1001
|
Wilson LD, Ross SA, Lepore DA, Wada T, Penninger JM, Thomas PQ. Developmentally regulated expression of the regulator of G-protein signaling gene 2 (Rgs2) in the embryonic mouse pituitary. Gene Expr Patterns 2005; 5:305-11. [PMID: 15661635 DOI: 10.1016/j.modgep.2004.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2004] [Revised: 10/22/2004] [Accepted: 10/22/2004] [Indexed: 11/20/2022]
Abstract
During the development of the anterior pituitary gland, five distinct hormone-producing cell types emerge in a spatially and temporally regulated pattern from an invagination of oral ectoderm termed Rathke's Pouch. Evidence from mouse knockout and ectopic expression studies indicates that 12.5 days post coitum (dpc) to 14.5 dpc is a critical period for the expansion of the progenitor cell pool and the determination of most hormone-secreting cell types. While signaling proteins and transcription factors have been identified as having key roles in pituitary cell differentiation, little is known about the identity and function of proteins that mediate signal transduction in progenitor cells. To identify genes that are enriched in the embryonic pituitary gland, we compared gene expression in 14.5 dpc pituitary and 14.5 dpc embryo minus pituitary tissues using the NIA 15K microarray. Analysis of the data using the R program revealed that the Regulator of G Protein Signaling 2 (Rgs2) gene was 3.9-fold more abundant in the 14.5 dpc pituitary. In situ hybridisation confirmed this finding, and showed that Rgs2 expression in midline tissues was restricted to the pituitary and discrete regions of the nervous system. Within the pituitary, Rgs2 was expressed in undifferentiated cells, and was downregulated at the completion of the hormone cell differentiation. To investigate Rgs2 function in the pituitary, we examined hormone cell differentiation in Rgs2 null neonate mice. Pituitary cell differentiation and morphology appeared normal in the Rgs2 mutant animals, suggesting that other Rgs family members with similar activities may be present in the developing pituitary.
Collapse
Affiliation(s)
- L D Wilson
- Murdoch Childrens Research Institute, Royal Childrens Hospital, Melbourne, Vic. 3052, Australia
| | | | | | | | | | | |
Collapse
|
1002
|
Wilson HL, O'Neill HC. Preparation of label from small cell numbers for microarray screening. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2005; 8:190-6. [PMID: 15669712 DOI: 10.1089/omi.2004.8.190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Common methods for amplification of labelled cRNA for hybridisation to Affymetrix GeneChips (Affymetrix Inc., Santa Clara, CA) assume that starting material is not limiting and require 2-5 microg of total RNA. However, often the target population of cells under study is a rare subset like stem cells or dendritic cells. To bypass this difficulty in the past, either the whole tissue or a representative cell line was used to obtain enough cells for experimentation. There are obvious limitations with these approaches. In the case of whole tissue, there are contaminating cells types, and cell lines may not exactly reflect cells in vivo. It has been reported that two cycles of amplification can generate enough labelled cRNA for hybridisation from as little as 2 ng of total RNA. This allows Affymetrix technology to be used to screen the gene expression of cells in low number, rare cell subsets or small patient biopsies. Adoption of this approach can be used to give an accurate profile of genes expressed in the specific cell subset of interest. Published methods and successful variations applied to these are discussed here.
Collapse
Affiliation(s)
- Heather L Wilson
- School of Biochemistry and Molecular Biology, Faculty of Science, Australian National University, Canberra, Australia
| | | |
Collapse
|
1003
|
Flaim CJ, Chien S, Bhatia SN. An extracellular matrix microarray for probing cellular differentiation. Nat Methods 2005; 2:119-25. [PMID: 15782209 DOI: 10.1038/nmeth736] [Citation(s) in RCA: 574] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Accepted: 12/21/2004] [Indexed: 01/06/2023]
Abstract
We present an extracellular matrix (ECM) microarray platform for the culture of patterned cells atop combinatorial matrix mixtures. This platform enables the study of differentiation in response to a multitude of microenvironments in parallel. The fabrication process required only access to a standard robotic DNA spotter, off-the-shelf materials and 1,000 times less protein than conventional means of investigating cell-ECM interactions. To demonstrate its utility, we applied this platform to study the effects of 32 different combinations of five extracellular matrix molecules (collagen I, collagen III, collagen IV, laminin and fibronectin) on cellular differentiation in two contexts: maintenance of primary rat hepatocyte phenotype indicated by intracellular albumin staining and differentiation of mouse embryonic stem (ES) cells toward an early hepatic fate, indicated by expression of a beta-galactosidase reporter fused to the fetal liver-specific gene, Ankrd17 (also known as gtar). Using this technique, we identified combinations of ECM that synergistically impacted both hepatocyte function and ES cell differentiation. This versatile technique can be easily adapted to other applications, as it is amenable to studying almost any insoluble microenvironmental cue in a combinatorial fashion and is compatible with several cell types.
Collapse
Affiliation(s)
- Christopher J Flaim
- Departments of Bioengineering and Medicine, University of California San Diego, 9500 Gilman Drive- MC 0412, La Jolla, California 92093-0412, USA
| | | | | |
Collapse
|
1004
|
Schmittwolf C, Kirchhof N, Jauch A, Dürr M, Harder F, Zenke M, Müller AM. In vivo haematopoietic activity is induced in neurosphere cells by chromatin-modifying agents. EMBO J 2005; 24:554-66. [PMID: 15660132 PMCID: PMC548653 DOI: 10.1038/sj.emboj.7600546] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2004] [Accepted: 12/14/2004] [Indexed: 11/08/2022] Open
Abstract
Modifications of DNA and chromatin are fundamental for the establishment and maintenance of cell type-specific gene expression patterns that constitute cellular identities. To test whether the developmental potential of fetal brain-derived cells that form floating sphere colonies (neurospheres) can be modified by destabilizing their epigenotype, neurosphere cells were treated with chemical compounds that alter the acetylation and methylation patterns of chromatin and DNA. Intravenous infusion of bulk or clonally derived neurosphere cells treated with a combination of trichostatin A (TSA) plus 5-aza-2'-deoxycytidine (AzaC) (TSA/AzaC neurosphere cells) yielded long-term, multilineage and transplantable neurosphere-derived haematopoietic repopulation. Untreated neurosphere cells exhibited no haematopoietic repopulation activity. The neurosphere-derived haematopoietic cells showed a diploid karyotype, indicating that they are unlikely to be products of cell fusion events, a conclusion strengthened by multicolour fluorescence in situ hybridization. Our results indicate that altering the epigenotype of neurosphere cells followed by transplantation enables the generation of neurosphere-derived haematopoietic cells.
Collapse
Affiliation(s)
- Carolin Schmittwolf
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Würzburg, Würzburg, Germany
| | - Nicole Kirchhof
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Würzburg, Würzburg, Germany
| | - Anna Jauch
- Institut für Humanmedizin, Universität Heidelberg, Heidelberg, Germany
| | - Michael Dürr
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Würzburg, Würzburg, Germany
| | - Friedrich Harder
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Würzburg, Würzburg, Germany
| | - Martin Zenke
- Institute for Biomedical Engineering, Cell Biology, Aachen, Germany
| | - Albrecht M Müller
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Würzburg, Würzburg, Germany
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Würzburg, Versbacherstr. 5, 97078 Würzburg, Germany. Tel.: +49 931 201 45848; Fax: +49 931 201 45147; E-mail:
| |
Collapse
|
1005
|
Yao S, Sukonnik T, Kean T, Bharadwaj RR, Pasceri P, Ellis J. Retrovirus silencing, variegation, extinction, and memory are controlled by a dynamic interplay of multiple epigenetic modifications. Mol Ther 2005; 10:27-36. [PMID: 15233939 DOI: 10.1016/j.ymthe.2004.04.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2004] [Accepted: 04/08/2004] [Indexed: 12/22/2022] Open
Abstract
Retrovirus silencing in stem cells produces silent or variegated provirus. Additional memory and extinction mechanisms act during differentiation. Here we show that retrovirus is silent or variegated in mouse embryonic stem (ES) cells that are de novo methyltransferase (dnmt3a and dnmt3b) null. Memory is maintained during differentiation, and extinction occurs on variegated retrovirus, indicating that DNA methylation is dispensable for all forms of retrovirus silencing. Silent and variegated provirus are marked by hypoacetylated histone H3 and bound H1. In wild-type ES cells, silent and variegated proviruses are methylated and bound by hypoacetylated H3, MeCP2, and less H1. Silencing, variegation, and extinction are partially reactivated by 5-AzaC in this context. Lentivirus vectors are also silent or variegated, marked by silent chromatin, and exhibit memory and extinction. We conclude that the universal epigenetic mark of retrovirus silencing is silent chromatin established via the dynamic interplay of multiple epigenetic modifications that include but do not require DNA methylation. A molecular mechanism of competitive H1 and MeCP2 binding may account for this epigenetic interplay, and a model for variegation is discussed.
Collapse
Affiliation(s)
- Shuyuan Yao
- Developmental Biology Program, Hospital for Sick Children, 555 University Avenue, Toronto, ON, Canada M5G 1X8
| | | | | | | | | | | |
Collapse
|
1006
|
Maraldi NM, Lattanzi G, Squarzoni S, Capanni C, Cenni V, Manzoli FA. Implications for nuclear organization and gene transcription of lamin A/C specific mutations. ACTA ACUST UNITED AC 2005; 45:1-16. [PMID: 16185751 DOI: 10.1016/j.advenzreg.2005.02.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
1007
|
Capp JP. Stochastic gene expression, disruption of tissue averaging effects and cancer as a disease of development. Bioessays 2005; 27:1277-85. [PMID: 16299757 DOI: 10.1002/bies.20326] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite the extensive literature describing the somatic genetic alterations in cancer cells, the precise origins of cancer cells remain controversial. In this article, I suggest that the etiology of cancer and the generation of genetic instability in cancer cells should be considered in the light of recent findings on both the stochastic nature of gene expression and its regulation at tissue level. By postulating that gene expression is intrinsically probabilistic and that stabilization of gene expression arises by cellular interactions in "morphogenetic fields", development and cellular differentiation can be rethought in an evolutionary perspective. In particular, this article proposes that disruptions of cellular interactions are the initial source of abnormal gene expression in cancer cells. Consequently, cancer phenotypes such as genetic and epigenetic instabilities, and also the presence of cells with stem cell-like properties, may result from inaccurate and aberrant patterns of gene expression generated by microenvironmental alterations. Finally, the therapeutic implications of this view are discussed.
Collapse
Affiliation(s)
- Jean-Pascal Capp
- Genetic Instability and Cancer Group, Institute of Pharmacology and Structural Biology, CNRS UMR 5089, 205 route de Narbonne, 31077 Toulouse cedex 4, France.
| |
Collapse
|
1008
|
Abstract
Cord blood (CB) has served as a clinically beneficial source of hematopoietic stem (HSC) and progenitor (HPC) cells for transplantation and correction of a large number of malignant and non-malignant disorders. The capacity of CB to perform these functions is intimately related to the quality and quantity of HSC and HPC present in CB. This review covers the biology of HSC and HPC, efforts to expand these cells ex vivo for enhanced clinical utility that has thus far not been very successful, and recent studies on attempts to enhance the homing and engrafting capability of HSC as an alternative means for more effective use of the limited numbers of CB cells collected. This review also highlights the presence in CB of mesenchymal stem cells, unrestricted somatic stem cells, endothelial progenitor cells and immune cells. The presence and biology of these non-HSC/HPC may open up future possibilities for additional clinical benefit of CB, a product considered mainly for discard before its clinical transplantation potential was realized in the late 1980s.
Collapse
Affiliation(s)
- H E Broxmeyer
- Department of Microbiology and Immunology, and the Walther Oncology Center, Indiana univrsity School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
1009
|
Dontu G, Wicha MS. Survival of mammary stem cells in suspension culture: implications for stem cell biology and neoplasia. J Mammary Gland Biol Neoplasia 2005; 10:75-86. [PMID: 15886888 DOI: 10.1007/s10911-005-2542-5] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There is increasing evidence that a variety of neoplasms including breast cancer may result from transformation of normal stem and progenitor cells. In the past, isolation and characterization of mammary stem cells has been limited by the lack of suitable culture systems able to maintain these cells in an undifferentiated state in vitro. We have recently described a culture system in which human mammary stem and progenitor cells are able to survive in suspension and produce spherical colonies composed of both stem and progenitor cells. Recent observation that adult stem cells from other tissues may also retain the capacity for growth under anchorage independent conditions suggests a common underlying mechanism. We propose that this mechanism involves the interaction between the canonical Wnt signal pathway and E-cadherin. The Wnt pathway has been implicated in normal stem cell self-renewal in vivo. Furthermore, there is evidence that deregulation of this pathway in the mammary gland and other organs may play a key role in carcinogenesis. Thus, the development of in vitro suspension culture systems not only provides an important new tool for the study of mammary cell biology, but also may have important implications for understanding key molecular pathways in both normal and neoplastic stem cells.
Collapse
Affiliation(s)
- Gabriela Dontu
- Department of Internal Medicine, Hematology-Oncology, Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA.
| | | |
Collapse
|
1010
|
Hoelters J, Ciccarella M, Drechsel M, Geissler C, Gülkan H, Böcker W, Schieker M, Jochum M, Neth P. Nonviral genetic modification mediates effective transgene expression and functional RNA interference in human mesenchymal stem cells. J Gene Med 2005; 7:718-28. [PMID: 15712343 DOI: 10.1002/jgm.731] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Human mesenchymal stem cells (hMSC) are increasingly the focus of both basic and clinical research due to their ability to strike a balance between self-renewal and commitment to mesodermal differentiation. However, the promising therapeutic utility of hMSC in regenerative medical approaches requires detailed knowledge about their molecular characteristics. Therefore, genetic modification of hMSC provides a powerful tool to understand their complex molecular regulation mechanisms. METHODS Here we describe a proof of concept approach of separate and combined gene transfer and gene silencing by nonviral DNA transfection of enhanced green fluorescent protein (EGFP) and EGFP-targeted small interfering RNAs (siRNAs) in hMSC. For optimization of nonviral DNA and siRNA transfer different liposomal-based transfection strategies were validated. RESULTS The highest fraction of EGFP-expressing hMSC was obtained using Lipofectamine 2000 (50%) which also mediated the highest transfection rates of siRNAs into hMSC (>or=92%). Stably EGFP-expressing hMSC maintained their proliferation capacity paired with the ability to differentiate into different mesodermal lineages (bone, cartilage, and fat) without loss of transgene expression. Based on our nonviral nucleic acid delivery technique we showed efficient, functional, and long-term RNA interference (RNAi) in hMSC by gene specific knock-down of transiently and stably expressed EGFP (88-98%). CONCLUSIONS This is the first demonstration of efficient nonviral transfer of both nucleic acids (DNA and siRNA) into hMSC, exhibiting the potential of targeted modification of hMSC. In particular, the combination of these techniques represents a powerful gene transfer/silencing strategy, thus facilitating detailed genetic approaches to study regulatory networks in stem cell differentiation processes.
Collapse
Affiliation(s)
- Juergen Hoelters
- Chirurgisches Forschungslabor, Chirurgische Klinik und Poliklinik-Innenstadt, Ludwig-Maximilians-Universität München, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
1011
|
Abstract
Stem cells are characterized by their potential immortality and are capable of self-renewal and differentiation. Stem cells are proposed to provide the potential to cure degenerative diseases and to give important clues regarding human development and aging. However, stem cell research has evoked enthusiasm and passionate debate regarding the ethics of their use in medicine and reproduction. In this article, the current understanding of the biology of stem cells, their application in urology, and some of the controversies regarding their use are discussed. Although the clinical application of stem cell technologies to urologic practice is likely to be well in the future, advances in this field hold great promise for the correction of a number of illnesses. Nevertheless, scientists and ethicists will continue to struggle with their ethical responsibilities to the patient and society.
Collapse
Affiliation(s)
- Kirk C Lo
- Department of Urology, Baylor College of Medicine, One Baylor Plaza, Alkek N 730, Houston, TX 77030, USA
| | | | | |
Collapse
|
1012
|
Israeli D, Ziaei S, Gonin P, Garcia L. A proposal for the physiological significance of mdr1 and Bcrp1/Abcg2 gene expression in normal tissue regeneration and after cancer therapy. J Theor Biol 2005; 232:41-5. [PMID: 15498591 DOI: 10.1016/j.jtbi.2004.07.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2004] [Revised: 06/16/2004] [Accepted: 07/22/2004] [Indexed: 10/26/2022]
Abstract
Cellular multi-drug resistance (MDR), which often develops in cancer cells of patients subjected to anti-cancer treatment, remains a significant barrier to successful cancer therapy. One of the principal causes of cellular MDR development is an increased expression of ABC-transporter genes such as mdr1 and Bcrp1/Abcg2. Despite many years of intensive research, the natural biological role of mdr1 in the context of cancer has remained elusive. Some hints about this role came, however, from an observation that P-gp, the mdr1 encoded protein, is expressed widely in stem cells and from the discovery that P-gp possesses an anti-apoptotic activity independently of exogenous drug application. Here, we discuss our own and other groups' recently published works and propose an integrated view of mdr1 and Bcrp1/Abcg2 activity during tissue regeneration in normal tissues as part of a stress-induced regeneration genetic program and in cancerous tissues in response to cancer therapy.
Collapse
Affiliation(s)
- David Israeli
- Genethon Centre National de la Recherche Scientifique UMR 8115, 1 bis rue de l'Internationale, 91002 Evry cedex, France.
| | | | | | | |
Collapse
|
1013
|
Ebata KT, Zhang X, Nagano MC. Expression patterns of cell-surface molecules on male germ line stem cells during postnatal mouse development. Mol Reprod Dev 2005; 72:171-81. [PMID: 16010662 DOI: 10.1002/mrd.20324] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Spermatogonial stem cells (SSCs) are stem cells of the male germ line. In mice, SSCs are quiescent at birth but actively proliferate during the first postnatal week, while they rarely divide in adult, suggesting an age-dependent difference in SSC characteristics. As an approach to evaluate this possibility, we studied the expression pattern of cell-surface molecules on neonatal, pup, and adult mouse SSCs. Using immunomagnetic cell sorting, testis cells were selected for the expression of alpha(6) integrin, alpha(v) integrin, c-kit receptor tyrosine kinase (Kit), or a binding subunit of glial-cell-line-derived neurotrophic factor (GDNF) receptor, GFRalpha1. Selected cells were assayed for their stem cell activity using spermatogonial transplantation. The results showed that SSCs expressed alpha(6) integrin, but not alpha(v) integrin and Kit, regardless of age. The SSC activity in pup GFRalpha1(+) cells was higher than that in adult and neonatal cells, indicating that the expression pattern of GFRalpha1 varied age-dependently. To evaluate if SSCs show an age-dependent difference in their response to GDNF, we cultured highly enriched pup and adult SSCs with GDNF: we could not observe such an age-dependent difference in vitro. In addition, we failed to immunologically detect the expression of two types of GDNF receptor signaling subunits on SSCs. These results indicate that SSCs may change the expression patterns of cell-surface molecules during postnatal development, and suggest that GDNF receptor molecules may not be abundantly or specifically expressed in the in vivo population of mouse SSCs.
Collapse
Affiliation(s)
- Kevin T Ebata
- Department of Obstetrics and Gynecology, Royal Victoria Hospital, McGill University, 687 Pine Avenue West, Montreal, Quebec, Canada H3A 1A1
| | | | | |
Collapse
|
1014
|
Ho AD. Kinetics and symmetry of divisions of hematopoietic stem cells. Exp Hematol 2005; 33:1-8. [PMID: 15661392 DOI: 10.1016/j.exphem.2004.09.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2004] [Accepted: 08/27/2004] [Indexed: 11/20/2022]
Abstract
To fulfill the dual abilities to self-renew and to differentiate into cells of multiple lineages, stem cells must undergo, at some stage, asymmetric divisions to generate cells to sustain the stem cell pool as well as the various progeny cells of the distinct lineages. A central question in developmental biology is how a single cell can divide to produce two progeny cells that adopt different fates. Different daughter cells can theoretically arise by uneven distribution of determinants upon cell division, i.e., due to intrinsic factors, or become different upon subsequent exposure to environmental signals, i.e., due to extrinsic factors. Recent advances in the understanding of stem cell biology in Drosophila and murine models have served as a model for hematopoietic stem cell (HSC) development. Provided with advances in molecular and cellular biology, we have gained insight into the mechanisms governing self-renewing asymmetric divisions of primitive HSC. Direct contact with cellular determinants in the niche has been shown to play an essential role in the balance between self-renewing asymmetric division versus differentiation. Identification of the molecular interactions between stem cells and their niche will lead to an understanding of the mechanisms controlling the long-term destiny of stem cells. Ultimately, molecular signals triggered by adhesion and junction complexes are probably responsible for the specific adoption of differentiation pathways.
Collapse
Affiliation(s)
- Anthony D Ho
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
1015
|
Ha Y, Kim YS, Cho JM, Yoon SH, Park SR, Yoon DH, Kim EY, Park HC. Role of granulocyte—macrophage colony—stimulating factor in preventing apoptosis and improving functional outcome in experimental spinal cord contusion injury. J Neurosurg Spine 2005; 2:55-61. [PMID: 15658127 DOI: 10.3171/spi.2005.2.1.0055] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object. Granulocyte—macrophage colony—stimulating factor (GM-CSF) is a potent hemopoietic cytokine that stimulates stem cell proliferation in the bone marrow and inhibits apoptotic cell death in leukocytes. Its effects in the central nervous system, however, are still unclear. The present study was undertaken to determine if GM-CSF can rescue neuronal cells from apoptosis and improve neurological function in a spinal cord injury (SCI) model.
Methods. To study the effect of GM-CSF on apoptotic neuronal death, the authors used a staurosporine-induced neuronal death model in an N2A cell line (in vitro) and in a rat SCI model (in vivo). The N2A cells were preincubated with GM-CSF for 60 minutes before being exposed to staurosporine for 24 hours. To inhibit GM-CSF, N2A cells were pretreated with antibodies against the GM-CSF receptor for 60 minutes. Clip compression was used to induce SCI. Animals were treated with daily doses of GM-CSF (20 µg/day) for 5 days. The number of apoptotic cells in the spinal cord and neurological improvements were assessed.
Pretreatment with GM-CSF was found to protect N2A cells significantly from apoptosis, and neutralizing antibodies for the GM-CSF receptors inhibited the rescuing effect of GM-CSF on apoptosis. In the rat SCI model, neurological function improved significantly in the GM-CSF—treated group compared with controls treated with phosphate-buffered saline. Terminal deoxynucleotidyl transferase—mediated deoxyuridine triphosphate nick-end labeling staining showed that GM-CSF administration reduced apoptosis in the injured spinal cord.
Conclusions. Treatment of SCI with GM-CSF showed beneficial effects. Neuronal protection against apoptosis is viewed as a likely mechanism underlying the therapeutic effect of GM-CSF in SCI.
Collapse
Affiliation(s)
- Yoon Ha
- Departments of Neurosurgery and Physiology, Inha University College of Medicine, Inchon, Korea.
| | | | | | | | | | | | | | | |
Collapse
|
1016
|
Itokazu Y, Kitada M, Dezawa M, Mizoguchi A, Matsumoto N, Shimizu A, Ide C. Choroid plexus ependymal cells host neural progenitor cells in the rat. Glia 2005; 53:32-42. [PMID: 16158416 DOI: 10.1002/glia.20255] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We previously demonstrated that choroid plexus epithelial (modified ependymal) cells (CPECs) differentiated into astrocytes after grafting into the spinal cord. In the present study, we examined whether CPECs from rats at postnatal 1 day (P1), 7 day (P7), and 8 weeks (P8W) can function as neural progenitor cells that give rise to neurons and glial cells. Cell spheres were produced in cultures of whole tissue of the choroid plexus from the fourth ventricle of rats at each postnatal period. beta-tubulin class III (Tuj-1), glial fibrillary acid protein (GFAP)-, and O4-positive cells differentiated from cell spheres in the differentiation medium. We produced a monoclonal antibody 3E6 specifically labeling microvilli of CPECs. Using this monoclonal antibody, CPECs were isolated from the choroid plexus of P8W rats by cell sorter (FACS). Immunocytochemistry confirmed that there was no contamination from fibroblasts, endothelial cells, macrophages, or Schwann cells in the FACS-isolated 3E6-labeled cells. Cell spheres formed in the cultures of these 3E6-labeled CPECs. After expansion, these cell spheres gave rise to Tuj-1- (5%), GFAP- (45%), and O4-positive cells (0.16%). The remaining cells (45%) were unlabeled neural or glial markers. Some CPECs of the P8W rat were immunohistochemically stained with lineage-associated markers of Musashi-1 and epidermal growth factor-receptor (EGF-R). In addition, infusion of EGF or fibroblast growth factor-2 (FGF2) into the ventricle increased the number of bromodeoxyuridine (BrdU)-positive cells among CPECs from 0.03% (untreated) to 1.14% (38-fold, EGF) and 1.03% (35-fold, FGF2), respectively. These findings indicate that neural progenitor cells exist among CPECs in the rat.
Collapse
Affiliation(s)
- Yutaka Itokazu
- Department of Anatomy and Neurobiology, Kyoto University, Graduate School of Medicine, Kyoto, Japan.
| | | | | | | | | | | | | |
Collapse
|
1017
|
Brendel C, Kuklick L, Hartmann O, Kim TD, Boudriot U, Schwell D, Neubauer A. Distinct gene expression profile of human mesenchymal stem cells in comparison to skin fibroblasts employing cDNA microarray analysis of 9600 genes. Gene Expr 2005; 12:245-57. [PMID: 16355723 PMCID: PMC6009126 DOI: 10.3727/000000005783992043] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Broad differentiation capacity has been described for mesenchymal stem cells (MSC) from human bone marrow. We sought to identify genes associated with the immature state and pluripotency of this cell type. To prove the pluripotent state of the MSC, differentiation into osteocytes, adipocytes, and chondrocytes was performed in vitro. In contrast, normal skin cells did not harbor these differentiation abilities. We compared the expression profile of human bone marrow MSC with cDNA from one primary human skin cell line as control, using a cDNA chip providing 9600 genes. The identity of all relevant genes was confirmed by direct sequencing. Data of gene array expression were corroborated employing quantitative PCR analysis. About 80 genes were differently expressed more than threefold in MSC compared to mature skin fibroblasts. Interestingly, primary human MSC were found to upregulate a number of genes important for embryogenesis such as distal-less homeo box 5, Eyes absent homolog 2, inhibitor of DNA binding 3, and LIM protein. In contrast, mesenchymal lineage genes were downregulated in MSC in comparison to skin cells. We also detected expression of some genes involved in neural development, indicating the broad differentiation capabilities of MSC. We conclude that human mesenchymal stem cells harbor an expression profile distinct from mature skin fibroblast, and genes associated with developmental processes and stem cell function are highly expressed in adult mesenchymal stem cells.
Collapse
Affiliation(s)
- Cornelia Brendel
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, Baldingerstrasse, Marburg, Germany
| | | | | | | | | | | | | |
Collapse
|
1018
|
Metcalfe SM, De S Muthukumarana PA. Transplantation tolerance: gene expression profiles comparing allotolerance vs. allorejection. Int Immunopharmacol 2005; 5:33-9. [PMID: 15589457 DOI: 10.1016/j.intimp.2004.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
An understanding of the molecular basis of immune regulation will allow development of therapies for diseases caused by immune dysregulation and for therapeutic exploitation of the immune response in transplantation of organ grafts or stem cells. To identify critical regulatory factors in immunity, we have used a mouse model wherein infectious regulatory tolerance is inducible by CD4/CD8 blockade in recipients of vascularised heart grafts. Once established, this transplantation tolerance is robust and isolated "tolerant" spleen cells show powerful immune regulatory properties, being able to impose donor-specific allotolerance upon fully immune competent naive recipients. Here, we present a compound comparison of four gene arrays (tolerance vs. rejection, at 48 h, and at 123 h) where a relatively small number of differentially expressed genes occurred. In rejection, there was a strong progressive amplification of IFNgamma and granzyme B mRNAs. In tolerance, both ELKL motif kinase and axotrophin occurred in the group of upregulated genes. Mice lacking ELKL motif kinase develop autoimmune disease, whilst axotrophin is a newly discovered stem cell gene that has only been explored in the context of neural development. This gene expression data is the first to demonstrate a link between axotrophin and regulatory tolerance and, since axotrophin, LIF, STAT3 and c-kit each function in stem cells, we propose that common mechanisms play a central role both in developmental regulation of stem cells, and in immune regulation.
Collapse
Affiliation(s)
- Su M Metcalfe
- Department of Surgery, University of Cambridge, Box 202, Level E9, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK.
| | | |
Collapse
|
1019
|
Bianco C, Strizzi L, Normanno N, Khan N, Salomon DS. Cripto-1: an oncofetal gene with many faces. Curr Top Dev Biol 2005; 67:85-133. [PMID: 15949532 DOI: 10.1016/s0070-2153(05)67003-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human Cripto-1 (CR-1), a member of the epidermal growth factor (EGF)-CFC family, has been implicated in embryogenesis and in carcinogenesis. During early vertebrate development, CR-1 functions as a co-receptor for Nodal, a transforming growth factor beta (TGFbeta) family member and is essential for mesoderm and endoderm formation and anterior-posterior and left-right axis establishment. In adult tissues, CR-1 is expressed at a low level in all stages of mammary gland development and expression increases during pregnancy and lactation. Overexpression of CR-1 in mouse mammary epithelial cells leads to their transformation in vitro and, when injected into mammary glands, produces ductal hyperplasias. CR-1 can also enhance migration, invasion, branching morphogenesis and epithelial to mesenchymal transition (EMT) of several mouse mammary epithelial cell lines. Furthermore, transgenic mouse studies have shown that overexpression of a human CR-1 transgene in the mammary gland under the transcriptional control of the mouse mammary tumor virus (MMTV) promoter results in mammary hyperplasias and papillary adenocarcinomas. Finally, CR-1 is expressed at high levels in approximately 50 to 80% of different types of human carcinomas, including breast, cervix, colon, stomach, pancreas, lung, ovary, and testis. In conclusion, EGF-CFC proteins play dual roles as embryonic pattern formation genes and as oncogenes. While during embryogenesis EGF-CFC proteins perform specific and regulatory functions related to cell and tissue patterning, inappropriate expression of these molecules in adult tissues can lead to cellular proliferation and transformation and therefore may be important in the etiology and/or progression of cancer.
Collapse
Affiliation(s)
- Caterina Bianco
- Tumor Growth Factor Section, Mammary Biology & Tumorigenesis Laboratory Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
1020
|
Hart AH, Hartley L, Ibrahim M, Robb L. Identification, cloning and expression analysis of the pluripotency promoting Nanog genes in mouse and human. Dev Dyn 2004; 230:187-98. [PMID: 15108323 DOI: 10.1002/dvdy.20034] [Citation(s) in RCA: 233] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The murine Nanog gene, a member of the homeobox family of DNA binding transcription factors, has been shown recently to maintain pluripotency of embryonic stem cells. We have used a sequence homology and expression screen to identify and clone the mouse and human Nanog genes and characterized their phylogenetic context and expression patterns. We report here the gene structure and expression patterns of the mouse Nanog gene, the human Nanog and Nanog2 genes, and six processed human Nanog pseudogenes. Mouse Nanog expression is high in undifferentiated embryonic stem cells and is down-regulated during embryonic stem cell differentiation, concomitant with loss of pluripotency. Murine embryonic Nanog expression is detected in the inner cell mass of the blastocyst. After implantation, Nanog is detectable at embryonic day (E) 6 in proximal epiblast in the region of the presumptive primitive streak. Expression extends distally as the streak elongates during gastrulation and remains restricted to epiblast. Nanog RNA is down-regulated in cells ingressing through the streak to form mesoderm and definitive endoderm. Nanog expression also marks the pluripotent germ cells of the nascent gonad at E11.5-E12.5 and is highly expressed in germ cell tumour and teratoma-derived cell lines. Reverse transcriptase-polymerase chain reaction analysis detected mouse Nanog expression at low levels in several adult tissues. The human Nanog genes are expressed in embryonic stem cells and down-regulated in all adult tissues and differentiated cell lines examined. High levels of human Nanog expression were detected by Northern analysis in the undifferentiated N-Tera embryonal carcinoma cell line. The conservation in gene sequence, structure, and expression of mouse and human Nanog and Nanog2 genes may reflect a common role in the maintenance of pluripotency in both species.
Collapse
Affiliation(s)
- Adam H Hart
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
| | | | | | | |
Collapse
|
1021
|
Gurok U, Steinhoff C, Lipkowitz B, Ropers HH, Scharff C, Nuber UA. Gene expression changes in the course of neural progenitor cell differentiation. J Neurosci 2004; 24:5982-6002. [PMID: 15229246 PMCID: PMC6729244 DOI: 10.1523/jneurosci.0809-04.2004] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The molecular changes underlying neural progenitor differentiation are essentially unknown. We applied cDNA microarrays with 13,627 clones to measure dynamic gene expression changes during the in vitro differentiation of neural progenitor cells that were isolated from the subventricular zone of postnatal day 7 mice and grown in vitro as neurospheres. In two experimental series in which we withdrew epidermal growth factor and added the neurotrophins Neurotrophin-4 or BDNF, four time points were investigated: undifferentiated cells grown as neurospheres, and cells 24, 48, and 96 hr after differentiation. Expression changes of selected genes were confirmed by semiquantitative RT-PCR. Ten different groups of gene expression dynamics obtained by cluster analysis are described. To correlate selected gene expression changes to the localization of respective proteins, we performed immunostainings of cultured neurospheres and of brain sections from adult mice. Our results provide new insights into the genetic program of neural progenitor differentiation and give strong hints to as yet unknown cellular communications within the adult subventricular zone stem cell niche.
Collapse
Affiliation(s)
- Ulf Gurok
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
1022
|
Abstract
Neural stem cells (NSC) are a tissue-specific subtype of self-renewing and multipotent cells that can give rise to all neural populations. In this review, the importance of maintaining cell-cell contacts in the study of NSC is highlighted, and data obtained from some crucial single-cell studies is compared to results obtained from neurospheres, where aggregates of NSC are grown in suspension. In particular, results that indicate how this culture system may be well suited to analyze NSC plasticity, cell-cell, and cell-extracellular matrix (ECM) interactions are pointed out, and the hypothesis that cell-cell and cell-ECM contacts may be essential for NSC maintenance, survival, and proliferation is highlighted. Finally, it is suggested that neurospheres might play a role in the study of context-dependent behavior of NSC in niches by providing a system where NSC can be challenged chemically or biologically and analyzed in vitro, in a time- and context-dependent manner.
Collapse
Affiliation(s)
- Lia Scotti Campos
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom.
| |
Collapse
|
1023
|
Szaniszlo P, Wang N, Sinha M, Reece LM, Van Hook JW, Luxon BA, Leary JF. Getting the right cells to the array: Gene expression microarray analysis of cell mixtures and sorted cells. Cytometry A 2004; 59:191-202. [PMID: 15170598 DOI: 10.1002/cyto.a.20055] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Most biological samples are cell mixtures. Some basic questions are still unanswered about analyzing these heterogeneous samples using gene expression microarray technology (MAT). How meaningful is a cell mixture's overall gene expression profile (GEP)? Is it necessary to purify the cells of interest before microarray analysis, and how much purity is needed? How much does the purification itself distort the GEP, and how well can the GEP of a small cell subset be recovered? METHODS Model cell mixtures with different cell ratios were analyzed by both spotted and Affymetrix MAT. GEP distortion during cell purification and GEPs of purified cells were studied. CD34+ cord blood cells were purified and analyzed by MAT. RESULTS GEPs for mixed cell populations were found to mirror the cell ratios in the mixture. Over 75% pure samples were indistinguishable from pure cells by their overall GEP. Cell purification preserved the GEP. The GEPs of small cell subsets could be accurately recovered by cell sorting both from model cell mixtures and from cord blood. CONCLUSIONS Purification of small cell subsets from a mixture prior to MAT is necessary for meaningful results. Even completely hidden GEPs of small cell subpopulations can be recovered by cell sorting.
Collapse
Affiliation(s)
- Peter Szaniszlo
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, 77555, USA
| | | | | | | | | | | | | |
Collapse
|
1024
|
Wilson A, Murphy MJ, Oskarsson T, Kaloulis K, Bettess MD, Oser GM, Pasche AC, Knabenhans C, Macdonald HR, Trumpp A. c-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation. Genes Dev 2004; 18:2747-63. [PMID: 15545632 PMCID: PMC528895 DOI: 10.1101/gad.313104] [Citation(s) in RCA: 605] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The activity of adult stem cells is essential to replenish mature cells constantly lost due to normal tissue turnover. By a poorly understood mechanism, stem cells are maintained through self-renewal while concomitantly producing differentiated progeny. Here, we provide genetic evidence for an unexpected function of the c-Myc protein in the homeostasis of hematopoietic stem cells (HSCs). Conditional elimination of c-Myc activity in the bone marrow (BM) results in severe cytopenia and accumulation of HSCs in situ. Mutant HSCs self-renew and accumulate due to their failure to initiate normal stem cell differentiation. Impaired differentiation of c-Myc-deficient HSCs is linked to their localization in the differentiation preventative BM niche environment, and correlates with up-regulation of N-cadherin and a number of adhesion receptors, suggesting that release of HSCs from the stem cell niche requires c-Myc activity. Accordingly, enforced c-Myc expression in HSCs represses N-cadherin and integrins leading to loss of self-renewal activity at the expense of differentiation. Endogenous c-Myc is differentially expressed and induced upon differentiation of long-term HSCs. Collectively, our data indicate that c-Myc controls the balance between stem cell self-renewal and differentiation, presumably by regulating the interaction between HSCs and their niche.
Collapse
Affiliation(s)
- Anne Wilson
- Genetics and Stem Cell Laboratory, Swiss Institute for Experimental Cancer Research (ISREC), CH-1066 Epalinges, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1025
|
Abstract
Hematopoietic stem cells (HSC) must balance self-renewal and differentiation to provide sufficient primitive cells to sustain hematopoiesis, while generating more mature cells with specialized capabilities. The enhanced self-renewal capacity of primitive HSCs enables their ability to sustain hematopoiesis throughout decades of life and their ability to repopulate a host when used therapeutically in bone marrow transplantation. However, hematopoietic cell perturbations resulting in unchecked self-renewal participate in leukemogenesis. While mechanisms governing self-renewal are still being uncovered, they are thought to bear relationship to the malignant process in a variety of tumor types and may therefore provide useful therapeutic targets in putative cancer stem cells. This review discusses molecular mechanisms recently defined to participate in HSC governance and highlights features of stem cell interactions with the microenvironment that may help guide therapies directed at HSCs.
Collapse
Affiliation(s)
- E C Attar
- Center for Regenerative Medicine and Technology and Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | | |
Collapse
|
1026
|
Abstract
Stem cells in animal tissues are often located and controlled by special tissue microenvironments known as niches. Studies of stem cell niches in model systems such as Drosophila have revealed adhesive interactions, cell cycle modifications and intercellular signals that operate to control stem cell behavior. Candidate niches and regulatory molecules have also been identified in many mammalian tissues, including bone marrow, skin, gut and brain. While niches are an ancient evolutionary device with conserved features across diverse organisms, we suggest that certain niches display important differences in their organization and function.
Collapse
Affiliation(s)
- Benjamin Ohlstein
- Howard Hughes Medical Institute, Carnegie Institution of Washington, Department of Embryology, 115 W. University Pkwy., Baltimore, Maryland 21210, USA
| | | | | | | |
Collapse
|
1027
|
Abstract
Embryonic stem cells hold potential in the fields of regenerative medicine, developmental biology, tissue regeneration, disease pathogenicity, and drug discovery. Embryonic stem (ES) cell lines are now available in primates, including man, rhesus, and cynomologous monkeys. Monkey ES cells serve as invaluable clinically relevant models for studies that can't be conducted in humans because of practical or ethical limitations, or in rodents because of differences in physiology and anatomy. Here, we review the current status of nonhuman primate research with ES cells, beginning with a description of their isolation, characterization, and availability. Substantial limitations still plague the use of primate ES cells, such as their required growth on feeder layers, poor cloning efficiency, and restricted availability. The ability to produce homogenous populations of both undifferentiated as well as differentiated phenotypes is an important challenge, and genetic approaches to achieving these objectives are discussed. Finally, safety, efficiency, and feasibility issues relating to the transplantation of ES-derived cells are considered.
Collapse
Affiliation(s)
- Don P Wolf
- Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon 97006, USA.
| | | | | | | |
Collapse
|
1028
|
Meeson AP, Hawke TJ, Graham S, Jiang N, Elterman J, Hutcheson K, Dimaio JM, Gallardo TD, Garry DJ. Cellular and Molecular Regulation of Skeletal Muscle Side Population Cells. Stem Cells 2004; 22:1305-20. [PMID: 15579648 DOI: 10.1634/stemcells.2004-0077] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Muscle progenitor cells (satellite cells) function in the maintenance and repair of adult skeletal muscle. Side population (SP) cells are enriched in repopulating activity and also reside in adult skeletal muscle. In this study, we observed that Abcg2 is a determinant of the SP cell phenotype. Using reverse transcription polymerase chain reaction and immunohistochemical techniques, we localized Abcg2-expressing cells in the interstitium and in close approximation to the vasculature of adult skeletal muscle. Muscle SP cells are able to differentiate into myotubes and increase in number after cardiotoxin-induced muscle injury. Similar to myogenic progenitor cells, muscle SP cells express Foxk1 and are decreased in number in Foxk1 mutant skeletal muscle. Using emerging technologies, we examine the molecular signature of muscle SP cells from normal, injured, and Foxk1 mutant skeletal muscle to define common and distinct molecular programs. We propose that muscle SP cells are progenitor cells that participate in repair and regeneration of adult skeletal muscle.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 2
- ATP-Binding Cassette Transporters/physiology
- Animals
- Cell Differentiation
- Cell Separation
- Cobra Cardiotoxin Proteins/pharmacology
- Flow Cytometry
- Forkhead Transcription Factors
- Green Fluorescent Proteins/metabolism
- Immunohistochemistry
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscles/pathology
- Muscular Dystrophies/pathology
- Neoplasm Proteins/physiology
- Nuclear Proteins/genetics
- Nucleic Acid Hybridization
- Phenotype
- RNA/metabolism
- Regeneration
- Reverse Transcriptase Polymerase Chain Reaction
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/physiology
- Stem Cells/cytology
- Time Factors
- Transcription Factors/genetics
- Transcription, Genetic
Collapse
Affiliation(s)
- Annette P Meeson
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1029
|
Stewart R, Coyne L, Lako M, Halliwell RF, Przyborski SA. Human Embryonal Carcinoma Stem Cells Expressing Green Fluorescent Protein Form Functioning Neurons In Vitro: A Research Tool for Co-culture Studies. Stem Cells Dev 2004; 13:646-57. [PMID: 15684832 DOI: 10.1089/scd.2004.13.646] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Neural differentiation is controlled by complex molecular mechanisms that determine cell fate and diversity within the nervous system. Interactions between developing tissues play an important role in regulating this process. In vitro co-culture experiments offer a method to study cell differentiation and function under controlled conditions, with the additional benefit of investigating how interactions between populations of cells influence cell growth and behavior. However, it can often be difficult to distinguish between populations of co-cultured cells. Here we report the development of a human embryonal carcinoma (EC) stem cell line (named TERA2.cl.SP12-GFP) that expresses the genetic marker, green fluorescent protein (GFP). Here, we demonstrate that TERA2.cl.SP12-GFP stem cells stably express GFP and that this remains detectable during retinoic acid-induced differentiation. Regulated expression of neural markers during cell development correlated with the formation of morphologically identifiable neurons. Populations of post-mitotic GFP-positive neurons were readily purified and electrophysiological characterization confirmed that such neurons were functionally active. Thus, cultured TERA2.cl.SP12-GFP cells can be readily distinguished from alternative cell types in vitro and provide an amenable system for live cell imaging to study the development and function of human neurons in isolation, and in co-culture with other tissue types.
Collapse
Affiliation(s)
- R Stewart
- School of Biological and Biomedical Science, University of Durham, South Road, Durham DH1 3LE, UK
| | | | | | | | | |
Collapse
|
1030
|
Beggs ML, Nagarajan R, Taylor-Jones JM, Nolen G, Macnicol M, Peterson CA. Alterations in the TGFbeta signaling pathway in myogenic progenitors with age. Aging Cell 2004; 3:353-61. [PMID: 15569352 DOI: 10.1111/j.1474-9728.2004.00135.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Myogenic progenitors in adult muscle are necessary for the repair, maintenance and hypertrophy of post-mitotic muscle fibers. With age, fat deposition and fibrosis contribute to the decline in the integrity and functional capacity of muscles. In a previous study we reported increased accumulation of lipid in myogenic progenitors obtained from aged mice, accompanied by an up-regulation of genes involved in adipogenic differentiation. The present study was designed to extend our understanding of how aging affects the fate and gene expression profile of myogenic progenitors. Affymetrix murine U74 Genechip analysis was performed using RNA extracted from myogenic progenitors isolated from adult (8-month-old) and aged (24-month-old) DBA/2JNIA mice. The cells from the aged animals exhibited major alterations in the expression level of many genes directly or indirectly involved with the TGFbeta signaling pathway. Our data indicate that with age, myogenic progenitors acquire the paradoxical phenotype of being both TGFbeta activated based on overexpression of TGFbeta-inducible genes, but resistant to the differentiation-inhibiting effects of exogenous TGFbeta. The overexpression of TGFbeta-regulated genes, such as connective tissue growth factor, may play a role in increasing fibrosis in aging muscle.
Collapse
Affiliation(s)
- Marjorie L Beggs
- Department of Geriatrics, Reynolds Center on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | | | | | | | | | |
Collapse
|
1031
|
Carter LA, MacDonald JL, Roskams AJ. Olfactory horizontal basal cells demonstrate a conserved multipotent progenitor phenotype. J Neurosci 2004; 24:5670-83. [PMID: 15215289 PMCID: PMC6729216 DOI: 10.1523/jneurosci.0330-04.2004] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Stem cells of adult regenerative organs share a common goal but few established conserved mechanisms. Within the neural stem cell niche of the mouse olfactory epithelium, we identified a combination of extracellular matrix (ECM) receptors that regulate adhesion and mitosis in non-neural stem cells [intercellular adhesion molecule-1 (ICAM-1), beta1, beta4, and alpha-1, -3, and -6 integrins] and on horizontal basal cells (HBCs), candidate olfactory neuro-epithelial progenitors. Using ECM receptors as our guide, we recreated a defined microenvironment in vitro that mimics olfactory basal lamina and, when supplemented with epidermal growth factor, transforming growth factor alpha, and leukemia inhibitory factor, allows us to preferentially expand multiple clonal adherent colony phenotypes from individual ICAM-1+ and ICAM-1+/beta1 integrin+-selected HBCs. The most highly mitotic colony-forming HBCs demonstrate multipotency, spontaneously generating more ICAM-positive presumptive HBCs, a combination of olfactory neuroglial progenitors, and neurons of olfactory and potentially nonolfactory phenotypes. HBCs thus possess a conserved adhesion receptor expression profile similar to non-neural stem cells, preferential self-replication in an in vitro environment mimicking their in vivo niche, and contain subpopulations of cells that can produce multiple differentiated neuronal and glial progeny from within and beyond the olfactory system in vitro.
Collapse
Affiliation(s)
- Lindsay A Carter
- Department of Zoology and Center for Molecular Medicine and Therapeutics, University of British Columbia, V6T 1Z4 Vancouver, Canada
| | | | | |
Collapse
|
1032
|
Hiratani I, Leskovar A, Gilbert DM. Differentiation-induced replication-timing changes are restricted to AT-rich/long interspersed nuclear element (LINE)-rich isochores. Proc Natl Acad Sci U S A 2004; 101:16861-6. [PMID: 15557005 PMCID: PMC534734 DOI: 10.1073/pnas.0406687101] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The replication timing of some genes is developmentally regulated, but the significance of replication timing to cellular differentiation has been difficult to substantiate. Studies have largely been restricted to the comparison of a few genes in established cell lines derived from different tissues, and most of these genes do not change replication timing. Hence, it has not been possible to predict how many or what types of genes might be subject to such control. Here, we have evaluated the replication timing of 54 tissue-specific genes in mouse embryonic stem cells before and after differentiation to neural precursors. Strikingly, genes residing within isochores rich in GC and poor in long interspersed nuclear elements (LINEs) did not change their replication timing, whereas half of genes within isochores rich in AT and long interspersed nuclear elements displayed programmed changes in replication timing that accompanied changes in gene expression. Our results provide direct evidence that differentiation-induced autosomal replication-timing changes are a significant part of mammalian development, provide a means to predict genes subject to such regulation, and suggest that replication timing may be more related to the evolution of metazoan genomes than to gene function or expression pattern.
Collapse
Affiliation(s)
- Ichiro Hiratani
- Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | | | | |
Collapse
|
1033
|
Damiola F, Keime C, Gonin-Giraud S, Dazy S, Gandrillon O. Global transcription analysis of immature avian erythrocytic progenitors: from self-renewal to differentiation. Oncogene 2004; 23:7628-43. [PMID: 15378009 DOI: 10.1038/sj.onc.1208061] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The molecular mechanisms regulating the cell fate decision between self-renewal and differentiation/apoptosis in stem and progenitor cells are poorly understood. Here, we report the first comprehensive identification of genes potentially involved in the switch from self-renewal toward differentiation of primary, non-immortalized erythroid avian progenitor cells (T2EC cells). We used the Serial Analysis of Gene Expression (SAGE) technique in order to identify and quantify the genome fraction functionally active in a self-renewing versus a differentiating cell population. We generated two SAGE libraries and sequenced a total of 37,589 tags, thereby obtaining the first transcriptional profile characterization of a chicken cell. Tag identification was performed using a new relational database (Identitag) developed in the laboratory, which allowed a highly satisfactory level of identification. Among 123 differentially expressed genes, 11 were investigated further and for nine of them the differential expression was subsequently confirmed by real-time PCR. The comparison of tag abundance between the two libraries revealed that only a small fraction of transcripts was differentially expressed. The analysis of their functions argue against a prominent role for a master switch in T2EC cells decision-making, but are in favor of a critical role for coordinated small variations in a relatively small number of genes that can lead to essential cellular identity changes.
Collapse
Affiliation(s)
- Francesca Damiola
- Equipe 'Signalisations et identités cellulaires', Centre de Génétique Moléculaire et Cellulaire CNRS UMR 5534, France.
| | | | | | | | | |
Collapse
|
1034
|
Tao W, Wang M, Voss ED, Cocklin RR, Smith JA, Cooper SH, Broxmeyer HE. Comparative Proteomic Analysis of Human CD34+Stem/Progenitor Cells and Mature CD15+Myeloid Cells. Stem Cells 2004; 22:1003-14. [PMID: 15536191 DOI: 10.1634/stemcells.22-6-1003] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Human CD34(+) cells, highly enriched for hematopoietic stem and progenitors, and CD15(+) cells, more terminally differentiated myeloid cells in blood, represent distinct maturation/differentiation stages. A proteomic approach was used to identify proteins differentially present in these two populations from human cord blood. Cytosolic proteins were extracted and subjected to two-dimensional gel electrophoresis followed by mass spectrometry. On average, 460 protein spots on each gel were detected; 112 and 15 proteins, respectively, were found to be differentially expressed or post-translationally modified in CD34(+) and CD15(+) cells. This suggests that CD34(+) cells have a relatively larger proteome than mature CD15(+) myeloid cells and production of many stem/progenitor cell-associated proteins ceases or is dramatically down-regulated as the CD34(+) cells undergo differentiation. Of approximately 140 protein spots, 47 different proteins were positively identified by mass spectrometry and database search; these proteins belong to several functional categories, including cell signaling, transcription factors, cytoskeletal proteins, metabolism, protein folding, and vesicle trafficking. Multiple heat shock proteins and chaperones, as well as proteins important for intracellular trafficking, were predominantly present in CD34(+) cells. Most of the identified proteins in CD34(+) cells are expressed in germ cell tumors, as well as in embryonal carcinoma and neuroblastoma. Approximately eight novel proteins, whose functions are unknown, were identified. This study presents, for the first time, global cellular protein expression patterns in human CD34(+) and CD15(+) cells, which should help to better understand intracellular processes involved in myeloid differentiation and add insight into the functional capabilities of these distinct cell types.
Collapse
Affiliation(s)
- Wen Tao
- Department of Microbiology and Immunology, Indiana University School of Medicine, 950 West Walnut Street, Indianapolis, Indiana 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
1035
|
Boman BM, Walters R, Fields JZ, Kovatich AJ, Zhang T, Isenberg GA, Goldstein SD, Palazzo JP. Colonic crypt changes during adenoma development in familial adenomatous polyposis: immunohistochemical evidence for expansion of the crypt base cell population. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:1489-98. [PMID: 15509520 PMCID: PMC1618673 DOI: 10.1016/s0002-9440(10)63407-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/29/2004] [Indexed: 01/14/2023]
Abstract
Familial adenomatous polyposis patients, who have a germline APC mutation, develop adenomas in normal-appearing colonic mucosa, and in the process usually acquire a mutation in the other APC allele as well. Nonetheless, the cellular mechanisms that link these initiating genetic changes with the earliest tissue changes (upward shift in the labeling index) in colon tumorigenesis are unclear. Based on the tenet that colorectal cancer originates from crypt stem cells (SCs) and on our kinetic modeling, we hypothesized that overpopulation of mutant colonic SCs is the missing link. Directly testing this hypothesis requires measuring changes in the size of the SC population, but specific markers for human colonic SCs are lacking. Hence, we used immunohistochemical mapping to study crypt base cells, of which SCs are a subset. Using colectomy specimens from 16 familial adenomatous polyposis and 11 control cases, we determined the topographic profiles of various cell populations along the crypt axis and the proportions of each cell type. In the formation of adenomatous crypts, the distribution of cells expressing crypt base cell markers (MSH2, Bcl-2, survivin) expanded toward the crypt surface and showed the greatest proportional increase (fivefold to eightfold). Cells expressing a marker for the upper crypt (p27(kip1)) shifted to the crypt bottom and showed the smallest increase. This suggests that: 1) during adenoma development, APC mutations cause expansion of the crypt base cell population, including crypt SCs; 2) SC overpopulation can explain the shifts in pattern of proliferative crypt cell populations in early colon tumorigenesis, and 3) mutant crypt SCs clonally expand to form colonic adenomas and carcinomas.
Collapse
Affiliation(s)
- Bruce M Boman
- Division of Genetic and Preventive Medicine, Kimmel Cancer Center, 1100 Walnut St., Suite 400, Thomas Jefferson University, Philadelphia PA 19107, USA.
| | | | | | | | | | | | | | | |
Collapse
|
1036
|
Abstract
Stem cells are endowed with self-renewal and multipotential differentiation capacities. Contrary to the expectation that stem cells would selectively express specific genes, these cells have a highly promiscuous gene-expression pattern. Here, I suggest that the transient stem cell state, termed the 'stem state', may be assumed by any cell and that the search for specific genes expressed by all stem cells, which would characterize the stem cell as a cell type, might be futile.
Collapse
Affiliation(s)
- Dov Zipori
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
1037
|
Carpenter L, Zernicka-Goetz M. Directing pluripotent cell differentiation using ?diced RNA? in transient transfection. Genesis 2004; 40:157-63. [PMID: 15515021 DOI: 10.1002/gene.20078] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Embryonic stem (ES) and embryonic carcinoma (EC) cells are pluripotent and have the capacity to differentiate into many cell types. The ability to direct their differentiation should have considerable practical applications. Here, we first report the use of diced short interfering RNAi against Oct4 in a transient approach, to direct differentiation of ES towards the trophectoderm lineage. We then apply this approach to downregulate Smad4 in mouse P19 EC cells. We have found that this leads to an increase in the levels of Pax6 (a neuroectoderm marker), reduction in the levels of Brachyury (a mesoderm marker), and a 3-fold increase in the number of betaIII tubulin-positive colonies when these cells were allowed to differentiate. This indicates a redirection of cell fate towards the neuroectoderm lineage. Thus, transient RNAi could provide a valuable tool to direct pluripotent cells along specific pathways of differentiation while circumventing permanent genetic changes.
Collapse
Affiliation(s)
- Lee Carpenter
- University of Cambridge, Department of Genetics, Downing Street, Cambridge CB2 3EH, UK
| | | |
Collapse
|
1038
|
Abstract
PURPOSE OF REVIEW Recent studies that might help in the search for stem cells in adult kidney and clarify the origin of proliferating cells during kidney repair are reviewed. RECENT FINDINGS Some of the most notable recent findings are as follows: (1) the 'stemness' profile may be determined by approximately 250 genes; (2) organ-specific stem-cell growth and differentiation are stimulated during the reparative phase following transient injury; (3) two bone marrow stem-cell types show a remarkable degree of differentiation potential; (4) some organs contain resident marrow-derived stem cells, and their differentiation potential may only be expressed during repair; (5) the metanephric mesenchyme contains pluripotent and self-renewing stem cells; (6) marrow-derived cells invade the kidney and differentiate into mesangial and tubular epithelial cells, and these processes are increased following renal injury; and (7) epithelial-to-mesenchymal transition generates renal fibroblasts. SUMMARY While it remains unknown whether there is a stem cell in the adult kidney, characterization of the cell populations involved in renal repair and misrepair is allowing a new understanding of the mechanisms that are responsible for renal homeostasis. The most surprising results suggest a very prominent role for cells exogenous to the kidney. Two recently published transcription profiles of 'stemness' and the phenotype of pluripotent metanephric mesenchymal cells may help in the search for adult renal stem cells.
Collapse
Affiliation(s)
- Juan A Oliver
- Department of Medicine, College of Physicians and Surgeons, Columbia University, 630 West 168 Street, New York, NY 10032, USA.
| |
Collapse
|
1039
|
Czyz J, Wiese C, Rolletschek A, Blyszczuk P, Cross M, Wobus AM. Potential of embryonic and adult stem cells in vitro. Biol Chem 2004; 384:1391-409. [PMID: 14669982 DOI: 10.1515/bc.2003.155] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recent developments in the field of stem cell research indicate their enormous potential as a source of tissue for regenerative therapies. The success of such applications will depend on the precise properties and potentials of stem cells isolated either from embryonic, fetal or adult tissues. Embryonic stem cells established from the inner cell mass of early mouse embryos are characterized by nearly unlimited proliferation, and the capacity to differentiate into derivatives of essentially all lineages. The recent isolation and culture of human embryonic stem cell lines presents new opportunities for reconstructive medicine. However, important problems remain; first, the derivation of human embryonic stem cells from in vitro fertilized blastocysts creates ethical problems, and second, the current techniques for the directed differentiation into somatic cell populations yield impure products with tumorigenic potential. Recent studies have also suggested an unexpectedly wide developmental potential of adult tissue-specific stem cells. Here too, many questions remain concerning the nature and status of adult stem cells both in vivo and in vitro and their proliferation and differentiation/transdifferentiation capacity. This review focuses on those issues of embryonic and adult stem cell biology most relevant to their in vitro propagation and differentiation. Questions and problems related to the use of human embryonic and adult stem cells in tissue regeneration and transplantation are discussed.
Collapse
Affiliation(s)
- Jaroslaw Czyz
- In Vitro Differentiation Group, IPK Gatersleben, Corrensstrasse 3, D-06466 Gatersleben, Germany
| | | | | | | | | | | |
Collapse
|
1040
|
Couldrey C, Bradley HL, Bunting KD. A STAT5 modifier locus on murine chromosome 7 modulates engraftment of hematopoietic stem cells during steady-state hematopoiesis. Blood 2004; 105:1476-83. [PMID: 15498858 DOI: 10.1182/blood-2004-06-2302] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Homologous disruption of expression of signal transducer and activator of transcription 5a (STAT5a) and STAT5b (STAT5ab(-/-)) in mice results in hematopoietic stem cells (HSCs) that can engraft irradiated hosts alone but are noncompetitive against wild-type HSCs. To explore mechanisms for this phenotype, we crossed the STAT5 mutations onto an HW80 background congenic to the original C57BL/6 that differs in a small chromosome 7 genomic locus. We previously demonstrated that C57BL/6 or HW80 background STAT5ab(-/-) bone marrow (BM) cells showed equal repopulating function either competitively or noncompetitively in irradiated hosts. However, one intraperitoneal injection of wild-type green fluorescent protein (GFP) transgenic BM cells into unconditioned newborn STAT5ab(-/-) recipients of either background was sufficient for high-level donor engraftment. Furthermore, haploinsufficiency of STAT5 (STAT5ab(+/-)) allowed improved engraftment over wild-type recipients, indicating a dose-dependent requirement for STAT5 activation. In reciprocal experiments, STAT5ab(-/-) BM was transplanted into nonirradiated W/W(v) hosts. In these mice, C57BL/6 STAT5ab(-/-) BM cells were 10-fold more defective in long-term engraftment than control wild-type BM cells and HW80 STAT5ab(-/-) BM cells were 5- to 10-fold more defective than C57BL/6 STAT5ab(-/-) BM cells. Therefore, we conclude that STAT5 plays a critical role during steady-state HSC engraftment and a chromosome 7 modifier locus regulates this activity.
Collapse
Affiliation(s)
- Christine Couldrey
- Hematopoiesis Department, Jerome H. Holland Laboratory for the Biomedical Sciences, American Red Cross, Rockville, MD, USA
| | | | | |
Collapse
|
1041
|
Golan-Mashiach M, Dazard JE, Gerecht-Nir S, Amariglio N, Fisher T, Jacob-Hirsch J, Bielorai B, Osenberg S, Barad O, Getz G, Toren A, Rechavi G, Itskovitz-Eldor J, Domany E, Givol D. Design principle of gene expression used by human stem cells: implication for pluripotency. FASEB J 2004; 19:147-9. [PMID: 15498892 DOI: 10.1096/fj.04-2417fje] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Human embryonic stem cells (ESC) are undifferentiated and are endowed with the capacities of self-renewal and pluripotential differentiation. Adult stem cells renew their own tissue, but whether they can transdifferentiate to other tissues is still controversial. To understand the genetic program that underlies the pluripotency of stem cells, we compared the transcription profile of ESC with that of progenitor/stem cells of human hematopoietic and keratinocytic origins, along with their mature cells to be viewed as snapshots along tissue differentiation. ESC gene profiles show higher complexity with significantly more highly expressed genes than adult cells. We hypothesize that ESC use a strategy of expressing genes that represent various differentiation pathways and selection of only a few for continuous expression upon differentiation to a particular target. Such a strategy may be necessary for the pluripotency of ESC. The progenitors of either hematopoietic or keratinocytic cells also follow the same design principle. Using advanced clustering, we show that many of the ESC expressed genes are turned off in the progenitors/stem cells followed by a further down-regulation in adult tissues. Concomitantly, genes specific to the target tissue are up-regulated toward mature cells of skin or blood.
Collapse
Affiliation(s)
- Michal Golan-Mashiach
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1042
|
Blanpain C, Lowry WE, Geoghegan A, Polak L, Fuchs E. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell 2004; 118:635-48. [PMID: 15339667 DOI: 10.1016/j.cell.2004.08.012] [Citation(s) in RCA: 1049] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2004] [Revised: 07/23/2004] [Accepted: 07/23/2004] [Indexed: 12/16/2022]
Abstract
In adult skin, each hair follicle contains a reservoir of stem cells (the bulge), which can be mobilized to regenerate the new follicle with each hair cycle and to reepithelialize epidermis during wound repair. Here we report new methods that permit their clonal analyses and engraftment and demonstrate the two defining features of stem cells, namely self-renewal and multipotency. We also show that, within the bulge, there are two distinct populations, one of which maintains basal lamina contact and temporally precedes the other, which is suprabasal and arises only after the start of the first postnatal hair cycle. This spatial distinction endows them with discrete transcriptional programs, but surprisingly, both populations are growth inhibited in the niche but can self-renew in vitro and make epidermis and hair when grafted. These findings suggest that the niche microenvironment imposes intrinsic "stemness" features without restricting the establishment of epithelial polarity and changes in gene expression.
Collapse
Affiliation(s)
- Cedric Blanpain
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
1043
|
Abstract
Proteins from the Polycomb group (PcG) are epigenetic chromatin modifiers involved in cancer development and also in the maintenance of embryonic and adult stem cells. The therapeutic potential of stem cells and the growing conviction that tumors contain stem cells highlights the importance of understanding the extrinsic and intrinsic circuitry controlling stem cell fate and their connections to cancer.
Collapse
Affiliation(s)
- Merel E Valk-Lingbeek
- The Netherlands Cancer Institute, Department of Molecular Genetics, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | |
Collapse
|
1044
|
Kim JK, Choi BH, Park HC, Park SR, Kim YS, Yoon SH, Park HS, Kim EY, Ha Y. Effects of GM-CSF on the neural progenitor cells. Neuroreport 2004; 15:2161-5. [PMID: 15371725 DOI: 10.1097/00001756-200410050-00003] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Granulocyte macrophage colony stimulating factor (GM-CSF) is a potent hematopoietic cytokine, which stimulates stem cell proliferation in the bone marrow. We now report that GM-CSF receptors expressed on neural progenitor cells and can mediate a biological response in cells to treat with GM-CSF treated neural progenitor cells exhibited a proliferative response and a marked decrease in terminal differentiation to mature neuron or astrocytes. GM-CSF treatment also suppressed neural progenitor cell apoptosis. These findings suggest that GM-CSF can stimulate the proliferation and inhibit the apoptosis of neural progenitor cells to expand the progenitor population, and that GM-CSF has a potential role in neural development or repair.
Collapse
Affiliation(s)
- Jin Kyun Kim
- Department of Neurosurgery, Inha University College of Medicine, 7-206, shinheung-dong 3ga, Chung-gu, Inchon, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
1045
|
Baker SA, Baker KA, Hagg T. Dopaminergic nigrostriatal projections regulate neural precursor proliferation in the adult mouse subventricular zone. Eur J Neurosci 2004; 20:575-9. [PMID: 15233767 DOI: 10.1111/j.1460-9568.2004.03486.x] [Citation(s) in RCA: 192] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An understanding of the regulators of neurogenesis in the normal and diseased brain is necessary in order to recruit endogenously produced neural precursors for cell replacement in neurodegenerative disorders such as Parkinson's disease. The location of dopaminergic projections from the midbrain to the neostriatum and nucleus accumbens overlaps with the most active region of neurogenesis in the adult brain, the subventricular zone of the anterior lateral ventricle. This suggests that dopamine may contribute to regulation of the subventricular niche of adult neurogenesis. Here, we show in adult mice that destruction of the dopaminergic neurons in the substantia nigra and ventral tegmental area in a 6-hydroxydopamine model of Parkinson's disease reduced the number of proliferating neural precursors in the subventricular zone of the anterior lateral ventricle by approximately 40%. The effect on neural precursor proliferation correlated with the extent of dopaminergic denervation in the neighboring neostriatum. This identifies dopamine as one of the few known endogenous regulators of adult neurogenesis with implications for the potential use of endogenous neural precursors in cell replacement strategies for Parkinson's disease.
Collapse
Affiliation(s)
- Sarah A Baker
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 S Floyd Street, MDR Room 616, Louisville, KY 40292, USA
| | | | | |
Collapse
|
1046
|
Venezia TA, Merchant AA, Ramos CA, Whitehouse NL, Young AS, Shaw CA, Goodell MA. Molecular signatures of proliferation and quiescence in hematopoietic stem cells. PLoS Biol 2004; 2:e301. [PMID: 15459755 PMCID: PMC520599 DOI: 10.1371/journal.pbio.0020301] [Citation(s) in RCA: 262] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2004] [Accepted: 07/13/2004] [Indexed: 12/05/2022] Open
Abstract
Stem cells resident in adult tissues are principally quiescent, yet harbor enormous capacity for proliferation to achieve self renewal and to replenish their tissue constituents. Although a single hematopoietic stem cell (HSC) can generate sufficient primitive progeny to repopulate many recipients, little is known about the molecular mechanisms that maintain their potency or regulate their self renewal. Here we have examined the gene expression changes that occur over a time course when HSCs are induced to proliferate and return to quiescence in vivo. These data were compared to data representing differences between naturally proliferating fetal HSCs and their quiescent adult counterparts. Bioinformatic strategies were used to group time-ordered gene expression profiles generated from microarrays into signatures of quiescent and dividing stem cells. A novel method for calculating statistically significant enrichments in Gene Ontology groupings for our gene lists revealed elemental subgroups within the signatures that underlie HSC behavior, and allowed us to build a molecular model of the HSC activation cycle. Initially, quiescent HSCs evince a state of readiness. The proliferative signal induces a preparative state, which is followed by active proliferation divisible into early and late phases. Re-induction of quiescence involves changes in migratory molecule expression, prior to reestablishment of homeostasis. We also identified two genes that increase in both gene and protein expression during activation, and potentially represent new markers for proliferating stem cells. These data will be of use in attempts to recapitulate the HSC self renewal process for therapeutic expansion of stem cells, and our model may correlate with acquisition of self renewal characteristics by cancer stem cells. This comprehensive study of gene expression in hematopoietic stem cells reveals some key cellular changes that occur when the stem cells transition from quiescence to proliferation and back again
Collapse
Affiliation(s)
- Teresa A Venezia
- 1Cell and Molecular Biology Program, Baylor College of MedicineHouston, TexasUnited States of America
- 2Center for Cell and Gene Therapy, Baylor College of MedicineHouston, TexasUnited States of America
| | - Akil A Merchant
- 2Center for Cell and Gene Therapy, Baylor College of MedicineHouston, TexasUnited States of America
- 3Department of Medicine, Baylor College of MedicineHouston, TexasUnited States of America
| | - Carlos A Ramos
- 2Center for Cell and Gene Therapy, Baylor College of MedicineHouston, TexasUnited States of America
- 3Department of Medicine, Baylor College of MedicineHouston, TexasUnited States of America
| | - Nathan L Whitehouse
- 4Department of Human and Molecular Genetics, Baylor College of MedicineHouston, TexasUnited States of America
| | - Andrew S Young
- 4Department of Human and Molecular Genetics, Baylor College of MedicineHouston, TexasUnited States of America
| | - Chad A Shaw
- 4Department of Human and Molecular Genetics, Baylor College of MedicineHouston, TexasUnited States of America
| | - Margaret A Goodell
- 1Cell and Molecular Biology Program, Baylor College of MedicineHouston, TexasUnited States of America
- 2Center for Cell and Gene Therapy, Baylor College of MedicineHouston, TexasUnited States of America
- 4Department of Human and Molecular Genetics, Baylor College of MedicineHouston, TexasUnited States of America
- 5Department of Pediatrics, Baylor College of MedicineHouston, TexasUnited States of America
| |
Collapse
|
1047
|
Cai J, Weiss ML, Rao MS. In search of "stemness". Exp Hematol 2004; 32:585-98. [PMID: 15246154 PMCID: PMC3279197 DOI: 10.1016/j.exphem.2004.03.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2004] [Revised: 03/22/2004] [Accepted: 03/25/2004] [Indexed: 01/02/2023]
Abstract
Stem cells have been identified and characterized in a variety of tissues. In this review we examine possible shared properties of stem cells. We suggest that irrespective of their lineal origin, stem cells have to respond in similar ways to regulate self-renewal and differentiation and it is likely that cell-cycle control, asymmetry/differentiation controls, cellular protective and DNA repair mechanisms, and associated apoptosis/senescence signaling pathways all might be expected to be more highly regulated in stem cells, likely by similar mechanisms. We review the literature to suggest a set of candidate stemness genes that may serve as universal stem cell markers. While we predict many similarities, we also predict that differences will exist between stem cell populations and that when transdifferentiation is considered genes expected to be both similar and different need to be examined.
Collapse
Affiliation(s)
- Jingli Cai
- Gerontology Research Center, Stem Cell Biology Unit/Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | |
Collapse
|
1048
|
Kim JY, Sawada A, Tokimasa S, Endo H, Ozono K, Hara J, Takihara Y. Defective long-term repopulating ability in hematopoietic stem cells lacking the Polycomb-group gene rae28. Eur J Haematol 2004; 73:75-84. [PMID: 15245505 DOI: 10.1111/j.1600-0609.2004.00268.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The rae28 gene (rae28) is a member of a Polycomb-group (PcG) complex 1, which is known to help maintain transcription states once these have been initiated, by generating heritable higher-order chromatin structures. In this study, we examined the capacity of rae28-deficient (rae28-/-) hematopoietic stem cells (HSCs) to generate long-term marrow reconstitution. rae28-/- fetal liver cells containing 20 competitive repopulation units (CRUs) were able to support the survival of lethally irradiated congenic mice for as long as 6 months. The marrow reconstituted with the rae28-/- cells, however, could not increase HSCs efficiently. This was evidenced by its inability to reconstitute marrow in serial transplantation experiments, as well as by the reduction in HSC-enriched Lin- c-kit+ Sca-1high+ subpopulation in the bone marrow cells. Moreover, the reconstituted marrow produced less than half of the peripheral blood cells in each of the lineages examined. We also monitored the mean stem cell activity (MAS). MAS of rae28-/- CRUs was progressively reduced after transplantation, and after 12 months it was reduced to one-tenth of that of the wild-type. These in vivo results clearly indicate that rae28 is indispensable for the long-term repopulating ability of HSCs. We further referred to the plausible mechanisms underlying defective long-term repopulating ability of rae28-deficient HSCs and argued for its involvement in maintenance of cell proliferation capability as well as that in self-renewal ability.
Collapse
Affiliation(s)
- Ji Yoo Kim
- Department of Developmental Biology and Medicine, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
1049
|
Qiu J, Takagi Y, Harada J, Rodrigues N, Moskowitz MA, Scadden DT, Cheng T. Regenerative response in ischemic brain restricted by p21cip1/waf1. ACTA ACUST UNITED AC 2004; 199:937-45. [PMID: 15067031 PMCID: PMC2211886 DOI: 10.1084/jem.20031385] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neural precursor cells from adults have exceptional proliferative and differentiative capability in vitro yet respond minimally to in vivo brain injury due to constraining mechanisms that are poorly defined. We assessed whether cell cycle inhibitors that restrict stem cell populations in other tissues may participate in limiting neural stem cell reactivity in vivo. The cyclin-dependent kinase inhibitor, p21cip1/waf1 (p21), maintains hematopoietic stem cell quiescence, and we evaluated its role in the regenerative response of neural tissue after ischemic injury using the mice deficient in p21. Although steady-state conditions revealed no increase in primitive cell proliferation in p21-null mice, a significantly larger fraction of quiescent neural precursors was activated in the hippocampus and subventricular zone after brain ischemia. The hippocampal precursors migrated and differentiated into a higher number of neurons after injury. Therefore, p21 is an intrinsic suppressor to neural regeneration after brain injury and may serve as a common molecular regulator restricting proliferation among stem cell pools from distinct tissue types.
Collapse
Affiliation(s)
- Jianhua Qiu
- Neuroscience Center, Harvard Medical School, Boston, MA 02129, USA
| | | | | | | | | | | | | |
Collapse
|
1050
|
Tagliafico E, Brunelli S, Bergamaschi A, De Angelis L, Scardigli R, Galli D, Battini R, Bianco P, Ferrari S, Cossu G, Ferrari S. TGFβ/BMP activate the smooth muscle/bone differentiation programs in mesoangioblasts. J Cell Sci 2004; 117:4377-88. [PMID: 15331661 DOI: 10.1242/jcs.01291] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Mesoangioblasts are vessel-derived stem cells that can be induced to differentiate into different cell types of the mesoderm such as muscle and bone. The gene expression profile of four clonal derived lines of mesoangioblasts was determined by DNA micro-array analysis: it was similar in the four lines but different from 10T1/2 embryonic fibroblasts, used as comparison. Many known genes expressed by mesoangioblasts belong to response pathways to developmental signalling molecules, such as Wnt or TGFβ/BMP. Interestingly, mesoangioblasts express receptors of the TGFβ/BMP family and several Smads and, accordingly, differentiate very efficiently into smooth muscle cells in response to TGFβ and into osteoblasts in response to BMP. In addition, insulin signalling promotes adipogenic differentiation, possibly through the activation of IGF-R. Several Wnts and Frizzled, Dishevelled and Tcfs are expressed, suggesting the existence of an autocrine loop for proliferation and indeed, forced expression of Frzb-1 inhibits cell division. Mesoangioblasts also express many neuro-ectodermal genes and yet undergo only abortive neurogenesis, even after forced expression of neurogenin 1 or 2, MASH or NeuroD. Finally, mesoangioblasts express several pro-inflammatory genes, cytokines and cytokine receptors, which may explain their ability to be recruited by tissue inflammation. Our data define a unique phenotype for mesoangioblasts, explain several of their biological features and set the basis for future functional studies on the role of these cells in tissue histogenesis and repair.
Collapse
Affiliation(s)
- Enrico Tagliafico
- Dipartimento di Scienze Biomediche, Università di Modena e Reggio Emilia, Via G. Campi 287, 41100 Modena, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|