet, insulin signaling, and cancer risk in humans.
Collapse
Affiliation(s)
- Hwei-Jan Hsu
- Department of Cell and Developmental Biology, 4120B Medical Research Building III, Vanderbilt University Medical Center, 465 21st Avenue South, Nashville, TN 37232-8240, USA
| | | | | |
Collapse
|
589
|
Abstract
PURPOSE OF REVIEW Current research and clinical developments on hematogeneous micrometastasis in breast cancer patients are summarized. RECENT FINDINGS Distant metastasis is the leading cause of cancer-related death in breast cancer and bone marrow is a common homing organ for blood-borne disseminated tumor cells derived from primary breast carcinomas. Sensitive immunocytochemical or molecular assays now allow the detection of single disseminated tumor cells in bone marrow or the peripheral blood at a frequency of 10 and these cells are detected in 10-60% of breast cancer patients without clinical or even histopathologic signs of metastasis. Recently, evidence has emerged that the detection of disseminated tumor cells and circulating tumor cells may provide important prognostic information, and in particular might help to monitor efficacy of therapy. Moreover, the characterization of disseminated tumor cells/circulating tumor cells has shed new light on the complex process underlying early tumor cell dissemination and metastatic progression in cancer patients. SUMMARY Research on disseminated tumor cells/circulating tumor cells will help to identify novel targets for biological therapies aimed at preventing metastatic relapse and to monitor the efficacy of these therapies. In particular, understanding tumor dormancy and identifying metastatic stem cells might result in the development of new concepts for antimetastatic therapies.
Collapse
|
590
|
Rajasekhar VK, Begemann M. Concise Review: Roles of Polycomb Group Proteins in Development and Disease: A Stem Cell Perspective. Stem Cells 2007; 25:2498-510. [PMID: 17600113 DOI: 10.1634/stemcells.2006-0608] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The acquisition and maintenance of cell fate are essential for metazoan growth and development. A strict coordination between genetic and epigenetic programs regulates cell fate determination and maintenance. Polycomb group (PcG) genes are identified as essential in these epigenetic developmental processes. These genes encode components of multimeric transcriptional repressor complexes that are crucial in maintaining cell fate. PcG proteins have also been shown to play a central role in stem cell maintenance and lineage specification. PcG proteins, together with a battery of components including sequence-specific DNA binding/accessory factors, chromatin remodeling factors, signaling pathway intermediates, noncoding small RNAs, and RNA interference machinery, generally define a dynamic cellular identity through tight regulation of specific gene expression patterns. Epigenetic modification of chromatin structure that results in expression silencing of specific genes is now emerging as an important molecular mechanism in this process. In embryonic stem (ES) cells and adult stem cells, such specific genes represent those associated with differentiation and development, and silencing of these genes in a PcG protein-dependent manner confers stemness. ES cells also contain novel chromatin motifs enriched in epigenetic modifications associated with both activation and repression of genes, suggesting that certain genes are poised for activation or repression. Interestingly, these chromatin domains are highly coincident with the promoters of developmental regulators, which are also found to be occupied by PcG proteins. The epigenetic integrity is compromised, however, by mutations or other alterations that affect the function of PcG proteins in stem cells leading to aberrant cell proliferation and tissue transformation, a hallmark of cancer. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Vinagolu K Rajasekhar
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Rockefeller Research Laboratories, Room #945, New York, New York 10021, USA.
| | | |
Collapse
|
591
|
Abstract
Parallel to the role that normal stem cells play in organogenesis, cancer stem cells are thought to be crucial for tumorigenesis. Understanding normal development might therefore lead to better treatments of cancer. We review recent data that stem cells of glioblastoma, a highly malignant brain tumour, seem to be dependent on cues from aberrant vascular niches that mimic the normal neural stem cell niche. These data have direct implications for cancer, highlighting the similarity between normal and malignant stem cells and identifying the tumour microenvironment as a target for new therapies.
Collapse
Affiliation(s)
- Richard J Gilbertson
- Department of Developmental Neurobiology and Oncology, St. Jude Children's Research Hospital, 332 North Lauderdale Street, Memphis, Tennessee 38105, USA.
| | | |
Collapse
|
592
|
Wen YH, Giashuddin S, Shapiro RL, Velazquez E, Melamed J. Unusual occurrence of a melanoma with intermixed epithelial component: a true melanocarcinoma?: case report and review of epithelial differentiation in melanoma by light microscopy and immunohistochemistry. Am J Dermatopathol 2007; 29:395-9. [PMID: 17667176 DOI: 10.1097/dad.0b013e31812f5235] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We report a case of a 27-year-old woman with a nonpigmented lesion on the right scalp. Histological examination showed a malignant nodular neoplasm with 2 distinct but intimately admixed components: a malignant melanoma with a spindle component and an unusual glandular component. Immunohistochemical studies demonstrated epithelial differentiation on the basis of cytokeratin (CAM5.2 and AE1/AE3) expression in the glandular component and melanocytic differentiation (HMB-45, PNL2, MITF, and S-100) of the spindle cell component. A single melanocytic marker (MITF) was expressed in both components, raising the possibility of dual differentiation in a single tumor, rather than the alternative considerations of a collision tumor or a reactive pseudoepitheliomatous hyperplasia with eccrine duct lumen formation within a melanoma. This unusual tumor with both melanocytic and epithelial components may represent a true melanocarcinoma, which becomes a plausible consideration, in view of melanoma plasticity and recent experimental evidence and speculation about the role of stem cells in melanoma.
Collapse
Affiliation(s)
- Y Hannah Wen
- Department of Pathology, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | | | |
Collapse
|
593
|
Lonergan T, Bavister B, Brenner C. Mitochondria in stem cells. Mitochondrion 2007; 7:289-96. [PMID: 17588828 PMCID: PMC3089799 DOI: 10.1016/j.mito.2007.05.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Revised: 04/06/2007] [Accepted: 05/11/2007] [Indexed: 12/23/2022]
Abstract
The current status of knowledge about mitochondrial properties in mouse, monkey and human embryonic, adult and precursor stem cells is discussed. Topics include mitochondrial localization patterns, oxygen consumption and ATP content in cells as they relate to the maintenance of stem cell properties and subsequent differentiation of stem cells into specific cell types. The significance of the perinuclear arrangement of mitochondria, which may be a characteristic feature of stem cells, as well as the expression of mitochondrial DNA regulatory proteins and mutations in the mitochondrial stem cell genome is also discussed.
Collapse
Affiliation(s)
- Thomas Lonergan
- Department of Biological Sciences, University of New Orleans, 2000 Lakeshore Drive, New Orleans, LA 70148, USA.
| | | | | |
Collapse
|
594
|
Inagaki A, Soeda A, Oka N, Kitajima H, Nakagawa J, Motohashi T, Kunisada T, Iwama T. Long-term maintenance of brain tumor stem cell properties under at non-adherent and adherent culture conditions. Biochem Biophys Res Commun 2007; 361:586-92. [PMID: 17673180 DOI: 10.1016/j.bbrc.2007.07.037] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 07/09/2007] [Indexed: 12/22/2022]
Abstract
There is increasing evidence for the presence of cancer stem-like progenitors in malignant brain tumors. This subpopulation of progenitor cells, the so-called "cancer stem cells (CSCs)", may play a pivotal role in brain tumor initiation, growth, and recurrence. Here we describe the establishment of one permanent brain tumor stem cell line that able to form new spheres after culture under adherent monolayer conditions and to recapitulate the properties of the original tumor upon transplantation into immunodeficient mice. Re-formed spheres retained their stem cell properties and isolated single CSCs from these spheres formed spheres/tumors even after long-term cultures (over 2 years). These data suggested that a small population of CSCs preserved its stem cell properties even after serial passages under non-adherent/adherent culture conditions. Evaluation of underlying metabolic events and assessment of the biological features of these viable cell lines will yield useful knowledge on the in situ behavior of brain tumors.
Collapse
Affiliation(s)
- Akihito Inagaki
- Department of Tissue and Organ Development Regeneration and Advanced Medical Science, Gifu University School of Medicine, Gifu City, Gifu, Japan
| | | | | | | | | | | | | | | |
Collapse
|
595
|
Abstract
Metastasis is the most lethal attribute of cancer cells and clinical decisions regarding treatment are based largely upon the likelihood of developing metastases. However, improvements in detection as well as recent experimental data have raised questions about the most appropriate definition of a metastasis, especially whether the mere presence of cells at secondary sites constitute a metastatic lesion. After reviewing the experimental basis of metastasis, a definition of metastasis is proffered along with a proposal to consider regarding modification of staging parameters.
Collapse
Affiliation(s)
- Danny R Welch
- Department of Pathology, Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA.
| |
Collapse
|
596
|
Chen C, Ridzon D, Lee CT, Blake J, Sun Y, Strauss WM. Defining embryonic stem cell identity using differentiation-related microRNAs and their potential targets. Mamm Genome 2007; 18:316-27. [PMID: 17610011 DOI: 10.1007/s00335-007-9032-6] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Accepted: 04/10/2007] [Indexed: 01/07/2023]
Abstract
Defining the identity of embryonic stem (ES) cells in quantitative molecular terms is a prerequisite to understanding their functional characteristics. Little is known about the role of microRNAs (miRNAs) in the regulation of ES cell identity. Statistical analysis of miRNA expression revealed unique expression signatures that could definitively classify mouse ES (mES), embryoid bodies (mEB), and somatic tissues. Analysis of these data sets also provides further confirmation of the nonrestrictive expression of miRNAs during murine development. Using combined genome-wide expression analyses of both miRNAs and mRNAs, we observed both negative and positive correlations in gene expression between miRNAs and their predicted targets. ES-specific miRNAs were positively correlated with their predicted targets, suggesting that mES-specific miRNAs may have a different role or mechanism in regulating their targets in mES maintenance or differentiation. The concept of cellular identity has changed with technology; this study redefines cellular identity by a generic statistical method of known dimension.
Collapse
Affiliation(s)
- Caifu Chen
- R&D, Applied Biosystems, Foster City, California 94404, USA
| | | | | | | | | | | |
Collapse
|
597
|
Oka N, Soeda A, Inagaki A, Onodera M, Maruyama H, Hara A, Kunisada T, Mori H, Iwama T. VEGF promotes tumorigenesis and angiogenesis of human glioblastoma stem cells. Biochem Biophys Res Commun 2007; 360:553-9. [PMID: 17618600 DOI: 10.1016/j.bbrc.2007.06.094] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2007] [Accepted: 06/08/2007] [Indexed: 11/27/2022]
Abstract
There is increasing evidence for the presence of cancer stem cells (CSCs) in malignant brain tumors, and these CSCs may play a pivotal role in tumor initiation, growth, and recurrence. Vascular endothelial growth factor (VEGF) promotes the proliferation of vascular endothelial cells (VECs) and the neurogenesis of neural stem cells. Using CSCs derived from human glioblastomas and a retrovirus expressing VEGF, we examined the effects of VEGF on the properties of CSCs in vitro and in vivo. Although VEGF did not affect the property of CSCs in vitro, the injection of mouse brains with VEGF-expressing CSCs led to the massive expansion of vascular-rich GBM, tumor-associated hemorrhage, and high morbidity, suggesting that VEGF promoted tumorigenesis via angiogenesis. These results revealed that VEGF induced the proliferation of VEC in the vascular-rich tumor environment, the so-called stem cell niche.
Collapse
Affiliation(s)
- Naoki Oka
- Department of Neurosurgery, Gifu University School of Medicine, 1-1 Yanagido, Gifu City, Gifu 501-1194, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
598
|
Ren L, Gao G, Zhao D, Ding M, Luo J, Deng H. Developmental stage related patterns of codon usage and genomic GC content: searching for evolutionary fingerprints with models of stem cell differentiation. Genome Biol 2007; 8:R35. [PMID: 17349061 PMCID: PMC1868930 DOI: 10.1186/gb-2007-8-3-r35] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 01/08/2007] [Accepted: 03/12/2007] [Indexed: 11/26/2022] Open
Abstract
Developmental-stage-related patterns of gene expression correlate with codon usage and genomic GC content in stem cell hierarchies. Background The usage of synonymous codons shows considerable variation among mammalian genes. How and why this usage is non-random are fundamental biological questions and remain controversial. It is also important to explore whether mammalian genes that are selectively expressed at different developmental stages bear different molecular features. Results In two models of mouse stem cell differentiation, we established correlations between codon usage and the patterns of gene expression. We found that the optimal codons exhibited variation (AT- or GC-ending codons) in different cell types within the developmental hierarchy. We also found that genes that were enriched (developmental-pivotal genes) or specifically expressed (developmental-specific genes) at different developmental stages had different patterns of codon usage and local genomic GC (GCg) content. Moreover, at the same developmental stage, developmental-specific genes generally used more GC-ending codons and had higher GCg content compared with developmental-pivotal genes. Further analyses suggest that the model of translational selection might be consistent with the developmental stage-related patterns of codon usage, especially for the AT-ending optimal codons. In addition, our data show that after human-mouse divergence, the influence of selective constraints is still detectable. Conclusion Our findings suggest that developmental stage-related patterns of gene expression are correlated with codon usage (GC3) and GCg content in stem cell hierarchies. Moreover, this paper provides evidence for the influence of natural selection at synonymous sites in the mouse genome and novel clues for linking the molecular features of genes to their patterns of expression during mammalian ontogenesis.
Collapse
Affiliation(s)
- Lichen Ren
- College of Life Sciences, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Ge Gao
- Center for Bioinformatics, College of Life Sciences, National Laboratory of Protein Engineering and Plant Genetics Engineering, Peking University, Beijing, 100871, PR China
| | - Dongxin Zhao
- Department of Cell Biology and Genetics, College of Life Sciences, Peking University, Beijing, 100871, PR China
| | - Mingxiao Ding
- Department of Cell Biology and Genetics, College of Life Sciences, Peking University, Beijing, 100871, PR China
| | - Jingchu Luo
- Center for Bioinformatics, College of Life Sciences, National Laboratory of Protein Engineering and Plant Genetics Engineering, Peking University, Beijing, 100871, PR China
| | - Hongkui Deng
- Department of Cell Biology and Genetics, College of Life Sciences, Peking University, Beijing, 100871, PR China
| |
Collapse
|
599
|
Shiras A, Chettiar ST, Shepal V, Rajendran G, Prasad GR, Shastry P. Spontaneous Transformation of Human Adult Nontumorigenic Stem Cells to Cancer Stem Cells Is Driven by Genomic Instability in a Human Model of Glioblastoma. Stem Cells 2007; 25:1478-89. [PMID: 17332509 DOI: 10.1634/stemcells.2006-0585] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The presence of a CD133+/nestin+ population in brain tumors suggests that a normal neural stem cell may be the cell of origin for gliomas. We have identified human CD133-positive NSCs from adult glioma tissue and established them as long-term in vitro cultures human neuroglial culture (HNGC)-1. Replicative senescence in HNGC-1 led to a high level of genomic instability and emergence of a spontaneously immortalized clone that developed into cell line HNGC-2 with features of cancer stem cells (CSCs), which include the ability for self-renewal and the capacity to form CD133-positive neurospheres and develop intracranial tumors. The data from our study specify an important role of genomic instability in initiation of transformed state as well as its progression into highly tumorigenic CSCs. The activated forms of Notch and Hes isoforms were expressed in both non-neoplastic neural stem cells and brain tumor stem cells derived from it. Importantly, a significant overexpression of these molecules was found in the brain tumor stem cells. These findings suggest that this model comprised of HNGC-1 and HNGC-2 cells would be a useful system for studying pathways involved in self-renewal of stem cells and their transformation to cancer stem cells. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Anjali Shiras
- National Centre for Cell Science (NCCS), NCCS Complex, University of Pune Campus, Ganeshkhind, Pune 411007, Maharashtra, India.
| | | | | | | | | | | |
Collapse
|
600
|
|