1
|
Pan Y, Yang X, Chen M, Shi K, Lyu Y, Meeson AP, Lash GE. Role of Cancer Side Population Stem Cells in Ovarian Cancer Angiogenesis. Med Princ Pract 2024; 33:403-413. [PMID: 39068919 PMCID: PMC11460956 DOI: 10.1159/000539642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/03/2024] [Indexed: 07/30/2024] Open
Abstract
Ovarian cancer is one of the most common gynecologic malignancies. Recurrence and metastasis often occur after treatment, and it has the highest mortality rate of all gynecological tumors. Cancer stem cells (CSCs) are a small population of cells with the ability of self-renewal, multidirectional differentiation, and infinite proliferation. They have been shown to play an important role in tumor growth, metastasis, drug resistance, and angiogenesis. Ovarian cancer side population (SP) cells, a type of CSC, have been shown to play roles in tumor formation, colony formation, xenograft tumor formation, ascites formation, and tumor metastasis. The rapid progression of tumor angiogenesis is necessary for tumor growth; however, many of the mechanisms driving this process are unclear as is the contribution of CSCs. The aim of this review was to document the current state of knowledge of the molecular mechanism of ovarian cancer stem cells (OCSCs) in regulating tumor angiogenesis.
Collapse
Affiliation(s)
- Yue Pan
- Division of Uterine Vascular Biology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - XueFen Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Miaojuan Chen
- Division of Uterine Vascular Biology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Kun Shi
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yuan Lyu
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | | | - Gendie E. Lash
- Division of Uterine Vascular Biology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, Third Affiliate Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Doerfler R, Yerneni S, Newby A, Chaudhary N, Shu A, Fein K, Hofstatter Azambuja J, Whitehead KA. Characterization and comparison of human and mouse milk cells. PLoS One 2024; 19:e0297821. [PMID: 38295101 PMCID: PMC10830055 DOI: 10.1371/journal.pone.0297821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Recent data has characterized human milk cells with unprecedented detail and provided insight into cell populations. While such analysis of freshly expressed human milk has been possible, studies of cell functionality within the infant have been limited to animal models. One commonly used animal model for milk research is the mouse; however, limited data are available describing the composition of mouse milk. In particular, the maternal cells of mouse milk have not been previously characterized in detail, in part due to the difficulty in collecting sufficient volumes of mouse milk. In this study, we have established a method to collect high volumes of mouse milk, isolate cells, and compare the cell counts and types to human milk. Surprisingly, we found that mouse milk cell density is three orders of magnitude higher than human milk. The cell types present in the milk of mice and humans are similar, broadly consisting of mammary epithelial cells and immune cells. These results provide a basis of comparison for mouse and human milk cells and will inform the most appropriate uses of mouse models for the study of human phenomena.
Collapse
Affiliation(s)
- Rose Doerfler
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Saigopalakrishna Yerneni
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Alexandra Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Ashley Shu
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Katherine Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Juliana Hofstatter Azambuja
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| |
Collapse
|
3
|
Cortés-Llanos B, Wang Y, Sims CE, Allbritton NL. A technology of a different sort: microraft arrays. LAB ON A CHIP 2021; 21:3204-3218. [PMID: 34346456 PMCID: PMC8387436 DOI: 10.1039/d1lc00506e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A common procedure performed throughout biomedical research is the selection and isolation of biological entities such as organelles, cells and organoids from a mixed population. In this review, we describe the development and application of microraft arrays, an analysis and isolation platform which enables a vast range of criteria and strategies to be used when separating biological entities. The microraft arrays are comprised of elastomeric microwells with detachable polymer bases (microrafts) that act as capture and culture sites as well as supporting carriers during cell isolation. The technology is elegant in its simplicity and can be implemented for samples possessing tens to millions of objects yielding a flexible platform for applications such as single-cell RNA sequencing, subcellular organelle capture and assay, high-throughput screening and development of CRISPR gene-edited cell lines, and organoid manipulation and selection. The transparent arrays are compatible with a multitude of imaging modalities enabling selection based on 2D or 3D spatial phenotypes or temporal properties. Each microraft can be individually isolated on demand with retention of high viability due to the near zero hydrodynamic stress imposed upon the cells during microraft release, capture and deposition. The platform has been utilized as a simple manual add-on to a standard microscope or incorporated into fully automated instruments that implement state-of-the-art imaging algorithms and machine learning. The vast array of selection criteria enables separations not possible with conventional sorting methods, thus garnering widespread interest in the biological and pharmaceutical sciences.
Collapse
|
4
|
Abbas S, Rashid F, Ulker E, Zaib S, Ayub K, Ullah S, Nadeem MA, Yousuf S, Ludwig R, Ali S, Iqbal J. Anticancer evaluation of a manganese complex on HeLa and MCF-7 cancer cells: design, deterministic solvothermal synthesis approach, Hirshfeld analysis, DNA binding, intracellular reactive oxygen species production, electrochemical characterization and density functional theory. J Biomol Struct Dyn 2020; 39:1068-1081. [DOI: 10.1080/07391102.2020.1726818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Saghir Abbas
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
- Department of Chemistry, Quaid-I-Azam University Islamabad, Islamabad, Pakistan
| | - Faisal Rashid
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Emine Ulker
- Department of Chemistry, Faculty of Arts & Sciences, Recep Tayyip Erdogan University, Rize, Turkey
| | - Sumera Zaib
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Khurshid Ayub
- Department of Chemistry, COMSATS University, Abbottabad Campus, Abbottabad, Pakistan
| | - Sana Ullah
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Sammer Yousuf
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Ralf Ludwig
- Leibniz-Institut für Katalyse e. V. an der Universität Rostock, Rostock, Germany
| | - Saqib Ali
- Department of Chemistry, Quaid-I-Azam University Islamabad, Islamabad, Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad, Pakistan
| |
Collapse
|
5
|
Kwon S, Kim SS, Nebeck HE, Ahn EH. Immortalization of Different Breast Epithelial Cell Types Results in Distinct Mitochondrial Mutagenesis. Int J Mol Sci 2019; 20:E2813. [PMID: 31181796 PMCID: PMC6600575 DOI: 10.3390/ijms20112813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/27/2019] [Accepted: 06/02/2019] [Indexed: 11/16/2022] Open
Abstract
Different phenotypes of normal cells might influence genetic profiles, epigenetic profiles, and tumorigenicities of their transformed derivatives. In this study, we investigate whether the whole mitochondrial genome of immortalized cells can be attributed to the different phenotypes (stem vs. non-stem) of their normal epithelial cell originators. To accurately determine mutations, we employed Duplex Sequencing, which exhibits the lowest error rates among currently-available DNA sequencing methods. Our results indicate that the vast majority of the observed mutations of the whole mitochondrial DNA occur at low-frequency (rare mutations). The most prevalent rare mutation types are C→T/G→A and A→G/T→C transitions. Frequencies and spectra of homoplasmic point mutations are virtually identical between stem cell-derived immortalized (SV1) cells and non-stem cell-derived immortalized (SV22) cells, verifying that both cell types were derived from the same woman. However, frequencies of rare point mutations are significantly lower in SV1 cells (5.79 × 10-5) than in SV22 cells (1.16 × 10-4). The significantly lower frequencies of rare mutations are aligned with a finding of longer average distances to adjacent mutations in SV1 cells than in SV22 cells. Additionally, the predicted pathogenicity for rare mutations in the mitochondrial tRNA genes tends to be lower (by 2.5-fold) in SV1 cells than in SV22 cells. While four known/confirmed pathogenic mt-tRNA mutations (m.5650 G>A, m.5521 G>A, m.5690 A>G, m.1630 A>G) were identified in SV22 cells, no such mutations were observed in SV1 cells. Our findings suggest that the immortalization of normal cells with stem cell features leads to decreased mitochondrial mutagenesis, particularly in RNA gene regions. The mutation spectra and mutations specific to stem cell-derived immortalized cells (vs. non-stem cell derived) have implications in characterizing the heterogeneity of tumors and understanding the role of mitochondrial mutations in the immortalization and transformation of human cells.
Collapse
Affiliation(s)
- Sujin Kwon
- Department of Pathology, University of Washington, Seattle, WA 98195, USA.
| | - Susan S Kim
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| | - Howard E Nebeck
- Department of Pathology, University of Washington, Seattle, WA 98195, USA.
| | - Eun Hyun Ahn
- Department of Pathology, University of Washington, Seattle, WA 98195, USA.
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
6
|
Al-anbaky Q, Al-karakooly Z, Kilaparty SP, Agrawal M, Albkuri YM, RanguMagar AB, Ghosh A, Ali N. Cytotoxicity of Manganese (III) Complex in Human Breast Adenocarcinoma Cell Line Is Mediated by the Generation of Reactive Oxygen Species Followed by Mitochondrial Damage. Int J Toxicol 2016; 35:672-682. [DOI: 10.1177/1091581816659661] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Manganese (Mn) complexes are widely studied because of their important catalytic properties in synthetic and biochemical reactions. A Mn (III) complex of an amidoamine ligand was synthesized using a tetradentate amidoamine ligand. In this study, the Mn (III) complex was evaluated for its biological activity by measuring its cytotoxicity in human breast adenocarcinoma cell line (MCF-7). Cytotoxic effects of the Mn (III) complex were determined using established biomarkers in an attempt to delineate the mechanism of action and the utility of the complex as a potential anticancer drug. The Mn (III) complex induces cell death in a dose- and time-dependent manner as shown by microculture tetrazolium assay, a measure of cytotoxic cell death. Our results demonstrated that cytotoxic effects were significantly increased at higher concentrations of Mn (III) complex and with longer time of treatment. The IC50 (Inhibitor concentration that results in 50% cell death) value of Mn (III) complex in MCF-7 cells was determined to be 2.5 mmol/L for 24 hours of treatment. In additional experiments, we determined the Mn (III) complex–mediated cell death was due to both apoptotic and nonspecific necrotic cell death mechanisms. This was assessed by ethidium bromide/acridine orange staining and flow cytometry techniques. The Mn (III) complex produced reactive oxygen species (ROS) triggering the expression of manganese superoxide dismutase 1 and ultimately damaging the mitochondrial function as is evident by a decline in mitochondrial membrane potential. Treatment of the cells with free radical scavenger, N, N-dimethylthiourea decreased Mn (III) complex–mediated generation of ROS and attenuated apoptosis. Together, these results suggest that the Mn (III) complex–mediated MCF-7 cell death utilizes combined mechanism involving apoptosis and necrosis perhaps due to the generation of ROS.
Collapse
Affiliation(s)
- Qudes Al-anbaky
- Department of Biology, University of Arkansas at Little Rock, Little Rock, AR, USA
- Department of Biology, University of Diyala, Baqubah, Iraq
| | | | - Surya P. Kilaparty
- Department of Biology, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Megha Agrawal
- Department of Biology, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Yahya M. Albkuri
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Ambar B. RanguMagar
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Anindya Ghosh
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR, USA
| | - Nawab Ali
- Department of Biology, University of Arkansas at Little Rock, Little Rock, AR, USA
| |
Collapse
|
7
|
Madera-Sandoval RL, Reyes-Maldonado E, Dzul-Caamal R, Gallegos-Rangel E, Domínguez-López ML, García-Latorre E, Vega-López A. Fat-associated lymphoid cluster in Cyprinus carpio: Characterisation and its relation with peritoneal haemangiosarcoma. FISH & SHELLFISH IMMUNOLOGY 2015; 44:633-641. [PMID: 25804491 DOI: 10.1016/j.fsi.2015.03.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/21/2015] [Accepted: 03/14/2015] [Indexed: 06/04/2023]
Abstract
FALC cells are natural helper cells producing Th2-type cytokines, which express c-kit, Sca-1, IL7R and CD45 in mouse and human. These cells are involved in allergic responses and contribute to the inflammatory reactions of adipose tissue; however, a lack of information prevails about the presence of these cells in other species. The aim of the study was to identify and characterise FALC cells in the common carp (Cyprinus carpio) using immunohistochemistry and molecular biology techniques as well as to explore their relationships with their microenvironment. Histological description of the FALC was performed using H&E and polyclonal antibodies were used against cell-surface markers such as c-kit, Sca-1 and CD45. Furthermore, gene expression of c-kit, Sca-1 and IL7R was assessed. C. carpio FALC cells express the same surface markers reported in FALC of the mouse at both the pre- and post-transcriptional level. By exposure to the soluble fraction of helminths, FALC cells produce abundant Th2 cytokines (IL-5, IL-6 and IL-13) but do not synthesise IL-1α. Additionally, FALC cells probably participate in vascular remodelling of the intestine vessels, inducing tumours because a malignant haemangiosarcoma in the peritoneal cavity was found. In this tumour, abundant FALC with their characteristic cell-surface markers were detected. The findings of this study suggest the involvement of some proto-oncogenes such as c-kit and Sca-1, and the deregulation of Src kinases modulated by CD45 present in C. carpio FALC with the ontogeny of peritoneal haemangiosarcoma in this fish species.
Collapse
Affiliation(s)
- Ruth L Madera-Sandoval
- Laboratorio de Toxicología Ambiental, Departamento de Ingeniería en Sistemas Ambientales, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México, D.F. CP 07738, Mexico
| | - Elba Reyes-Maldonado
- Laboratorio de Citología, Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala s/n, Casco de Santo Tomás, México, D.F. CP 11340, Mexico
| | - Ricardo Dzul-Caamal
- Laboratorio de Toxicología Ambiental, Departamento de Ingeniería en Sistemas Ambientales, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México, D.F. CP 07738, Mexico
| | - Esperanza Gallegos-Rangel
- Laboratorio de Toxicología Ambiental, Departamento de Ingeniería en Sistemas Ambientales, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México, D.F. CP 07738, Mexico
| | - María Lilia Domínguez-López
- Laboratorio de Inmunoquímica I, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala s/n, Casco de Santo Tomás, México, D.F. CP 11340, Mexico
| | - Ethel García-Latorre
- Laboratorio de Inmunoquímica I, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala s/n, Casco de Santo Tomás, México, D.F. CP 11340, Mexico
| | - Armando Vega-López
- Laboratorio de Toxicología Ambiental, Departamento de Ingeniería en Sistemas Ambientales, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México, D.F. CP 07738, Mexico.
| |
Collapse
|
8
|
Marcelo KL, Sills TM, Coskun S, Vasavada H, Sanglikar S, Goldie LC, Hirschi KK. Hemogenic endothelial cell specification requires c-Kit, Notch signaling, and p27-mediated cell-cycle control. Dev Cell 2014; 27:504-15. [PMID: 24331925 DOI: 10.1016/j.devcel.2013.11.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 09/01/2013] [Accepted: 11/04/2013] [Indexed: 02/05/2023]
Abstract
Delineating the mechanism or mechanisms that regulate the specification of hemogenic endothelial cells from primordial endothelium is critical for optimizing their derivation from human stem cells for clinical therapies. We previously determined that retinoic acid (RA) is required for hemogenic specification, as well as cell-cycle control, of endothelium during embryogenesis. Herein, we define the molecular signals downstream of RA that regulate hemogenic endothelial cell development and demonstrate that cell-cycle control is required for this process. We found that re-expression of c-Kit in RA-deficient (Raldh2(-/-)) primordial endothelium induced Notch signaling and p27 expression, which restored cell-cycle control and rescued hemogenic endothelial cell specification and function. Re-expression of p27 in RA-deficient and Notch-inactivated primordial endothelial cells was sufficient to correct their defects in cell-cycle regulation and hemogenic endothelial cell development. Thus, RA regulation of hemogenic endothelial cell specification requires c-Kit, notch signaling, and p27-mediated cell-cycle control.
Collapse
Affiliation(s)
- Kathrina L Marcelo
- Interdepartmental Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Children's Nutrition Research Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Tiffany M Sills
- Interdisciplinary Program in Cell and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Children's Nutrition Research Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Suleyman Coskun
- Yale Cardiovascular Research Center and Yale Stem Cell Center, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA
| | - Hema Vasavada
- Yale Cardiovascular Research Center and Yale Stem Cell Center, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA
| | - Supriya Sanglikar
- Yale Cardiovascular Research Center and Yale Stem Cell Center, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA
| | - Lauren C Goldie
- Interdepartmental Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Children's Nutrition Research Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Karen K Hirschi
- Interdepartmental Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Interdisciplinary Program in Cell and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Children's Nutrition Research Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Yale Cardiovascular Research Center and Yale Stem Cell Center, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA.
| |
Collapse
|
9
|
Ratajczak MZ, Jadczyk T, Schneider G, Kakar SS, Kucia M. Induction of a tumor-metastasis-receptive microenvironment as an unwanted and underestimated side effect of treatment by chemotherapy or radiotherapy. J Ovarian Res 2013; 6:95. [PMID: 24373588 PMCID: PMC3880975 DOI: 10.1186/1757-2215-6-95] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 12/26/2013] [Indexed: 12/12/2022] Open
Abstract
There are well-known side effects of chemotherapy and radiotherapy that are mainly related to the toxicity and impaired function of vital organs; however, the induction by these therapies of expression of several pro-metastatic factors in various tissues and organs that in toto create a pro-metastatic microenvironment is still, surprisingly, not widely acknowledged. In this review, we support the novel concept that toxic damage in various organs leads to upregulation in "bystander" tissues of several factors such as chemokines, growth factors, alarmines, and bioactive phosphosphingolipids, which attract circulating normal stem cells for regeneration but unfortunately also provide chemotactic signals to cancer cells that survived the initial treatment. We propose that this mechanism plays an important role in the metastasis of cancer cells to organs such as bones, lungs, and liver, which are highly susceptible to chemotherapeutic agents as well as ionizing irradiation. This problem indicates the need to develop efficient anti-metastatic drugs that will work in combination with, or follow, standard therapies in order to prevent the possibility of therapy-induced spread of tumor cells.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA
| | - Tomasz Jadczyk
- Third Division of Cardiology, Silesian Medical University, Katowice, Poland
| | - Gabriela Schneider
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA
| | - Sham S Kakar
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA
| | - Magda Kucia
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA
| |
Collapse
|
10
|
Vu NB, Nguyen TT, Tran LCD, Do CD, Nguyen BH, Phan NK, Pham PV. Doxorubicin and 5-fluorouracil resistant hepatic cancer cells demonstrate stem-like properties. Cytotechnology 2013; 65:491-503. [PMID: 23104270 PMCID: PMC3720974 DOI: 10.1007/s10616-012-9511-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 10/16/2012] [Indexed: 12/11/2022] Open
Abstract
The efficacy of hepatocellular carcinoma (HCC) treatment is very low because of the high percentage of recurrence and resistance to anticancer agents. Hepatic cancer stem cells (HCSCs) are considered the origin of such recurrence and resistance. Our aim was to evaluate the stemness of doxorubicin and 5-fluorouracil resistant hepatic cancer cells and establish the new method to isolate the HCSCs from primary cultured HCC tumors. HCC biopsies were used to establish primary cultures. Then, primary cells were selected for HCSCs by culture in medium supplemented with doxorubicin (0, 0.1, 0.25, 0.5 or 1 μg/mL), 5-fluorouracil (0, 0.1, 0.25, 0.5 or 1 μg/mL) or their combination. Selection was confirmed by detection of HCSC markers such as CD133, CD13, CD90, and the side population was identified by rhodamine 123 efflux. The cell population with the strongest expression of these markers was used to evaluate the cell cycle, gene expression profile, tumor sphere formation, marker protein expression, and in vivo tumorigenesis. Selective culture of primary cells in medium supplemented with 0.5 μg/mL doxorubicin and 1 μg/mL 5-fluorouracil selected cancer cells with the highest stemness properties. Selected cells strongly expressed CD13, CD133, CD90, and CD326, efflux rhodamine 123 and formed tumor spheres in suspension. Moreover, selected cells were induced to differentiate into cells with high expression of CD19 and AFP (alpha-fetoprotein), and importantly, could form tumors in NOD/SCID mice upon injection of 1 × 10(5) cells/mouse. Selective culture with doxorubicin and 5-fluorouracil will enrich HCSCs, is an easy method to obtain HCSCs that can be used to develop better therapeutic strategies for patients with HCC, and particularly HCSC-targeting therapy.
Collapse
Affiliation(s)
- Ngoc Bich Vu
- />Laboratory of Stem Cell Research and Application, University of Science, VNU-HCM, HCM City, Vietnam
| | - Tam Thanh Nguyen
- />Laboratory of Stem Cell Research and Application, University of Science, VNU-HCM, HCM City, Vietnam
| | - Long Cong-Duy Tran
- />University of Medical Center, Ho Chi Minh University of Medicine and Pharmacy, HCM City, Vietnam
| | - Cong Dinh Do
- />University of Medical Center, Ho Chi Minh University of Medicine and Pharmacy, HCM City, Vietnam
| | - Bac Hoang Nguyen
- />University of Medical Center, Ho Chi Minh University of Medicine and Pharmacy, HCM City, Vietnam
| | - Ngoc Kim Phan
- />Laboratory of Stem Cell Research and Application, University of Science, VNU-HCM, HCM City, Vietnam
| | - Phuc Van Pham
- />Laboratory of Stem Cell Research and Application, University of Science, VNU-HCM, HCM City, Vietnam
| |
Collapse
|
11
|
Richter A, Nissen N, Mailänder P, Stang F, Siemers F, Kruse C, Danner S. Mammary gland-derived nestin-positive cell populations can be isolated from human male and female donors. Stem Cell Res Ther 2013; 4:78. [PMID: 23835213 PMCID: PMC3854770 DOI: 10.1186/scrt229] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 06/27/2013] [Indexed: 12/30/2022] Open
Abstract
Introduction Nestin-expressing cells isolated from different human tissues reveal self-renewal capacity and a multilineage differentiation potential. In particular, adult stem/progenitor cell populations from exocrine glands such as the pancreas, salivary gland and sweat gland are characterized by prominent nestin expression. Interestingly, human mammary gland histological examinations also demonstrated the existence of nestin-positive cells in the ductal compartments. Within the scope of our previous work we wonder whether an isolation of nestin-positive cell populations from human mammary gland biopsies is possible and what characteristics they have in vitro. Cell populations from both sexes were propagated and subjected to a comparison with other gland-derived cell populations. Methods Human mammary tissue biopsies were mechanically and enzymatically treated, and the isolated acini structures were observed with time-lapse microscopy to track adherently outgrowing cells. The proliferation potential of the cell population was assessed by performing growth curves. On the gene and protein levels we investigated the expression of stem cell markers as well as markers indicating multilineage differentiation. Results We succeeded in establishing proliferating cell populations from breast tissue biopsies of both sexes. Our results display several similarities to the glandular stem cell populations from other exocrine glands. Beside their proliferation capacity during in vitro culture, the obtained cell populations are characterized by their prominent nestin expression. The cells share surface proteins commonly expressed on adult stem cells. We demonstrated the expression of stem cell-related genes like Oct4, Sox2, KLF4 and Nanog, and confirmed multipotent differentiation capacity by detecting transcripts expressed in endodermal, mesodermal and ectodermal cell types. Conclusion With this study we present an efficient procedure for isolation and propagation of nestin-positive stem cells obtained from male and female breast tissue, which is frequently available. The established multipotent cell populations could be easily expanded in vitro and thus hold promise for cell-based therapies and personalized medicine.
Collapse
|
12
|
Wang Y, Phillips CN, Herrera GS, Sims CE, Yeh JJ, Allbritton NL. Array of Biodegradable Microraftsfor Isolation and Implantation of Living, Adherent Cells. RSC Adv 2013; 3:9264-9272. [PMID: 23930219 PMCID: PMC3733277 DOI: 10.1039/c3ra41764f] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A new strategy for efficient sorting and implantation of viable adherent cells into animals is described. An array of biodegradable micro-structures (microrafts) was fabricated using a polydimethylsiloxane substrate for micromolding poly(lactic-co-glycolic acid) (PLGA). Screening various forms of PLGA determined that the suitability of PLGA for microraft manufacture, biocompatibility and in vitro degradation was dependent on molecular weight and lactic/glycolic ratio. Cells plated on the array selectively attached to the microrafts and could be identified by their fluorescence, morphology or other criteria. The cells were efficiently dislodged and collected from the array using a microneedle device. The platform was used to isolate specific cells from a mixed population establishing the ability to sort target cells for direct implantation. As a proof of concept, fluorescently conjugated microrafts carrying tumor cells stably expressing luciferase were isolated from an array and implanted subcutaneously into mice. In vivo bio-luminescence imaging confirmed the growth of a tumor in the recipient animals. Imaging of tissue sections from the tumors demonstrated in vivo degradation of the implanted microrafts. The process is a new strategy for isolating and delivering a small number of adherent cells for animal implantation with potential applications in tissue repair, tumor induction, in vivo differentiation of stem cells and other biomedical research.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599
| | - Colleen N. Phillips
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599
| | - Gabriela S. Herrera
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599
| | - Jen Jen Yeh
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Departments of Surgery and Pharmacology, University of North Carolina, Chapel Hill, NC 27599
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599 and North Carolina State University, Raleigh, NC 27695
| |
Collapse
|
13
|
Weng D, Song B, Koido S, Calderwood SK, Gong J. Immunotherapy of radioresistant mammary tumors with early metastasis using molecular chaperone vaccines combined with ionizing radiation. THE JOURNAL OF IMMUNOLOGY 2013; 191:755-63. [PMID: 23772032 DOI: 10.4049/jimmunol.1203286] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In the current study, exposure of mammary tumor cells derived from mice transgenic for the polyomavirus middle T oncogene to ionizing radiation resulted in the generation of a tumor cell population that preferentially expressed cancer stem cell markers. In addition, these cells were more resistant to subsequent radiation treatments and appeared to acquire an enhanced capacity for dissemination to the lungs of mice. Therefore, we tested an immunotherapy approach to the treatment of local and disseminated mammary tumor cells in a murine model using a recently developed molecular chaperone-based vaccine that specifically targets the radioresistant subpopulation of tumor cells. Heat shock protein 70-peptide complexes (Hsp70.PC-F) were extracted from fusions of dendritic cells and radiation-enriched tumor cells, and the resulting chaperone vaccines were used to treat mice with pre-existing lung metastases. Immunization of mice with the Hsp70.PC-F vaccine resulted in a T cell-mediated immune response, including a significant increase in CD4 and CD8 T cell proliferation and the induction of effector T cells capable of targeting radioresistant tumor cells. Importantly, the growth of primary tumors was inhibited, and the number of tumor cells metastasizing to lung was reduced significantly by combining chaperone vaccine with radiotherapy. These results indicate that Hsp70.PC-F vaccine can induce specific immunity to radioresistant populations of mammary tumor cells and, thus, can complement radiotherapy, leading to synergistic killing.
Collapse
Affiliation(s)
- Desheng Weng
- Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
14
|
Du WW, Fang L, Yang X, Sheng W, Yang BL, Seth A, Zhang Y, Yang BB, Yee AJ. The Role of Versican in Modulating Breast Cancer Cell Self-renewal. Mol Cancer Res 2013; 11:443-55. [DOI: 10.1158/1541-7786.mcr-12-0461] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Yang J, Liao D, Chen C, Liu Y, Chuang TH, Xiang R, Markowitz D, Reisfeld RA, Luo Y. Tumor-Associated Macrophages Regulate Murine Breast Cancer Stem Cells Through a Novel Paracrine EGFR/Stat3/Sox-2 Signaling Pathway. Stem Cells 2013; 31:248-58. [DOI: 10.1002/stem.1281] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 10/28/2012] [Indexed: 01/09/2023]
|
16
|
Plaks V, Brenot A, Lawson DA, Linnemann JR, Van Kappel EC, Wong KC, de Sauvage F, Klein OD, Werb Z. Lgr5-expressing cells are sufficient and necessary for postnatal mammary gland organogenesis. Cell Rep 2013; 3:70-8. [PMID: 23352663 DOI: 10.1016/j.celrep.2012.12.017] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 09/14/2012] [Accepted: 12/26/2012] [Indexed: 01/10/2023] Open
Abstract
Mammary epithelial stem cells are vital to tissue expansion and remodeling during various phases of postnatal mammary development. Basal mammary epithelial cells are enriched in Wnt-responsive cells and can reconstitute cleared mammary fat pads upon transplantation into mice. Lgr5 is a Wnt-regulated target gene and was identified as a major stem cell marker in the small intestine, colon, stomach, and hair follicle, as well as in kidney nephrons. Here, we demonstrate the outstanding regenerative potential of a rare population of Lgr5-expressing (Lgr5(+)) mammary epithelial cells (MECs). We found that Lgr5(+) cells reside within the basal population, are superior to other basal cells in regenerating functional mammary glands (MGs), are exceptionally efficient in reconstituting MGs from single cells, and exhibit regenerative capacity in serial transplantations. Loss-of-function and depletion experiments of Lgr5(+) cells from transplanted MECs or from pubertal MGs revealed that these cells are not only sufficient but also necessary for postnatal mammary organogenesis.
Collapse
Affiliation(s)
- Vicki Plaks
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143-0452, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Banakh I, Gonez LJ, Sutherland RM, Naselli G, Harrison LC. Adult pancreas side population cells expand after β cell injury and are a source of insulin-secreting cells. PLoS One 2012; 7:e48977. [PMID: 23152835 PMCID: PMC3494669 DOI: 10.1371/journal.pone.0048977] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 10/02/2012] [Indexed: 12/28/2022] Open
Abstract
Pancreas stem cells are a potential source of insulin-producing β cells for the therapy of diabetes. In adult tissues the ‘side population’ (SP) of cells that effluxes the DNA binding dye Hoechst 33342 through ATP-binding cassette transporters has stem cell properties. We hypothesised therefore that the SP would expand in response to β cell injury and give rise to functional β cells. SP cells were flow sorted from dissociated pancreas cells of adult mice, analysed for phenotype and cultured with growth promoting and differentiation factors before analysis for hormone expression and glucose-stimulated insulin secretion. SP cell number and colony forming potential (CFP) increased significantly in models of type diabetes, and after partial pancreatectomy, in the absence of hyperglycaemia. SP cells, ∼1% of total pancreas cells at 1 week of age, were enriched >10-fold for CFP compared to non-SP cells. Freshly isolated SP cells contained no insulin protein or RNA but expressed the homeobox transcription factor Pdx1 required for pancreas development and β cell function. Pdx1, along with surface expression of CD326 (Ep-Cam), was a marker of the colony forming and proliferation potential of SP cells. In serum-free medium with defined factors, SP cells proliferated and differentiated into islet hormone-expressing cells that secreted insulin in response to glucose. Insulin expression was maintained when tissue was transplanted within vascularised chambers into diabetic mice. SP cells in the adult pancreas expand in response to β cell injury and are a source of β cell progenitors with potential for the treatment of diabetes.
Collapse
Affiliation(s)
- Ilia Banakh
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Leonel J. Gonez
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Robyn M. Sutherland
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Gaetano Naselli
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Leonard C. Harrison
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
18
|
Yang X, Sarvestani SK, Moeinzadeh S, He X, Jabbari E. Three-dimensional-engineered matrix to study cancer stem cells and tumorsphere formation: effect of matrix modulus. Tissue Eng Part A 2012; 19:669-84. [PMID: 23013450 DOI: 10.1089/ten.tea.2012.0333] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Maintenance of cancer stem cells (CSCs) is regulated by the tumor microenvironment. Synthetic hydrogels provide the flexibility to design three-dimensional (3D) matrices to isolate and study individual factors in the tumor microenvironment. The objective of this work was to investigate the effect of matrix modulus on tumorsphere formation by breast cancer cells and maintenance of CSCs in an inert microenvironment without the interference of other factors. In that regard, 4T1 mouse breast cancer cells were encapsulated in inert polyethylene glycol diacrylate hydrogels and the effect of matrix modulus on tumorsphere formation and expression of CSC markers was investigated. The gel modulus had a strong effect on tumorsphere formation and the effect was bimodal. Tumorsphere formation and expression of CSC markers peaked after 8 days of culture. At day 8, as the matrix modulus was increased from 2.5 kPa to 5.3, 26.1, and 47.1 kPa, the average tumorsphere size changed from 37±6 μm to 57±6, 20±4, and 12±2 μm, respectively; cell number density in the gel changed from 0.8±0.1×10⁵ cells/mL to 1.7±0.2×10⁵, 0.4±0.1×10⁵, and 0.2±0.1×10⁵ cells/mL after initial encapsulation of 0.14×10⁵ cells/mL; and the expression of CD44 breast CSC marker changed from 17±4-fold to 38±9-, 3±1-, and 2±1-fold increase compared with the initial level. Similar results were obtained with MCF7 human breast carcinoma cells. Mouse 4T1 and human MCF7 cells encapsulated in the gel with 5.3 kPa modulus formed the largest tumorspheres and highest density of tumorspheres, and had highest expression of breast CSC markers CD44 and ABCG2. The inert polyethylene glycol hydrogel can be used as a model-engineered 3D matrix to study the role of individual factors in the tumor microenvironment on tumorigenesis and maintenance of CSCs without the interference of other factors.
Collapse
Affiliation(s)
- Xiaoming Yang
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina, USA
| | | | | | | | | |
Collapse
|
19
|
Guo S, Wang Y, Allbritton N, Jiang X. Ultrasound-induced release of micropallets with cells. APPLIED PHYSICS LETTERS 2012; 101:163703. [PMID: 23152640 PMCID: PMC3487920 DOI: 10.1063/1.4757648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 09/21/2012] [Indexed: 05/22/2023]
Abstract
Separation of selected adherent live cells attached on an array of microelements, termed micropallets, from a mixed population is an important process in biomedical research. We demonstrated that adherent cells can be safely, selectively, and rapidly released from the glass substrate together with micropallets using an ultrasound wave. A 3.3-MHz ultrasound transducer was used to release micropallets (500 μm × 500 μm × 300 μm) with attached HeLa cells, and a cell viability of 92% was obtained after ultrasound release. The ultrasound-induced release process was recorded by a high-speed camera, revealing a proximate velocity of ∼0.5 m/s.
Collapse
Affiliation(s)
- Sijia Guo
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina 27695, USA
| | | | | | | |
Collapse
|
20
|
Akter R, Hossain MZ, Kleve MG, Gealt MA. Wortmannin induces MCF-7 breast cancer cell death via the apoptotic pathway, involving chromatin condensation, generation of reactive oxygen species, and membrane blebbing. BREAST CANCER-TARGETS AND THERAPY 2012; 4:103-13. [PMID: 24367198 DOI: 10.2147/bctt.s31712] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Apoptosis can be used as a reliable marker for evaluating potential chemotherapeutic agents. Because wortmannin is a microbial steroidal metabolite, it specifically inhibits the phosphatidyl inositol 3-kinase pathway, and could be used as a promising apoptosis-based therapeutic agent in the treatment of cancer. The objective of this study was to investigate the biomolecular mechanisms involved in wortmannin-induced cell death of breast cancer-derived MCF-7 cells. METHODS AND RESULTS Our experimental results demonstrate that wortmannin has strong apoptotic effects through a combination of different actions, including reduction of cell viability in a dose-dependent manner, inhibition of proliferation, and enhanced generation of intracellular reactive oxygen species. CONCLUSION Our findings suggest that wortmannin induces MCF-7 cell death via a programmed pathway showing chromatin condensation, nuclear fragmentation, reactive oxygen species, and membrane blebbing, which are characteristics typical of apoptosis.
Collapse
Affiliation(s)
- Rozina Akter
- Applied Biosciences Emphasis, Department of Applied Science, University Arkansas at Little Rock, Little Rock, AR, USA
| | - Md Zakir Hossain
- Graduate Institute of Technology, University Arkansas at Little Rock, Little Rock, AR, USA
| | - Maurice G Kleve
- Department of Biology, College of Science and of Mathematics, University Arkansas at Little Rock, Little Rock, AR, USA
| | - Michael A Gealt
- Department of Biology, College of Science and of Mathematics, University Arkansas at Little Rock, Little Rock, AR, USA
| |
Collapse
|
21
|
Abstract
Stem cells of normal mammalian tissues are defined as nonspecialized cells that have two critical properties: (a) the ability to renew themselves through cell division and (b) the potency to differentiate into other cell types. Therefore, they play a crucial role in development and in tissue homeostasis during adult life. Being long-lived, they can be the targets of environmental carcinogens leading to the accumulation of consecutive genetic changes. Hence, the genome of stem cells must be exceptionally well protected, and several protective mechanisms have evolved to ensure the genetic integrity of the stem cell compartment in any given tissue. Ionizing radiation exposure can disrupt tissue homeostasis both through the induction of cell killing/depletion of radiosensitive stem cells, leading to loss of tissue functionality and by genotoxic damage, increasing overall risk of cancer. We will review the current knowledge about radiation effects in adult stem cells of specific normal tissues, including skin, breast, and brain, examine parallels, as well as differences with cancer stem cells, and discuss the relevance of stem cell effects to radiation risk and radiotherapy.
Collapse
Affiliation(s)
- Kevin M Prise
- Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, United Kingdom.
| | | |
Collapse
|
22
|
Endometrial stem cells and reproduction. Obstet Gynecol Int 2012; 2012:851367. [PMID: 22287970 PMCID: PMC3263645 DOI: 10.1155/2012/851367] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 11/15/2011] [Accepted: 11/21/2011] [Indexed: 01/17/2023] Open
Abstract
Abnormal endometrial function remains a significant cause of implantation failure, recurrent pregnancy loss, and other pathologies responsible for female infertility. The development of novel therapies to treat infertility due to endometrial dysfunction requires an understanding of the latest advancements in endometrial cell biology, such as the role of endometrial stem cells. The remarkable regenerative capacity of the human endometrium is absolutely essential for successful reproduction and likely requires a population of stem cells in the endometrium. The purpose of this review is to provide an introduction to some of the newest concepts in endometrial stem cell biology.
Collapse
|
23
|
Yuan H, Upadhyay G, Yin Y, Kopelovich L, Glazer RI. Stem cell antigen-1 deficiency enhances the chemopreventive effect of peroxisome proliferator-activated receptorγ activation. Cancer Prev Res (Phila) 2012; 5:51-60. [PMID: 21955520 PMCID: PMC3252486 DOI: 10.1158/1940-6207.capr-11-0256] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Stem cell antigen-1 (Sca-1, Ly6A) is a glycerophosphatidylinositol (GPI)-anchored protein that was identified as a murine marker of bone marrow stem cells. Although Sca-1 is widely used to enrich for stem and progenitor cells in various tissues, little is known about its function and associated signaling pathways in normal and malignant cells. Here, we report that the absence of Sca-1 in the mammary gland resulted in higher levels of PPARγ and PTEN, and a reduction of pSer84PPARγ, pERK1/2, and PPARδ. This phenotype correlated with markedly increased sensitivity of Sca-1 null mice to PPARγ agonist GW7845 and insensitivity to PPARδ agonist GW501516. Reduction of Sca-1 expression in mammary tumor cells by RNA interference resulted in a phenotype similar to the Sca-1 deficient mammary gland, as evidenced by increased PPARγ expression and transcriptional activity, resulting in part from a lesser susceptibility to proteasomal degradation. These data implicate Sca-1 as a negative regulator of the tumor suppressor effects of PPARγ.
Collapse
Affiliation(s)
- Hongyan Yuan
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Geeta Upadhyay
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Yuzhi Yin
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Levy Kopelovich
- Chemoprevention Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Robert I. Glazer
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
24
|
Ma H, Mismar W, Wang Y, Small DW, Ras M, Allbritton NL, Sims CE, Venugopalan V. Impact of release dynamics of laser-irradiated polymer micropallets on the viability of selected adherent cells. J R Soc Interface 2011; 9:1156-67. [PMID: 22158840 DOI: 10.1098/rsif.2011.0691] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
We use time-resolved interferometry, fluorescence assays and computational fluid dynamics (CFD) simulations to examine the viability of confluent adherent cell monolayers to selection via laser microbeam release of photoresist polymer micropallets. We demonstrate the importance of laser microbeam pulse energy and focal volume position relative to the glass-pallet interface in governing the threshold energies for pallet release as well as the pallet release dynamics. Measurements using time-resolved interferometry show that increases in laser pulse energy result in increasing pallet release velocities that can approach 10 m s(-1) through aqueous media. CFD simulations reveal that the pallet motion results in cellular exposure to transient hydrodynamic shear stress amplitudes that can exceed 100 kPa on microsecond timescales, and which produces reduced cell viability. Moreover, CFD simulation results show that the maximum shear stress on the pallet surface varies spatially, with the largest shear stresses occurring on the pallet periphery. Cell viability of confluent cell monolayers on the pallet surface confirms that the use of larger pulse energies results in increased rates of necrosis for those cells situated away from the pallet centre, while cells situated at the pallet centre remain viable. Nevertheless, experiments that examine the viability of these cell monolayers following pallet release show that proper choices for laser microbeam pulse energy and focal volume position lead to the routine achievement of cell viability in excess of 90 per cent. These laser microbeam parameters result in maximum pallet release velocities below 6 m s(-1) and cellular exposure of transient hydrodynamic shear stresses below 20 kPa. Collectively, these results provide a mechanistic understanding that relates pallet release dynamics and associated transient shear stresses with subsequent cellular viability. This provides a quantitative, mechanistic basis for determining optimal operating conditions for laser microbeam-based pallet release systems for the isolation and selection of adherent cells.
Collapse
Affiliation(s)
- Huan Ma
- Department of Chemical Engineering and Materials Science, University of California, 916 Engineering Tower, Irvine, CA 92697-2575, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Lim JM, Lee M, Lee EJ, Gong SP, Lee ST. Stem cell engineering: limitation, alternatives, and insight. Ann N Y Acad Sci 2011; 1229:89-98. [PMID: 21793843 DOI: 10.1111/j.1749-6632.2011.06093.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The 21st century will see improvements in the quality of human life. The development of new therapeutic technologies will prevent prevalent diseases and enable recovery from currently incurable diseases. The development of cell and tissue replacement therapies using stem cells and their progenitors will accelerate the development of causative treatments. The effort expended thus far in developing cell therapies has revealed many technical limitations. Thus, we must explore conceptual changes in the feasibility of stem cell therapy. This paper introduces the current limitations to stem cell engineering and ways to overcome these limitations, which will provide new insight into their clinical application.
Collapse
Affiliation(s)
- Jeong Mook Lim
- WCU Biomodulation and Department of Agricultural Biotechnology, Seoul National University, Seoul, Korea.
| | | | | | | | | |
Collapse
|
26
|
CD49f and CD61 identify Her2/neu-induced mammary tumor-initiating cells that are potentially derived from luminal progenitors and maintained by the integrin-TGFβ signaling. Oncogene 2011; 31:2614-26. [PMID: 21996747 PMCID: PMC3260386 DOI: 10.1038/onc.2011.439] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
HER2/Neu is overexpressed in 20-30% of breast cancers and associated with aggressive phenotypes and poor prognosis. For deciphering the role of HER2/Neu in breast cancer, mouse mammary tumor virus (MMTV)-Her2/neu transgenic mice that develop mammary tumors resembling human HER2-subtype breast cancer have been established. Several recent studies have revealed that HER2/Neu is overexpressed in and regulates self renewal of breast tumor initiating cells (TICs). However, in the MMTV-Her2/neu transgenic mouse model, the identity of TICs remains elusive, despite previous studies showing supportive evidence for existence of TICs in Her2/neu-induced mammary tumors. Through systematic screening and characterization, we identified surface markers CD49f, CD61 and ESA were aberrantly overexpressed in Her2-overexpressing mammary tumor cells. Analysis of these markers as well as CD24 detected anomalous expansion of the luminal progenitor population in preneoplastic mammary glands of Her2/neu-transgenic mice, indicating that aberrant luminal progenitors originated Her2-induced mammary tumors. The combined markers, CD49f and CD61, further delineated the CD49fhighCD61high-sorted fraction as a TIC-enriched population, which displayed increased tumorsphere formation ability, enhanced tumorigenicity both in vitro and in vivo and drug resistance to pacitaxel and doxorubicin. Moreover, the TIC-enriched population manifested increased TGFβ signaling and exhibited gene expression signatures of stemness, TGFβ signaling and Epithelial-to-Mesenchymal Transition. Our findings that self-renewal and clonogenicity of TICs were suppressed by pharmacologically inhibiting the TGFβ signaling further indicate that the TGFβ pathway is vital for maintenance of the TIC population. Finally, we showed that the integrin β3 (CD61) signaling pathway was required for sustaining active TGFβ signaling and self-renewal of TICs. We for the first time developed a technique to highly enrich TICs from mammary tumors of Her2/neu-transgenic mice, unraveled their properties and identified the cooperative integrin β3-TGFβ signaling axis as a potential therapeutic target for HER2-induced TICs.
Collapse
|
27
|
Kent S, Hutchinson J, Balboni A, Decastro A, Cherukuri P, Direnzo J. ΔNp63α promotes cellular quiescence via induction and activation of Notch3. Cell Cycle 2011; 10:3111-8. [PMID: 21912215 DOI: 10.4161/cc.10.18.17300] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Genetic analysis of TP63 indicates that ΔNp63 isoforms are required for preservation of self-renewing capacity in the stem cell compartments of diverse epithelial structures; however, the underlying cellular and molecular mechanisms remain incompletely defined. Cellular quiescence is a common feature of adult stem cells that may account for their ability to retain long-term replicative capacity while simultaneously limiting cellular division. Similarly, quiescence within tumor stem cell populations may represent a mechanism by which these populations evade cytotoxic therapy and initiate tumor recurrence. Here, we present evidence that ΔNp63α, the predominant TP63 isoform in the regenerative compartment of diverse epithelial structuresm, promotes cellular quiescence via activation of Notch signaling. In HC11 cells, ectopic ΔNp63α mediates a proliferative arrest in the 2N state coincident with reduced RNA synthesis characteristic of cellular quiescence. Additionally, ΔNp63α and other quiescence-inducing stimuli enhanced expression of Notch3 in HC11s and breast cancer cell lines, and ectopic expression of the Notch3 intracellular domain (N3 (ICD) ) was sufficient to cause accumulation in G 0/G 1 and increased expression of two genes associated with quiescence, Hes1 and Mxi1. Pharmacologic inhibition of Notch signaling or shRNA-mediated suppression of Notch3 were sufficient to bypass quiescence induced by ΔNp63α and other quiescence-inducing stimuli. These studies identify a novel mechanism by which ΔNp63α preserves long-term replicative capacity by promoting cellular quiescence and identify the Notch signaling pathway as a mediator of multiple quiescence-inducing stimuli, including ΔNp63α expression.
Collapse
Affiliation(s)
- Sierra Kent
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH, USA
| | | | | | | | | | | |
Collapse
|
28
|
Stem cell antigen-1 enhances tumorigenicity by disruption of growth differentiation factor-10 (GDF10)-dependent TGF-beta signaling. Proc Natl Acad Sci U S A 2011; 108:7820-5. [PMID: 21518866 DOI: 10.1073/pnas.1103441108] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Stem cell antigen (Sca)-1/Ly6A, a glycerophosphatidylinositol-linked surface protein, was found to be associated with murine stem cell- and progenitor cell-enriched populations, and also has been linked to the capacity of tumor-initiating cells. Despite these interesting associations, this protein's functional role in these processes remains largely unknown. To identify the mechanism underlying the protein's possible role in mammary tumorigenesis, Sca-1 expression was examined in Sca-1(+/EGFP) mice during carcinogenesis. Mammary tumor cells derived from these mice readily engrafted in syngeneic mice, and tumor growth was markedly inhibited on down-regulation of Sca-1 expression. The latter effect was associated with significantly elevated expression of the TGF-β ligand growth differentiation factor-10 (GDF10), which was found to selectively activate TGF-β receptor (TβRI/II)-dependent Smad3 phosphorylation. Overexpression of GDF10 attenuated tumor formation; conversely, silencing of GDF10 expression reversed these effects. Sca-1 attenuated GDF10-dependent TGF-β signaling by disrupting the heterodimerization of TβRI and TβRII receptors. These findings suggest a new functional role for Sca-1 in maintaining tumorigenicity, in part by acting as a potent suppressor of TGF-β signaling.
Collapse
|
29
|
Pleniceanu O, Harari-Steinberg O, Dekel B. Concise review: Kidney stem/progenitor cells: differentiate, sort out, or reprogram? Stem Cells 2010; 28:1649-60. [PMID: 20652959 PMCID: PMC2996087 DOI: 10.1002/stem.486] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
End-stage renal disease (ESRD) is defined as the inability of the kidneys to remove waste products and excess fluid from the blood. ESRD progresses from earlier stages of chronic kidney disease (CKD) and occurs when the glomerular filtration rate (GFR) is below 15 ml/minute/1.73 m2. CKD and ESRD are dramatically rising due to increasing aging population, population demographics, and the growing rate of diabetes and hypertension. Identification of multipotential stem/progenitor populations in mammalian tissues is important for therapeutic applications and for understanding developmental processes and tissue homeostasis. Progenitor populations are ideal targets for gene therapy, cell transplantation, and tissue engineering. The demand for kidney progenitors is increasing due to severe shortage of donor organs. Because dialysis and transplantation are currently the only successful therapies for ESRD, cell therapy offers an alternative approach for kidney diseases. However, this approach may be relevant only in earlier stages of CKD, when kidney function and histology are still preserved, allowing for the integration of cells and/or for their paracrine effects, but not when small and fibrotic end-stage kidneys develop. Although blood- and bone marrow-derived stem cells hold a therapeutic promise, they are devoid of nephrogenic potential, emphasizing the need to seek kidney stem cells beyond known extrarenal sources. Moreover, controversies regarding the existence of a true adult kidney stem cell highlight the importance of studying cell-based therapies using pluripotent cells, progenitor cells from fetal kidney, or dedifferentiated/reprogrammed adult kidney cells. Stem Cells 2010; 28:1649–1660.
Collapse
Affiliation(s)
- Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer, Israel
| | | | | |
Collapse
|
30
|
Wang Y, Phillips C, Xu W, Pai JH, Dhopeshwarkar R, Sims CE, Allbritton N. Micromolded arrays for separation of adherent cells. LAB ON A CHIP 2010; 10:2917-24. [PMID: 20838672 PMCID: PMC2994190 DOI: 10.1039/c0lc00186d] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
We present an efficient, yet inexpensive, approach for isolating viable single cells or colonies from a mixed population. This cell microarray platform possesses innovations in both the array manufacture and the manner of target cell release. Arrays of microwells with bases composed of detachable concave elements, termed microrafts, were fabricated by a dip-coating process using a polydimethylsiloxane mold as the template and the array substrate. This manufacturing approach enabled the use of materials other than photoresists to create the array elements. Thus microrafts possessing low autofluorescence could be fabricated for fluorescence-based identification of cells. Cells plated on the microarray settled and attached at the center of the wells due to the microrafts' concavity. Individual microrafts were readily dislodged by the action of a needle inserted through the compliant polymer substrate. The hard polymer material (polystyrene or epoxy resin) of which the microrafts were composed protected the cells from damage by the needle. For cell analysis and isolation, cells of interest were identified using a standard inverted microscope and microrafts carrying target cells were dislodged with the needle. The released cells/microrafts could be efficiently collected, cultured and clonally expanded. During the separation and collection procedures, the cells remained adherent and provided a measure of protection during manipulation, thus providing an extremely high single-cell cloning rate (>95%). Generation of a transfected cell line based on expression of a fluorescent protein demonstrated an important application for performing on-chip cell separations.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Colitti M. Expression of putative stem cell markers related to developmental stage of sheep mammary glands. Anat Histol Embryol 2010; 39:555-62. [PMID: 20809917 DOI: 10.1111/j.1439-0264.2010.01028.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
It is thought that the regenerative capacity of the mammary gland following post-lactation involution resides in multipotent stem cells within the luminal tissue. Adult stem cells make up a small percentage of the cells found in mature organ systems, however to define useful markers has long been a challenge. c-Kit (KIT) and its ligand stem cell factor (KITLG), ATP-binding cassette sub-family G member 2 (ABCG2) and Musashi 1 (MSI1) are good candidate to identify progenitor cells in their niche. Using real-time PCR we showed that KIT, KITLG and MSI1 expressions were up regulated before lambing and at involution relatively to prepubertal stage. The in situ hybridization analysis for KIT gene confirmed and localized the expression in luminal epithelial cells. The changes in the expression profile of putative stem cell markers in mammary glands of sheep suggest that they modify with the progression of lactation cycle, being up regulated during differentiation and down regulated during lactation.
Collapse
Affiliation(s)
- M Colitti
- Department of Scienze Animali, Faculty of Veterinary Medicine, University of Udine, via delle Scienze, 208, 33100 Udine, Italy.
| |
Collapse
|
32
|
Pai JH, Xu W, Sims CE, Allbritton NL. Microtable arrays for culture and isolation of cell colonies. Anal Bioanal Chem 2010; 398:2595-604. [PMID: 20644916 DOI: 10.1007/s00216-010-3984-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 06/07/2010] [Accepted: 06/29/2010] [Indexed: 01/09/2023]
Abstract
Cell microarrays with culture sites composed of individually removable microstructures or micropallets have proven benefits for isolation of cells from a mixed population. The laser energy required to selectively remove these micropallets with attached cells from the array depends on the microstructure surface area in contact with the substrate. Laser energies sufficient to release micropallets greater than 100 μm resulted in loss of cell viability. A new three-dimensional culture site similar in appearance to a table was designed and fabricated using a simple process that relied on a differential sensitivity of two photoresists to UV-mediated photopolymerization. With this design, the larger culture area rests on four small supports to minimize the surface area in contact with the substrate. Microtables up to 250 × 250 μm were consistently released with single 10-μJ pulses to each of the four support structures. In contrast, microstructures with a 150 × 150-μm surface area in contact with the substrate could not be reliably released at pulse energies up to 212 μJ. Cassie-Baxter wetting is required to provide a barrier of air to localize and sequester cells to the culture sites. A second asset of the design was an increased retention of this air barrier under conditions of decreased surface tension and after prolonged culture of cells. The improved air retention was due to the hydrophobic cavity created beneath the table and above the substrate which entrapped air when an aqueous solution was added to the array. The microtables proved an efficient method for isolating colonies from the array with 100% of selected colonies competent to expand following release from the array.
Collapse
Affiliation(s)
- Jeng-Hao Pai
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
33
|
Hu FF, Jing Xu, Cui YG, Qian XQ, Mao YD, Liao LM, Liu JY. Isolation and Characterization of Side Population Cells in the Postpartum Murine Endometrium. Reprod Sci 2010; 17:629-42. [DOI: 10.1177/1933719110369180] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Fei-Fei Hu
- Jiangsu Province Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China, Centre of Clinical Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Centre of Clinical Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Gui Cui
- Jiangsu Province Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xiao-Qiao Qian
- Centre of Clinical Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yun-Dong Mao
- Centre of Clinical Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lian-Ming Liao
- Stem Cell Laboratory, Fujian Second People's Hospital, Fuzhou, China
| | - Jia-Yin Liu
- Jiangsu Province Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China, Centre of Clinical Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China,
| |
Collapse
|
34
|
Zeng YA, Nusse R. Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture. Cell Stem Cell 2010; 6:568-77. [PMID: 20569694 PMCID: PMC2917779 DOI: 10.1016/j.stem.2010.03.020] [Citation(s) in RCA: 317] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 02/19/2010] [Accepted: 03/05/2010] [Indexed: 02/06/2023]
Abstract
Adult stem cells have the ability to self-renew and to generate specialized cells. Self-renewal is dependent on extrinsic niche factors but few of those signals have been identified. In addition, stem cells tend to differentiate in the absence of the proper signals and are therefore difficult to maintain in cell culture. The mammary gland provides an excellent system to study self-renewal signals, because the organ develops postnatally, arises from stem cells, and is readily generated from transplanted cells. We show here that adult mammary glands contain a Wnt-responsive cell population that is enriched for stem cells. In addition, stem cells mutant for the negative-feedback regulator Axin2 and therefore sensitized to Wnt signals have a competitive advantage in mammary gland reconstitution assays. In cell culture experiments, exposure to purified Wnt protein clonally expands mammary stem cells for many generations and maintains their ability to generate functional glands in transplantation assays. We conclude that Wnt proteins serve as rate-limiting self-renewal signals acting directly on mammary stem cells.
Collapse
Affiliation(s)
- Yi Arial Zeng
- Howard Hughes Medical Institute, Department of Developmental Biology, Stanford University, School of Medicine, Stanford, CA 94305-5323, USA
| | | |
Collapse
|
35
|
Ratajczak MZ, Shin DM, Liu R, Marlicz W, Tarnowski M, Ratajczak J, Kucia M. Epiblast/germ line hypothesis of cancer development revisited: lesson from the presence of Oct-4+ cells in adult tissues. Stem Cell Rev Rep 2010; 6:307-16. [PMID: 20309650 PMCID: PMC2888917 DOI: 10.1007/s12015-010-9143-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The morphology of several tumors mimics developmentally early tissues; tumors often express early developmental markers characteristic for the germ line lineage. Recently, our group identified a population of very small stem cells (SCs) in murine bone marrow (BM) and other adult organs that express several markers characteristic for epiblast/germ line-derived SCs. We named these rare cells "Very Small Embryonic/Epiblast-like Stem Cells (VSELs)." We hypothesized that these cells that express both epiblast and germ line markers are deposited during early gastrulation in developing tissues and organs and play an important role in the turnover of tissue-committed (TC) SCs. To support this, we envision that the germ line is not only the origin of SCs, but also remains as a scaffold or back-up for the SC compartment in adult life. Furthermore, we noticed that VSELs are protected from uncontrolled proliferation and teratoma formation by a unique DNA methylation pattern in some developmentally crucial imprinted genes, which show hypomethylation or erasure of imprints in paternally methylated genes and hypermethylation of imprints in the maternally methylated. In pathological situations, however, we hypothesize that VSELs could be involved in the development of several malignancies. Therefore, potential involvement of VSELs in cancerogenesis could support century-old concepts of embryonic rest- or germ line-origin hypotheses of cancer development. However, we are aware that this working hypothesis requires further direct experimental confirmation.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Louisville, KY 40202, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The steroidal regulation of proliferation and differentiation in the rodent mammary gland is well described, but how ovarian hormones regulate these processes in the human remains poorly understood. To investigate this, we developed the athymic nude mouse model in which intact normal human breast tissue is grafted subcutaneously and treated with estrogen and/or progesterone at human physiological serum levels. We demonstrated, first, that estrogen and not progesterone is the major epithelial cell mitogen in the adult non-pregnant, non-lactating breast, second, that estrogen induces progesterone receptor (PR) expression and, third, that PR expression is maximally induced at low estrogen concentrations while a higher amount of estrogen was required to induce proliferation. These data raised the question of whether one cell type possessed differential responses to high and low estrogen concentrations or whether PR expression and proliferation occurred in two cell populations. Using double-label immunofluorescence, we demonstrated that steroid receptor expression and cell proliferation (Ki67 antigen) occurred in separate cell populations in normal human breast epithelium, and that cells expressing the estrogen receptor-alpha (ERalpha) invariably contained the PR. We also found that this dissociation between steroid receptor expression and cell proliferation in normal epithelium was disrupted at an early stage in breast tumor formation. Recent findings presented herein support the proposal that some ERalpha/PR-positive epithelial cells are quiescent breast stem cells that act as 'steroid hormone sensors'. Such hormone sensor cells are likely to secrete positive or negative paracrine/juxtacrine factors dependent on the prevailing estrogen or progesterone concentration to influence the proliferative activity of adjacent ERalpha/PR-negative epithelial cells.
Collapse
Affiliation(s)
- R B Clarke
- Breast Biology Group, Clinical Research Department, Christie Hospital, Manchester, UK
| |
Collapse
|
37
|
Oates JE, Grey BR, Addla SK, Samuel JD, Hart CA, Ramani VAC, Brown MD, Clarke NW. Hoechst 33342 side population identification is a conserved and unified mechanism in urological cancers. Stem Cells Dev 2009; 18:1515-22. [PMID: 19260804 DOI: 10.1089/scd.2008.0302] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mutation within the adult human stem cell (SC) compartment has been proposed as a factor in the initiation and promotion of carcinogenesis. Isolation of these cancer stem cells (CSCs) has proven difficult, limiting their subsequent phenotypic, functional, and genetic characterization. We have used the Hoechst 33342 dye efflux technique to isolate an epithelial side population (SP) from genitourinary (GU) cancers, which is enriched for cells with SC traits. With informed consent, samples were taken from patients with primary tumors and undergoing surgery for prostatic (CaP), invasive bladder transitional cell (TCC), and renal cell carcinomas (RCC). Single cell epithelial suspensions were extracted from these and incubated with Hoechst 33342. Hoechst SP/non-SP profiles were then generated by flow cytometry using standardized protocols. SP/non-SP cell cycle status was established by Hoechst 33342 and Pyronin Y staining. Immunocytochemistry staining was performed for markers suggested as stem markers as well as lineage-specific markers. Functionality was determined using colony-forming assays and long-term monolayer culture. A characteristic verapamil-sensitive SP was isolated from all 3 urological malignancies and represented 0.57% +/- 0.11% (CaP), 0.52% +/- 0.49% (TCC), and 5.9% +/- 0.9% (RCC) of the total epithelial population. Cell cycle analysis showed that the SP had enhanced numbers of cells in G(0) as compared to the total cell population (CaP 12.4% +/- 3.2 vs. 3.8% +/- 1.0, RCC 23.2% +/- 3.4 vs. 1.8% +/- 0.9, and TCC 28.5% +/- 4.9 vs. 4% +/- 1.3). Immunocytochemistry demonstrated an increased expression of proliferative and putative stem markers within the SP fraction. Cultures confirmed significant enhancement of colony-forming ability and proliferative capacity of the SP fraction. A characteristic SP enriched for stem-like cells has been isolated from the 3 most common urological malignancies. This provides strong evidence that Hoechst 33342 efflux is a conserved and unified mechanism in GU cancer.
Collapse
Affiliation(s)
- Jeremy E Oates
- Genito-Urinary Cancer Research Group, School of Cancer and Imaging Sciences, Paterson Institute for Cancer Research, Manchester M20 4BX, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Raiser DM, Kim CF. Commentary: Sca-1 and Cells of the Lung: A matter of Different Sorts. Stem Cells 2009; 27:606-11. [PMID: 19259938 DOI: 10.1002/stem.10] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In two separate articles published in this issue, Teisanu et al. and McQualter et al. report the use of flow cytometry and cell sorting to identify putative bronchiolar stem cells that are low in expression for the cell surface marker Sca-1 yet negative for CD34, and a mesenchymal, fibroblastic progenitor cell population from the lung that is positive for Sca-1, respectively. At first glance, these studies may seem to suggest that Sca-1 and CD34 are not markers of an epithelial stem cell population in the lung, as we previously determined in studies that identified bronchioalveolar stem cells (BASCs), and may also appear to contradict each other. However, here we point to evidence that the findings of these three studies are not mutually exclusive, and rather, that the different cell isolation and culturing protocols used in these studies have allowed for the identification of unique pulmonary cell populations. Rather than discounting previous work on BASCs, these studies reveal the existence of new methods and new cell types that will be interesting to use in future functional tests for their importance in lung biology and lung disease.
Collapse
|
39
|
Chen J, Gremeaux L, Fu Q, Liekens D, Van Laere S, Vankelecom H. Pituitary progenitor cells tracked down by side population dissection. Stem Cells 2009; 27:1182-95. [PMID: 19418455 DOI: 10.1002/stem.51] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The pituitary gland represents the endocrine core, governing the body's hormonal landscape by adapting its cellular composition to changing demands. It is assumed that stem/progenitor cells are involved in this remodeling. Recently, we uncovered a candidate stem/progenitor cell population in the anterior pituitary. Here, we scrutinized this "side population" (SP) and show that, unexpectedly, not the subset expressing high levels of "stem cell antigen-1" (Sca1(high)) but the remainder non-Sca1(high) fraction clusters the pituitary progenitor cells. Transcriptomal interrogation revealed in the non-Sca1(high) SP upregulated expression of the pituitary stem/progenitor cell markers Sox2 and Sox9, and of multiple factors critically involved in pituitary embryogenesis. The non-Sca1(high) SP encloses the cells that generate spheres and display multipotent hormone differentiation capacity. In culture conditions selecting for the non-Sca1(high) subset within the SP, stem cell growth factors that induce SP expansion, affect transcription of embryonic factors, suggesting impact on a developmental program that unfolds within this SP compartment. Non-Sca1(high) SP cells, revealed by Sox2 expression, are observed in the postulated periluminal stem/progenitor cell niche, but also in small groups scattered over the gland, thereby advocating the existence of multiple niches. In early postnatal mice undergoing a pituitary growth wave, Sox2(+) cells are more abundant than in adults, concordant with a larger SP and higher non-Sca1(high) proportion. Together, we tracked down pituitary progenitor cells by SP phenotype, and thus provide a straightforward method to isolate and scrutinize these cells from the plastic pituitary ex vivo, as well as a culture system for in-depth exploration of their regulatory network.
Collapse
Affiliation(s)
- Jianghai Chen
- Department of Molecular Cell Biology, Laboratory of Tissue Plasticity, University of Leuven (KU Leuven), Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Positive selection, sorting, and collection of single cells from within a heterogeneous population are required for many biological studies. We recently demonstrated a miniaturized cell array for this purpose; however, on-chip pre-enrichment and isolation of specific target cells would provide significant value for cell isolation. In the current work, mixed cell samples of fewer than 30,000 cells were used for panning by means of on-array antibody-capture to pre-enrich the target population. The cell surface receptors Fc(epsilon)R(1), c-Kit, and ErbB2 were used for positive selection of RBL, RBL, and SK-BR-3 cells, respectively, from the mixed population. The capture efficiency, selectivity, and enrichment for the target cells were calculated and compared with fibronectin-coated controls. As expected, the capture efficiency depended on the frequency of the target cell in the mixed population over the range of 0.3-33%. For a frequency of 5% target cells, the capture efficiency was 39%-53% for the three conditions, while the selectivity varied between 78% and 98% with 16-20-fold enrichment. Furthermore, single-cell cloning studies demonstrated a high cloning efficiency of target cells selectively isolated from the array. Antibody-based pre-enrichment in combination with micropallet-based cell selection will be a valuable tool for isolation and expansion of rare cells from small heterogeneous populations.
Collapse
Affiliation(s)
- Hamed Shadpour
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
41
|
Colitti M, Farinacci M. Expression of a putative stem cell marker, Musashi 1, in mammary glands of ewes. J Mol Histol 2009; 40:139-49. [DOI: 10.1007/s10735-009-9224-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 06/09/2009] [Indexed: 10/20/2022]
|
42
|
Ratajczak MZ, Shin DM, Kucia M. Very small embryonic/epiblast-like stem cells: a missing link to support the germ line hypothesis of cancer development? THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1985-1992. [PMID: 19406990 PMCID: PMC2684162 DOI: 10.2353/ajpath.2009.081143] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/13/2009] [Indexed: 11/20/2022]
Abstract
The morphology of several tumors mimics developmentally early tissues, and tumors often express early developmental markers characteristic of the germ line lineage. The presence of these markers in neoplastic cells could reflect the dedifferentiation of somatic cells in which cancer develops or cancer origination in primitive stem cells closely related to the epiblast/germ line. The identification of primitive germ line-derived very small embryonic/epiblast-like stem cells, which are deposited early in embryogenesis in developing organs and persist in several organs into adulthood, raised the possibility that cancer may originate in these cells. In this review, we hypothesize that very small embryonic/epiblast-like stem cells could be a missing link that support the more than 100-year-old concepts of the embryonic rest or germ line origin hypotheses of cancer development; however, further experimental evidence is needed to support this hypothesis.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- or Magda Kucia, Ph.D., Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202, USA.
| | | | | |
Collapse
|
43
|
Zhou J, Zhang H, Gu P, Margolick JB, Yin D, Zhang Y. Cancer stem/progenitor cell active compound 8-quinolinol in combination with paclitaxel achieves an improved cure of breast cancer in the mouse model. Breast Cancer Res Treat 2009; 115:269-77. [PMID: 18506619 PMCID: PMC3320107 DOI: 10.1007/s10549-008-0072-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 05/19/2008] [Indexed: 12/30/2022]
Abstract
Increasing evidence suggests that breast cancer is caused by cancer stem cells and the cure of breast cancer requires eradication of breast cancer stem cells. In this study, we established and characterized a sphere culture model derived from side population cells from the human breast cancer cell line MCF7. The sphere culture could be maintained long term and was enriched in cells expressing known breast cancer stem cell marker CD44+CD24-. These sphere cells showed higher colony formation ability in vitro and higher tumorigenicity in vivo than MCF7 cells, suggesting the enrichment of breast cancer stem/progenitor cells. To identify compounds that preferentially inhibit the sphere cells, we performed a compound library screening. Two lead compounds, NSC24076 and NSC125034 and an analog of NSC125034, 8-quinolinol (8Q), were identified as having preferential activity against the sphere cells. 8Q showed some antitumor activity alone but had much better therapeutic effect and relapse prevention when combined with paclitaxel than either 8Q or paclitaxel alone in both MCF7 and MDA-MB-435 xenograft models. We propose that compounds selectively targeting cancer stem/progenitor cells when combined with standard chemotherapy drugs may produce an improved treatment of cancer without significant relapse.
Collapse
Affiliation(s)
- Jiangbing Zhou
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, The Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Hao Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, The Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Peihua Gu
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, The Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Joseph B. Margolick
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, The Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Deling Yin
- Department of Internal Medicine, James Quillen College of Medicine, East Tennessee State University, P.O. Box 70622, Johnson City, TN 37614, USA
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, The Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
44
|
Woodward WA, Bristow RG. Radiosensitivity of cancer-initiating cells and normal stem cells (or what the Heisenberg uncertainly principle has to do with biology). Semin Radiat Oncol 2009; 19:87-95. [PMID: 19249646 PMCID: PMC2700289 DOI: 10.1016/j.semradonc.2008.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mounting evidence suggests that parallels between normal stem cell biology and cancer biology may provide new targets for cancer therapy. Prospective identification and isolation of cancer-initiating cells from solid tumors has promoted the descriptive and functional identification of these cells allowing for characterization of their response to contemporary cancer therapies, including chemotherapy and radiation. In clinical radiation therapy, the failure to clinically eradicate all tumor cells (eg, a lack of response, partial response, or nonpermanent complete response by imaging) is considered a treatment failure. As such, biologists have explored the characteristics of the small population of clonogenic cancer cells that can survive and are capable of repopulating the tumor after subcurative therapy. Herein, we discuss the convergence of these clonogenic studies with contemporary radiosensitivity studies that use cell surface markers to identify cancer-initiating cells. Implications for and uncertainties regarding incorporation of these concepts into the practice of modern radiation oncology are discussed.
Collapse
Affiliation(s)
- Wendy Ann Woodward
- The University of Texas M.D. Anderson Cancer Center, Department of Radiation Oncology, Houston, Texas
| | - Robert Glen Bristow
- Radiation Medicine Program, Princess Margaret Hospital (University Health Network) and Departments of Radiation Oncology and Medical Biophysics, University of Toronto, Toronto Canada
| |
Collapse
|
45
|
Tsuji S, Yoshimoto M, Takahashi K, Noda Y, Nakahata T, Heike T. Side population cells contribute to the genesis of human endometrium. Fertil Steril 2008; 90:1528-37. [DOI: 10.1016/j.fertnstert.2007.08.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2007] [Revised: 07/31/2007] [Accepted: 08/04/2007] [Indexed: 12/20/2022]
|
46
|
Abstract
INTRODUCTION The lineages assumed by stem cells during hematopoiesis can be identified by the pattern of protein markers present on the surface of cells at different stages of differentiation. Specific antibodies directed at these markers have facilitated the isolation of hematopoietic stem cells by flow cytometry. DISCUSSION Similarly, stem cells in solid organs also can be identified using cell surface markers. In addition, solid tumors have recently been found to contain small proportions of cells that are capable of proliferation, self-renewal, and differentiation into the various cell types seen in the bulk tumor. Of particular concern, these tumor-initiating cells (termed cancer stem cells when multipotency and self-renewal have been demonstrated) often display characteristics of treatment resistance, particularly to ionizing radiation. Thus, it is important to be able to identify these cells in order to better understand the mechanisms of resistance, and to be able to predict outcome and response to treatment. This depends, of course, on identifying markers that can be used to identify the cells, and for some solid tumors, a specific pattern of cell surface markers is emerging. In breast cancer, for example, the tumor-initiating cells have a characteristic Lin(-)CD44(+)CD24(-/lo) ESA(+) antigenic pattern. In cells derived from some high-grade gliomas, expression of CD133 on the cell surface appears to select for a population of tumor-initiating, treatment resistant cells. CONCLUSION Because multiple markers, typically examined on single cells using flow cytometry, are used routinely to identify the subpopulation of tumor-initiating cells, and because the number of these cells is small, the challenge remains to detect them in clinical samples and to determine their ability to predict outcome and/or response to treatment, the hallmarks of established biomarkers.
Collapse
|
47
|
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
|
48
|
Szotek PP, Chang HL, Brennand K, Fujino A, Pieretti-Vanmarcke R, Lo Celso C, Dombkowski D, Preffer F, Cohen KS, Teixeira J, Donahoe PK. Normal ovarian surface epithelial label-retaining cells exhibit stem/progenitor cell characteristics. Proc Natl Acad Sci U S A 2008; 105:12469-73. [PMID: 18711140 PMCID: PMC2527935 DOI: 10.1073/pnas.0805012105] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Indexed: 12/18/2022] Open
Abstract
Ovulation induces cyclic rupture and regenerative repair of the ovarian coelomic epithelium. This process of repeated disruption and repair accompanied by complex remodeling typifies a somatic stem/progenitor cell-mediated process. Using BrdU incorporation and doxycycline inducible histone2B-green fluorescent protein pulse-chase techniques, we identify a label-retaining cell population in the coelomic epithelium of the adult mouse ovary as candidate somatic stem/progenitor cells. The identified population exhibits quiescence with asymmetric label retention, functional response to estrous cycling in vivo by proliferation, enhanced growth characteristics by in vitro colony formation, and cytoprotective mechanisms by enrichment for the side population. Together, these characteristics identify the label-retaining cell population as a candidate for the putative somatic stem/progenitor cells of the coelomic epithelium of the mouse ovary.
Collapse
Affiliation(s)
- Paul P. Szotek
- *Pediatric Surgical Research Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Henry L. Chang
- *Pediatric Surgical Research Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Kristen Brennand
- Howard Hughes Medical Institute, Harvard Stem Cell Institute and the Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02139
| | - Akihiro Fujino
- *Pediatric Surgical Research Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Rafael Pieretti-Vanmarcke
- *Pediatric Surgical Research Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Cristina Lo Celso
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Massachusetts General Hospital, Boston, MA 02114
| | - David Dombkowski
- Flow Cytometry Laboratory, Department of Pathology and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
| | - Frederic Preffer
- Flow Cytometry Laboratory, Department of Pathology and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
| | - Kenneth S. Cohen
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Massachusetts General Hospital, Boston, MA 02114
| | - Jose Teixeira
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114; and
| | - Patricia K. Donahoe
- *Pediatric Surgical Research Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| |
Collapse
|
49
|
Wang XY, Yin Y, Yuan H, Sakamaki T, Okano H, Glazer RI. Musashi1 modulates mammary progenitor cell expansion through proliferin-mediated activation of the Wnt and Notch pathways. Mol Cell Biol 2008; 28:3589-99. [PMID: 18362162 PMCID: PMC2423292 DOI: 10.1128/mcb.00040-08] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 02/06/2008] [Accepted: 03/13/2008] [Indexed: 01/17/2023] Open
Abstract
The RNA-binding protein Musashi1 (Msi1) is a positive regulator of Notch-mediated transcription in Drosophila melanogaster and neural progenitor cells and has been identified as a putative human breast stem cell marker. Here we describe a novel functional role for Msi1: its ability to drive progenitor cell expansion along the luminal and myoepithelial lineages. Expression of Msi1 in mammary epithelial cells increases the abundance of CD24(hi) Sca-1(+), CD24(hi) CD29(+), CK19, CK6, and double-positive CK14/CK18 progenitor cells. Proliferation is associated with increased proliferin-1 (PLF1) and reduced Dickkopf-3 (DKK3) secretion into the conditioned medium from Msi-expressing cells, which is associated with increased colony formation and extracellular signal-regulated kinase (ERK) phosphorylation. Treatment with the MEK inhibitor U0126 inhibits ERK activation and decreases Notch and beta-catenin/T-cell factor (TCF) reporter activity resulting from Msi1 expression. Reduction of DKK3 in control cells with a short hairpin RNA (shRNA) increases Notch and beta-catenin/TCF activation, whereas reduction of PLF1 with a shRNA in Msi1-expressing cells inhibits these pathways. These results identify Msi1 as a key determinant of the mammary lineage through its ability to coordinate cell cycle entry and activate the Notch and Wnt pathways by a novel autocrine process involving PLF1 and DKK3.
Collapse
Affiliation(s)
- Xiao-Yang Wang
- Department of Oncology, Georgetown University, and Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| | | | | | | | | | | |
Collapse
|
50
|
Vassilopoulos A, Wang RH, Petrovas C, Ambrozak D, Koup R, Deng CX. Identification and characterization of cancer initiating cells from BRCA1 related mammary tumors using markers for normal mammary stem cells. Int J Biol Sci 2008; 4:133-42. [PMID: 18461147 PMCID: PMC2367429 DOI: 10.7150/ijbs.4.133] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Accepted: 05/04/2008] [Indexed: 12/14/2022] Open
Abstract
It is hypothesized that cancer stem cells arise either from normal stem cells or from progenitor cells that have gained the ability to self-renew. Here we determine whether mammary cancer stem cells can be isolated by using antibodies that have been used for the isolation of normal mammary stem cells. We show that BRCA1 mutant cancer cell lines contained a subpopulation of CD24+CD29+ or CD24+CD49f+ cells that exhibited increased proliferation and colony forming ability in vitro, and enhanced tumor-forming ability in vivo. The purified CD24+CD29+ cells could differentiate and reconstitute the heterogeneity found in parental cells when plated as a monolayer. Under low-attachment conditions, we detected "tumorspheres" only in the presence of double positive cells, which maintained their ability to self-renew. Furthermore, CD24+CD29+ cells could form tubular structures reminiscent of the mammary ductal tree when grown in three-dimensional cultures, implying that these cancer cells maintain some of the characteristics of the normal stem cells. Nevertheless, they could still drive tumor formation since as low as 500 double positive cells immediately after sorting from BRCA1 mutant primary tumors were able to form tumors with the same heterogeneity found in the original tumors. These data provide evidence that breast cancer stem cells originate from normal stem cells and advance our understanding of BRCA1-associated tumorigenesis with possible implications for future cancer treatment.
Collapse
Affiliation(s)
- Athanassios Vassilopoulos
- 1. Genetics of Development, Disease Branch, National Institute of Diabetes, Digestive, Kidney Diseases, National Institutes of Health, Bethesda, Maryland, MD 20892, USA
| | - Rui-Hong Wang
- 1. Genetics of Development, Disease Branch, National Institute of Diabetes, Digestive, Kidney Diseases, National Institutes of Health, Bethesda, Maryland, MD 20892, USA
| | - Constantinos Petrovas
- 2. Immunology Laboratory, Vaccine Research Center, National Institute of Allergy, Infectious Diseases (NIAID), National Institutes of Health, Bethesda, Maryland, MD 20892, USA
| | - David Ambrozak
- 2. Immunology Laboratory, Vaccine Research Center, National Institute of Allergy, Infectious Diseases (NIAID), National Institutes of Health, Bethesda, Maryland, MD 20892, USA
| | - Richard Koup
- 2. Immunology Laboratory, Vaccine Research Center, National Institute of Allergy, Infectious Diseases (NIAID), National Institutes of Health, Bethesda, Maryland, MD 20892, USA
| | - Chu-Xia Deng
- 1. Genetics of Development, Disease Branch, National Institute of Diabetes, Digestive, Kidney Diseases, National Institutes of Health, Bethesda, Maryland, MD 20892, USA
| |
Collapse
|