1101
|
|
1102
|
Li Q, Ford MC, Lavik EB, Madri JA. Modeling the neurovascular niche: VEGF- and BDNF-mediated cross-talk between neural stem cells and endothelial cells: An in vitro study. J Neurosci Res 2006; 84:1656-68. [PMID: 17061253 DOI: 10.1002/jnr.21087] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neural stem cells (NSCs) exist in vascularized niches. Although there has been ample evidence supporting a role for endothelial cell-derived soluble factors as modulators of neural stem cell self-renewal and neuronal differentiation there is a paucity of data reported on neural stem cell modulation of endothelial cell behavior. We show that co-culture of NSCs with brain-derived endothelial cells (BECs) either in direct contact or separated by a porous membrane elicited robust vascular tube formation and maintenance, mediated by induction of vascular vascular endothelial growth factor (VEGF) and brain-derived neurotrophic factor (BDNF) and activation of vascular VEGFR2 and TrkB by NSC NO. Nitric oxide (NO) scavengers and sequestration of VEGF and BDNF blunted this induction of tube formation, whereas addition of exogenous NO donor, rBDNF and rVEGF rescued the induction of tube formation. Further, rBDNF enhanced NSC eNOS activation and NO generation, suggesting an inducible positive feed-back signaling loop between NSCs and BECs, providing for homeostasis and responsiveness of the resident NSCs and BECs comprising the neurovascular niche. These findings show the importance of reciprocal modulation of NSCs and BECs in induction and maintenance of the neurovascular niche and underscores their dynamic interactions.
Collapse
Affiliation(s)
- Qi Li
- Department of Pathology, Yale University, School of Medicine, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
1103
|
Zhang RL, Zhang ZG, Chopp M. Neurogenesis in the adult ischemic brain: generation, migration, survival, and restorative therapy. Neuroscientist 2005; 11:408-16. [PMID: 16151043 DOI: 10.1177/1073858405278865] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This article reviews current data on the induction of neurogenesis after stroke in the adult brain. The discussion of neurogenesis is divided into production, migration, and survival of these newly formed cells. For production, the subpopulations and the types of cell division are presented. Discussion of cell migration entails presenting data on both the pathways as well as the molecular targeting of newly formed neural progenitor cells to sites of injury. The role of the vascular and the astrocytic microenvironment in promoting the survival and integration of progenitor cells is also presented. Cell-based and pharmacological therapies designed to restore neurological function that promote neurogenesis are described. These therapies also induce angiogenesis and astrocytic changes that brain tissue, which prime the ischemic brain to foster the survival of the newly formed progenitor cells. Signaling pathways that regulate neurogenesis and angiogenesis are also addressed. This review summarizes recent data on neurogenesis and provides insight into the potential for restorative treatments of stroke.
Collapse
Affiliation(s)
- Rui Lan Zhang
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, Michigan 48202, USA
| | | | | |
Collapse
|
1104
|
Abstract
The theory of the "stem cell niche" was originally proposed for the hematopoietic system, and the existence of the niche as an actual entity was proved in the Drosophila germ cell system. Historically, mammalian spermatogenesis has been studied extensively as a prime example of a stem cell system, and studies have established a stem-progenitor hierarchical order of spermatogonia. In the niche on the basal lamina of seminiferous tubules, spermatogonial stem cells (SSCs) are secluded from the outside world and divide constantly to self-renew and differentiate. During the last 10 years, the development and exploitation of the germ cell transplantation method has expanded our understanding of the nature of SSCs and their niches. The ability to maintain and expand SSCs in vitro, which recently became possible, has further reinforced this research area as a mecca of stem cell biology. Nonetheless, the mammalian germ stem cell and its niche remain to be defined more strictly and precisely. We are still on a journey in search of the real stem cell and its true niche.
Collapse
Affiliation(s)
- Takehiko Ogawa
- Department of Urology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan.
| | | | | |
Collapse
|
1105
|
Guillemot F. Cellular and molecular control of neurogenesis in the mammalian telencephalon. Curr Opin Cell Biol 2005; 17:639-47. [PMID: 16226447 DOI: 10.1016/j.ceb.2005.09.006] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Accepted: 09/29/2005] [Indexed: 10/25/2022]
Abstract
The mammalian telencephalon exhibits an amazing diversity of neuronal types. The generation of this diversity relies on multiple developmental strategies, including the regional patterning of progenitors, their temporal specification, and the generation of intermediate progenitor populations. Progress has recently been made in characterizing some of the mechanisms involved. In particular, intermediate progenitors have been shown to play important roles in the generation of neurons in the cerebral cortex, and the properties and lineage relationships between radial glial cells and these intermediate progenitors have recently been examined by elegant time-lapse microscopic studies. Multiple pathways control the progression of neural lineages from multipotent stem cells to intermediate progenitors, postmitotic precursors and finally mature neurons. The regulation of two essential steps, neuronal commitment and specification of subtype identities, is increasingly well understood. These two steps are clearly distinct but co-ordinately regulated by common transcription factors such as neurogenins and Pax6. As our knowledge of the mechanisms of subtype specification of telencephalic neurons progresses, it will become possible to direct stem cells into generating particular telencephalic neuronal populations, opening the way to efficient neuronal replacement therapies.
Collapse
Affiliation(s)
- François Guillemot
- Division of Molecular Neurobiology, National Institute for Medical Research, Mill Hill, London NW7 1AA, UK.
| |
Collapse
|
1106
|
Ma DK, Ming GL, Song H. Glial influences on neural stem cell development: cellular niches for adult neurogenesis. Curr Opin Neurobiol 2005; 15:514-20. [PMID: 16144763 DOI: 10.1016/j.conb.2005.08.003] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2005] [Accepted: 08/24/2005] [Indexed: 12/20/2022]
Abstract
Neural stem cells continually generate new neurons in very limited regions of the adult mammalian central nervous system. In the neurogenic regions there are unique and highly specialized microenvironments (niches) that tightly regulate the neuronal development of adult neural stem cells. Emerging evidence suggests that glia, particularly astrocytes, have key roles in controlling multiple steps of adult neurogenesis within the niches, from proliferation and fate specification of neural progenitors to migration and integration of the neuronal progeny into pre-existing neuronal circuits in the adult brain. Identification of specific niche signals that regulate these sequential steps during adult neurogenesis might lead to strategies to induce functional neurogenesis in other brain regions after injury or degenerative neurological diseases.
Collapse
Affiliation(s)
- Dengke K Ma
- Institute for Cell Engineering, Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
1107
|
Das AV, Edakkot S, Thoreson WB, James J, Bhattacharya S, Ahmad I. Membrane properties of retinal stem cells/progenitors. Prog Retin Eye Res 2005; 24:663-81. [PMID: 15939659 DOI: 10.1016/j.preteyeres.2005.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The membrane properties of cells help integrate extrinsic information relayed through growth factors, chemokines, extracellular matrix, gap junctions and neurotransmitters towards modulating cell-intrinsic properties, which in turn determine whether cells remain quiescent, proliferate, differentiate, establish contact with other cells or remove themselves by activating programmed cell death. This review highlights some of the membrane properties of early and late retinal stem cells/progenitors, which are likely to be helpful in the identification and enrichment of these cells and in understanding mechanisms underlying their maintenance and differentiation. Understanding of membrane properties of retinal stem cells/progenitors is essential for the successful formulation of approaches to treat retinal degeneration and diseases by cell therapy.
Collapse
Affiliation(s)
- Ani V Das
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198-7691, USA
| | | | | | | | | | | |
Collapse
|
1108
|
Kopp HG, Avecilla ST, Hooper AT, Rafii S. The bone marrow vascular niche: home of HSC differentiation and mobilization. Physiology (Bethesda) 2005; 20:349-56. [PMID: 16174874 DOI: 10.1152/physiol.00025.2005] [Citation(s) in RCA: 332] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The bone marrow vascular niche consists of a network of thin-walled and fenestrated sinusoidal vessels whose integrity is maintained and supported by surrounding hematopoietic cells. However, this dependence is highly reciprocal in that the bone marrow vasculature provides not only a conduit for mature hematopoietic cells to the peripheral circulation but also a site where hematopoietic progenitors, especially megakaryocytes, differentiate and set the stage for full reconstitution of hematopoiesis.
Collapse
Affiliation(s)
- Hans-Georg Kopp
- Weill Medical College of Cornell University, New York, New York, USA
| | | | | | | |
Collapse
|
1109
|
Meng H, Zhang Z, Zhang R, Liu X, Wang L, Robin AM, Chopp M. Biphasic effects of exogenous VEGF on VEGF expression of adult neural progenitors. Neurosci Lett 2005; 393:97-101. [PMID: 16269210 DOI: 10.1016/j.neulet.2005.09.044] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Revised: 08/30/2005] [Accepted: 09/19/2005] [Indexed: 01/17/2023]
Abstract
Vascular endothelial growth factor (VEGF) regulates neurogenesis. The present study investigated the direct effect of VEGF on the enhancement of proliferation and differentiation of the adult mouse subventricular zone (SVZ) neural progenitors in vitro. A high dose (500 ng/ml) of VEGF significantly downregulated endogenous VEGF receptors 1 and 2, which was associated with significantly reduced neural progenitor cell proliferation and enhancement of neuronal differentiation. A low dose (50 ng/ml) of VEGF significantly upregulated endogenous VEGF receptors 1 and 2 but did not increase proliferation and differentiation. These data suggest that exogenous VEGF has a biphasic effect on the expression of endogenous VEGF receptors, and the high dose of VEGF enhances adult neural progenitor cell differentiation into neurons.
Collapse
Affiliation(s)
- He Meng
- Department of Neurology, Henry Ford Health Sciences Center, Henry Ford Hospital, Neurology Division of Research, Detroit, MI 48202, USA
| | | | | | | | | | | | | |
Collapse
|
1110
|
Nyfeler Y, Kirch RD, Mantei N, Leone DP, Radtke F, Suter U, Taylor V. Jagged1 signals in the postnatal subventricular zone are required for neural stem cell self-renewal. EMBO J 2005; 24:3504-15. [PMID: 16163386 PMCID: PMC1276174 DOI: 10.1038/sj.emboj.7600816] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2005] [Accepted: 08/08/2005] [Indexed: 01/06/2023] Open
Abstract
Neural stem cells (NSCs) in the postnatal mammalian brain self-renew and are a source of neurons and glia. To date, little is known about the molecular and cellular mechanisms regulating the maintenance and differentiation of these multipotent progenitors. We show that Jagged1 is required by mitotic cells in the subventricular zone (SVZ) and stimulates self-renewal of multipotent epidermal growth factor-dependent NSCs. Jagged1-expressing cells line the adult SVZ and are juxtaposed to Notch1-expressing cells, some of which are putative NSCs. In vitro, endogenous Jagged1 acts through Notch1 to promote NSC maintenance and multipotency. In vivo, reducing Jagged1/Notch1 signaling decreases the number of proliferating cells in the SVZ. In addition, soluble Jagged1 promotes self-renewal and neurogenic potential of multipotent neural progenitors in vitro. Our findings suggest a central role for Jagged1 in the NSC niche in the SVZ for maintaining a population of NSCs in the postnatal brain.
Collapse
Affiliation(s)
- Yves Nyfeler
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology, Freiburg, Germany
| | - Robert D Kirch
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology, Freiburg, Germany
| | - Ned Mantei
- Institute of Cell Biology, Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Dino P Leone
- Institute of Cell Biology, Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Freddy Radtke
- Ludwig Institute, Lausanne Branch, University of Lausanne, Epalinges, Switzerland
| | - Ueli Suter
- Institute of Cell Biology, Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Verdon Taylor
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology, Freiburg, Germany
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology, Stubeweg 51, 79011 Freiburg, Germany. Tel.: +49 761 5108 487; Fax: +49 761 5108 474; E-mail:
| |
Collapse
|
1111
|
Hagg T. Molecular regulation of adult CNS neurogenesis: an integrated view. Trends Neurosci 2005; 28:589-95. [PMID: 16153715 DOI: 10.1016/j.tins.2005.08.009] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2005] [Revised: 07/21/2005] [Accepted: 08/30/2005] [Indexed: 01/17/2023]
Abstract
Neural stem cells in the adult subventricular zone and dentate gyrus might be utilized to replace lost neurons in neurological disorders. The development of treatments would be facilitated by identifying the mechanisms that contribute to functional neurogenesis in adult animals. This review focuses on the emerging view that localized and overlapping pathways of growth factors, metalloproteases, neurotransmitters and hormones regulate different aspects of neurogenesis within the neurogenic niches. Neuroblast migration is precisely regulated by cooperation between several repellants, attractants and guidance molecules that are located within specific CNS regions. Further elucidation of crucial molecular regulators and integration of their signaling cascades should lead to more rational and effective approaches to harness the exciting phenomenon of adult CNS neurogenesis.
Collapse
Affiliation(s)
- Theo Hagg
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
1112
|
Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 2005; 121:1109-21. [PMID: 15989959 DOI: 10.1016/j.cell.2005.05.026] [Citation(s) in RCA: 2363] [Impact Index Per Article: 118.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Revised: 04/29/2005] [Accepted: 05/13/2005] [Indexed: 12/21/2022]
Abstract
To improve our ability to identify hematopoietic stem cells (HSCs) and their localization in vivo, we compared the gene expression profiles of highly purified HSCs and non-self-renewing multipotent hematopoietic progenitors (MPPs). Cell surface receptors of the SLAM family, including CD150, CD244, and CD48, were differentially expressed among functionally distinct progenitors. HSCs were highly purified as CD150(+)CD244(-)CD48(-) cells while MPPs were CD244(+)CD150(-)CD48(-) and most restricted progenitors were CD48(+)CD244(+)CD150(-). The primitiveness of hematopoietic progenitors could thus be predicted based on the combination of SLAM family members they expressed. This is the first family of receptors whose combinatorial expression precisely distinguishes stem and progenitor cells. The ability to purify HSCs based on a simple combination of SLAM receptors allowed us to identify HSCs in tissue sections. Many HSCs were associated with sinusoidal endothelium in spleen and bone marrow, though some HSCs were associated with endosteum. HSCs thus occupy multiple niches, including sinusoidal endothelium in diverse tissues.
Collapse
Affiliation(s)
- Mark J Kiel
- Howard Hughes Medical Institute and Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | |
Collapse
|
1113
|
Conti L, Pollard SM, Gorba T, Reitano E, Toselli M, Biella G, Sun Y, Sanzone S, Ying QL, Cattaneo E, Smith A. Niche-independent symmetrical self-renewal of a mammalian tissue stem cell. PLoS Biol 2005; 3:e283. [PMID: 16086633 PMCID: PMC1184591 DOI: 10.1371/journal.pbio.0030283] [Citation(s) in RCA: 687] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Accepted: 06/14/2005] [Indexed: 12/11/2022] Open
Abstract
Pluripotent mouse embryonic stem (ES) cells multiply in simple monoculture by symmetrical divisions. In vivo, however, stem cells are generally thought to depend on specialised cellular microenvironments and to undergo predominantly asymmetric divisions. Ex vivo expansion of pure populations of tissue stem cells has proven elusive. Neural progenitor cells are propagated in combination with differentiating progeny in floating clusters called neurospheres. The proportion of stem cells in neurospheres is low, however, and they cannot be directly observed or interrogated. Here we demonstrate that the complex neurosphere environment is dispensable for stem cell maintenance, and that the combination of fibroblast growth factor 2 (FGF-2) and epidermal growth factor (EGF) is sufficient for derivation and continuous expansion by symmetrical division of pure cultures of neural stem (NS) cells. NS cells were derived first from mouse ES cells. Neural lineage induction was followed by growth factor addition in basal culture media. In the presence of only EGF and FGF-2, resulting NS cells proliferate continuously, are diploid, and clonogenic. After prolonged expansion, they remain able to differentiate efficiently into neurons and astrocytes in vitro and upon transplantation into the adult brain. Colonies generated from single NS cells all produce neurons upon growth factor withdrawal. NS cells uniformly express morphological, cell biological, and molecular features of radial glia, developmental precursors of neurons and glia. Consistent with this profile, adherent NS cell lines can readily be established from foetal mouse brain. Similar NS cells can be generated from human ES cells and human foetal brain. The extrinsic factors EGF plus FGF-2 are sufficient to sustain pure symmetrical self-renewing divisions of NS cells. The resultant cultures constitute the first known example of tissue-specific stem cells that can be propagated without accompanying differentiation. These homogenous cultures will enable delineation of molecular mechanisms that define a tissue-specific stem cell and allow direct comparison with pluripotent ES cells.
Collapse
Affiliation(s)
- Luciano Conti
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
- 2Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Steven M Pollard
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Thorsten Gorba
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Erika Reitano
- 2Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Mauro Toselli
- 3Institute of Physiological and Pharmacological Sciences, University of Pavia, Pavia, Italy
| | - Gerardo Biella
- 3Institute of Physiological and Pharmacological Sciences, University of Pavia, Pavia, Italy
| | - Yirui Sun
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Sveva Sanzone
- 2Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Qi-Long Ying
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Elena Cattaneo
- 2Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Austin Smith
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
1114
|
Eckfeldt CE, Mendenhall EM, Verfaillie CM. The molecular repertoire of the 'almighty' stem cell. Nat Rev Mol Cell Biol 2005; 6:726-37. [PMID: 16103873 DOI: 10.1038/nrm1713] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Stem cells share the defining characteristics of self-renewal, which maintains or expands the stem-cell pool, and multi-lineage differentiation, which generates and regenerates tissues. Stem-cell self-renewal and differentiation are influenced by the convergence of intrinsic cellular signals and extrinsic microenvironmental cues from the surrounding stem-cell niche, but the specific signals involved are poorly understood. Recently, several studies have sought to identify the genetic mechanisms that underlie the stem-cell phenotype. Such a molecular road map of stem-cell function should lead to an understanding of the true potential of stem cells.
Collapse
Affiliation(s)
- Craig E Eckfeldt
- Department of Medicine and Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
1115
|
Abstract
Angiogenesis has critical roles in normal vascular development and in important pathologies including cancer, wound healing and inflammation. This brief article will review the angiogenic response induced by the vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) family of proteins and particularly VEGF-A, thought to be the single most important angiogenic factor. It will also review the steps and mechanisms by which VEGF-A induces the formation of new blood vessels and will provide an initial classification of the abnormal blood vessels that form in pathological angiogenesis. Finally, it will touch on the exciting relationships that are emerging between angiogenesis and the hemostatic and nervous systems.
Collapse
Affiliation(s)
- H F Dvorak
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, USA.
| |
Collapse
|
1116
|
Xi R, Kirilly D, Xie T. Molecular mechanisms controlling germline and somatic stem cells: similarities and differences. Curr Opin Genet Dev 2005; 15:381-7. [PMID: 15950455 DOI: 10.1016/j.gde.2005.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2005] [Accepted: 05/23/2005] [Indexed: 12/25/2022]
Abstract
Germline and somatic stem cells are distinct types of stem cells that are dedicated to reproduction and somatic tissue homeostasis, respectively. Extensive studies on these two stem cell types in different organisms over the past few years have revealed some commonalities in the mechanisms controlling their self-renewal and differentiation. Furthermore, germline or somatic cells in various organisms and sexes also exhibit their own unique ways of regulating stem cell function. By understanding these similarities and differences we might gain a better insight into how stem cells are regulated in general and how germline and somatic stem cell types are regulated differently.
Collapse
Affiliation(s)
- Rongwen Xi
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | | | | |
Collapse
|
1117
|
Getchell TV, Liu H, Vaishnav RA, Kwong K, Stromberg AJ, Getchell ML. Temporal profiling of gene expression during neurogenesis and remodeling in the olfactory epithelium at short intervals after target ablation. J Neurosci Res 2005; 80:309-29. [PMID: 15795924 DOI: 10.1002/jnr.20411] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neurogenesis in the olfactory epithelium (OE) is induced by olfactory bulbectomy (OBX), which effectively axotomizes olfactory sensory neurons (OSNs) and removes their synaptic targets, resulting in apoptosis. We used Affymetrix high-density oligonucleotide arrays to investigate changes in gene expression during initiation of signaling in pathways that regulate apoptosis and neurogenesis in the murine OE at 2, 8, 16, and 48 hr after bilateral OBX compared to that in sham-operated controls. We focused on regulation of a defined set of genes associated with apoptosis, stem/progenitor cell regulation, and cell cycle progression because of the activation of these processes in OE degeneration and remodeling after OBX. After data scrubbing and categorical analysis, one-way analysis of variance identified 72 genes (4.9% of the present known genes) as being regulated significantly (P < 0.05) at one or more points; 50 were defined as regulated differentially with the false discovery rate at 10%. Significant changes in gene expression occurred in all categories as early as 2 hr post-OBX, with the greatest number of differentially regulated genes at 16 and 48 hr. Hierarchical cluster analysis and correlation coefficients were used to identify similarities in patterns of gene expression changes within and across categories. Validation was carried out with SuperArray macroarrays and real-time RT-PCR. Our results confirmed the participation of many genes in known signaling pathways and identified changes in the expression of 42 genes not identified previously as participating in apoptosis and neurogenesis in the OE. Additionally, our analyses indicated the early involvement of genes regulating cytoskeletal reorganization and angiogenesis in the response to OBX. These studies are an important first step in defining early time-dependent changes in gene expression after target ablation that lead to neurogenesis in the olfactory sensory epithelium.
Collapse
Affiliation(s)
- Thomas V Getchell
- Department of Physiology, University of Kentucky, Lexington, 40536-0230, USA.
| | | | | | | | | | | |
Collapse
|
1118
|
Hauck AL, Swanson KS, Kenis PJA, Leckband DE, Gaskins HR, Schook LB. Twists and turns in the development and maintenance of the mammalian small intestine epithelium. ACTA ACUST UNITED AC 2005; 75:58-71. [PMID: 15838920 DOI: 10.1002/bdrc.20032] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Experimental studies during the last decade have revealed a number of signaling pathways that are critical for the development and maintenance of the intestinal epithelium and that demonstrate the molecular basis for a variety of diseases. The Notch-Delta, Wnt, Hedge Hog, TGF-beta, and other signaling pathways have been shown to form and steadily maintain the crypt-villus system, generating the proper quantities of highly-specialized cells, and ultimately defining the architectural shape of the system. Based on the characterized phenotypes and functional defects of mice resulting from various targeted knockouts, and overexpression and misexpressions of genes, a picture is emerging of the sequence of gene expression events from within the epithelium, and in the underlying mesenchyme that contribute to the regulation of cell differentiation and proliferation. This review focuses on the contributions of multiple signaling pathways to intestinal epithelial proliferation, differentiation, and structural organization, as well as the possible opportunities for cross-talk between pathways. The Notch pathway's potential ability to maintain and regulate the intestinal epithelial stem cell is discussed, in addition to its role as the primary mediator of lineage specification. Recent research that has shed light on the function of Wnt signaling and epithelial-mesenchymal cross-talk during embryonic and postnatal development is examined, along with data on the interplay of heparan sulfate proteoglycans in the signaling process.
Collapse
Affiliation(s)
- Andrew L Hauck
- Department of Cell and Structural Biology, University of Illinois, Urbana, Illinois 61801, USA
| | | | | | | | | | | |
Collapse
|
1119
|
Hauwel M, Furon E, Gasque P. Molecular and cellular insights into the coxsackie-adenovirus receptor: role in cellular interactions in the stem cell niche. ACTA ACUST UNITED AC 2005; 48:265-72. [PMID: 15850666 DOI: 10.1016/j.brainresrev.2004.12.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2004] [Accepted: 12/09/2004] [Indexed: 11/21/2022]
Abstract
In recent years, progress has been made in characterizing the molecular and cellular elements that are responsible for the regeneration in the damaged brain and highlighting the key role of the stromal-vascular 'environment' to orchestrate secondary neurogenesis and repair. Indeed, the ability of the stem cells to self-renew and differentiate is tightly regulated by stromal ependymal cells and endothelial cells expressing molecular cues that constitute the extracellular stem cell 'niche'. Several soluble growth factors such as EGF, TGFbeta, FGF2, SDF-1alpha and Noggin are important signals for the stem cell niche but little is known about the role of membrane-bound molecules in intercellular communications between the niche and the stem cells. In this mini-review, we highlight the emerging role of a family of adhesion molecules in the control of secondary neurogenesis. The coxsackie-adenovirus receptor (CAR) is a 46 kDa transmembrane protein and a member of the immunoglobulin super family. It is close structurally and evolutionary to other adhesion molecules involved in cell-cell interactions during embryogenesis, broadly expressed in the developing central nervous system but restricted to ependymal cells in the adult brain. This unique location and its newly established signalling properties further support the role of CAR in intercellular communications. Elucidating the other signalling molecules and manipulating the stromal-vascular niche for example by adenovirus gene therapy remain important goals for future clinical applications.
Collapse
Affiliation(s)
- Mathieu Hauwel
- Department of Medical biochemistry and Immunology, Brain Inflammation and Immunity Group (BIIG), Cardiff University, Heath Park, UK.
| | | | | |
Collapse
|
1120
|
Gotts JE, Chesselet MF. Vascular changes in the subventricular zone after distal cortical lesions. Exp Neurol 2005; 194:139-50. [PMID: 15899251 DOI: 10.1016/j.expneurol.2005.02.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2004] [Revised: 01/30/2005] [Accepted: 02/01/2005] [Indexed: 01/19/2023]
Abstract
One of the effects of cortical lesions is to produce cell proliferation in the subventricular zone (SVZ), a neurogenic zone of the adult brain distal from the lesion. The mechanisms of these effects are unknown. Recent evidence points to a relationship between the vasculature and neurogenesis both in vitro and in vivo. In the present study, we asked whether cortical lesions induced vascular modifications in the distal SVZ in vivo. Lesions of the frontoparietal cortex were produced by thermocoagulation of pial blood vessels, a method that leads to highly reproducible loss of all cortical layers, sparing the corpus callosum and underlying striatum. These lesions induced increased immunoreactivity for vascular endothelial growth factor (VEGF) around the walls of SVZ vessels, at a considerable distance from the lesion. Vascular permeability was markedly increased in both the SVZ and RMS by 3 days after the injury. A dramatic increase in endothelial proliferation was followed by expansion of the local SVZ vascular tree 7 days after the injury. This time course corresponded to the proliferative changes in the SVZ, and a tight correlation was observed between the number of blood vessels and the increase in SVZ cell number. The data demonstrate that thermocoagulatory cortical lesions induce distal vascular changes that could play a role in lesion-induced SVZ expansion.
Collapse
Affiliation(s)
- Jeffrey E Gotts
- Department of Neurology, Geffen School of Medicine at UCLA, 710 Westwood Plaza, RNRC B114, Los Angeles, CA 90095, USA
| | | |
Collapse
|
1121
|
Fre S, Huyghe M, Mourikis P, Robine S, Louvard D, Artavanis-Tsakonas S. Notch signals control the fate of immature progenitor cells in the intestine. Nature 2005; 435:964-8. [PMID: 15959516 DOI: 10.1038/nature03589] [Citation(s) in RCA: 685] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2004] [Accepted: 04/04/2005] [Indexed: 01/27/2023]
Abstract
The Notch signalling pathway plays a crucial role in specifying cellular fates in metazoan development by regulating communication between adjacent cells. Correlative studies suggested an involvement of Notch in intestinal development. Here, by modulating Notch activity in the mouse intestine, we directly implicate Notch signals in intestinal cell lineage specification. We also show that Notch activation is capable of amplifying the intestinal progenitor pool while inhibiting cell differentiation. We conclude that Notch activity is required for the maintenance of proliferating crypt cells in the intestinal epithelium.
Collapse
Affiliation(s)
- Silvia Fre
- Department of Cell Biology, Harvard Medical School, Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | |
Collapse
|
1122
|
Scheffler B, Walton NM, Lin DD, Goetz AK, Enikolopov G, Roper SN, Steindler DA. Phenotypic and functional characterization of adult brain neuropoiesis. Proc Natl Acad Sci U S A 2005; 102:9353-8. [PMID: 15961540 PMCID: PMC1150897 DOI: 10.1073/pnas.0503965102] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The modern concept of neurogenesis in the adult brain is predicated on the premise that multipotent glial cells give rise to new neurons throughout life. Although extensive evidence exists indicating that this is the case, the transition from glial to neuronal phenotype remains poorly understood. A unique monolayer cell-culture system was developed to induce, expose, and recapitulate the entire developmental series of events of subventricular zone (SVZ) neurogenesis. We show here, using immunophenotypic, ultrastructural, electrophysiological, and time-lapse analyses, that SVZ-derived glial fibrillary acidic protein(low)/A2B5+/nestin+ candidate founder cells undergo metamorphosis to eventually generate large numbers of fully differentiated interneuron phenotypes. A model of postnatal neurogenesis is considered in light of known embryonic events and reveals a limited developmental potential of SVZ stem/progenitor cells, whereby ancestral cells in both embryonic and postnatal/adult settings give rise to glia and GABAergic interneurons.
Collapse
Affiliation(s)
- Bjorn Scheffler
- Departments of Neuroscience and Neurosurgery, University of Florida, McKnight Brain Institute, Gainesville, FL 32610, USA
| | | | | | | | | | | | | |
Collapse
|
1123
|
Yamashita YM, Fuller MT, Jones DL. Signaling in stem cell niches: lessons from the Drosophila germline. J Cell Sci 2005; 118:665-72. [PMID: 15701923 DOI: 10.1242/jcs.01680] [Citation(s) in RCA: 157] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Stem cells are cells that, upon division, can produce new stem cells as well as daughter cells that initiate differentiation along a specific lineage. Studies using the Drosophila germline as a model system have demonstrated that signaling from the stem cell niche plays a crucial role in controlling stem cell behavior. Surrounding support cells secrete growth factors that activate signaling within adjacent stem cells to specify stem cell self-renewal and block differentiation. In addition, cell-cell adhesion between stem cells and surrounding support cells is important for holding stem cells close to self-renewal signals. Furthermore, a combination of localized signaling and autonomously acting proteins might polarize stem cells in such a way as to ensure asymmetric stem cell divisions. Recent results describing stem cell niches in other adult stem cells, including hematopoietic and neural stem cells, have demonstrated that the features characteristic of stem cell niches in Drosophila gonads might be conserved.
Collapse
Affiliation(s)
- Yukiko M Yamashita
- Department of Development Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|
1124
|
Nagase T, Nagase M, Yoshimura K, Fujita T, Koshima I. Angiogenesis within the developing mouse neural tube is dependent on sonic hedgehog signaling: possible roles of motor neurons. Genes Cells 2005; 10:595-604. [PMID: 15938717 DOI: 10.1111/j.1365-2443.2005.00861.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Embryonic morphogenesis of vascular and nervous systems is tightly coordinated, and recent studies revealed that some neurogenetic factors such as Sonic hedgehog (Shh) also exhibit angiogenetic potential. Vascularization within the developing mouse neural tube depends on vessel sprouting from the surrounding vascular plexus. Previous studies implicated possible roles of VEGF/Flk-1 and Angiopoietin-1(Ang-1)/Tie-2 signaling as candidate molecules functioning in this process. Examining gene expressions of these factors at embryonic day (E) 9.5 and 10.5, we unexpectedly found that both VEGF and Ang-1 were expressed in the motor neurons in the ventral neural tube. The motor neurons were indeed located in the close vicinity of the infiltrating vessels, suggesting involvement of motor neurons in the sprouting. To substantiate this possibility, we inhibited induction of the motor neurons in the cultured mouse embryos by cyclopamine, a Shh signaling blocker. The vessel sprouting was dramatically impaired by inhibition of Shh signaling, together with nearly complete loss of the motor neurons. Expression of Ang-1, but not VEGF, within the neural tube was remarkably reduced in the cyclopamine treated embryos. These results suggest that the neural tube angiogenesis is dependent on Shh signaling, and mediated, at least in part, by the Ang-1 positive motor neurons.
Collapse
Affiliation(s)
- Takashi Nagase
- Department of Plastic and Reconstructive Surgery, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | | | | | | | | |
Collapse
|
1125
|
|
1126
|
Bermpohl D, Halle A, Freyer D, Dagand E, Braun JS, Bechmann I, Schröder NWJ, Weber JR. Bacterial programmed cell death of cerebral endothelial cells involves dual death pathways. J Clin Invest 2005; 115:1607-15. [PMID: 15902310 PMCID: PMC1088015 DOI: 10.1172/jci23223] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Accepted: 03/23/2005] [Indexed: 01/19/2023] Open
Abstract
Major barriers separating the blood from tissue compartments in the body are composed of endothelial cells. Interaction of bacteria with such barriers defines the course of invasive infections, and meningitis has served as a model system to study endothelial cell injury. Here we report the impressive ability of Streptococcus pneumoniae, clinically one of the most important pathogens, to induce 2 morphologically distinct forms of programmed cell death (PCD) in brain-derived endothelial cells. Pneumococci and the major cytotoxins H2O2 and pneumolysin induce apoptosis-like PCD independent of TLR2 and TLR4. On the other hand, pneumococcal cell wall, a major proinflammatory component, causes caspase-driven classical apoptosis that is mediated through TLR2. These findings broaden the scope of bacterial-induced PCD, link these effects to innate immune TLRs, and provide insight into the acute and persistent phases of damage during meningitis.
Collapse
Affiliation(s)
- Daniela Bermpohl
- Department of Neurology, Center of Anatomy, Charité--Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
1127
|
Fabricius D, Bonde S, Zavazava N. Induction of Stable Mixed Chimerism by Embryonic Stem Cells Requires Functional Fas/FasL Engagement. Transplantation 2005; 79:1040-4. [PMID: 15880040 DOI: 10.1097/01.tp.0000159142.62535.37] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Recent data show the efficacy of embryonic stem cells (ESC) to engraft in allogeneic recipients without host pretreatment. This property is due to their low expression of major histocompatibility complex (MHC) class I antigens and lack of MHC class II expression. Here, we tested the hypothesis that the constitutive FasL expression by ESC is a requirement for their stable engraftment in allogeneic recipients. METHODS MRL and MRL-lpr/lpr mice (H-2k) were infused allogeneic 129SvJ RW-4 (H-2b) ESC without host preconditioning. The development of mixed chimerism was monitored over 100 days by flow cytometry. RESULTS Mixed chimerism was detectable by day 7. The amount of donor cells detected varied between 3-5.5% and were lymphoid, but nonmyeloid. Only 50% of lpr/lpr mice engrafted and lost donor cells by day 28 post-ESC infusion. In contrast, >80% wild type mice engrafted and maintained mixed chimerism up to day 100. CONCLUSIONS These data suggest a critical role for Fas-FasL engagement in ESC engraftment. We conclude that ESC may induce clonal deletion of alloreactive T cells by Fas-induced apoptosis in recipient T cells, protecting them from rejection. The data provide a rationale for improved protocols for the achievement of robust ESC-induced mixed chimerism.
Collapse
Affiliation(s)
- Dorit Fabricius
- University of Iowa Hospitals and Clinics and VAMC Iowa City, Iowa City, Iowa 52242, USA
| | | | | |
Collapse
|
1128
|
Krylyshkina O, Chen J, Mebis L, Denef C, Vankelecom H. Nestin-immunoreactive cells in rat pituitary are neither hormonal nor typical folliculo-stellate cells. Endocrinology 2005; 146:2376-87. [PMID: 15677762 DOI: 10.1210/en.2004-1209] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Nestin is an intermediate filament protein that has originally been identified as a marker of neuroepithelial stem/progenitor cells. The present study explored whether nestin immunoreactivity (nestin-ir) is present in the rat pituitary and in which cell type(s). Nestin-ir was observed in scattered cells in the anterior, intermediate, and neural lobes. Nestin-ir cells were predominantly of stellate shape and were more numerous in immature than in adult animals. Nestin-ir did not colocalize with any pituitary hormone, and did not colocalize or only very sporadically with the folliculo-stellate cell marker S100. In the intermediate lobe, nestin-ir cells contained glial fibrillary acidic protein in an age-dependent manner. Nestin-ir cells were closely associated with endothelial and fibronectin-ir cells, but did mostly not coincide. Nestin-ir was not found in alpha-smooth muscle actin-ir myofibroblasts or in microglial cells. Regardless of age, nestin-ir was detected in some unidentifiable cells that border the pituitary cleft. Nestin-ir remained present in pituitary cultured as three-dimensional aggregates. Treatment with basic fibroblast growth factor or leukemia inhibitory factor increased the number of nestin-ir cells. Starting from anterior lobe cell monolayer cultures, nestin-ir cells could be selected and propagated to a virtually pure population. These nestin-ir cells displayed remarkable motility and proliferative activity, and did not express hormones, glial fibrillary acidic protein, or S100, but contained vimentin-, fibronectin-, and alpha-smooth muscle actin-ir. In conclusion, nestin-ir is present in the pituitary in cells that are neither hormonal nor typical folliculo-stellate. The expression pattern depends on age and lobe examined. Pericapillar localization suggests a pericyte phenotype for some of them. Whether the heterogeneous nestin-ir population also contains pituitary progenitor cells remains to be explored.
Collapse
Affiliation(s)
- Olga Krylyshkina
- Laboratory of Cell Pharmacology, Department of Molecular Cell Biology, University of Leuven, Campus Gasthuisberg, B-3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|
1129
|
Eichmann A, Le Noble F, Autiero M, Carmeliet P. Guidance of vascular and neural network formation. Curr Opin Neurobiol 2005; 15:108-15. [PMID: 15721752 DOI: 10.1016/j.conb.2005.01.008] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Blood vessels and nerves are structurally similar complex branched systems. Their guidance must be exquisitely regulated to ensure proper wiring of both networks. Recent results showed that specialized endothelial cells, resembling axonal growth cones, form the tips of growing capillaries. These endothelial tip cells guide outgrowing capillaries in response to gradients of extracellular matrix-bound vascular endothelial growth factor. Several axon guidance molecules, including Semaphorins, Netrins, Ephrins and Slits, have also been implicated in vessel pathfinding and network formation. In particular, Semaphorin3E and its receptor plexinD1 in addition to the Netrin receptor UNC5B have recently been shown to direct endothelial tip cell navigation.
Collapse
Affiliation(s)
- Anne Eichmann
- INSERM U 36, Collège de France, 11 Place Marcelin Berthelot, 75005 Paris, France.
| | | | | | | |
Collapse
|
1130
|
Limbourg FP, Takeshita K, Radtke F, Bronson RT, Chin MT, Liao JK. Essential role of endothelial Notch1 in angiogenesis. Circulation 2005; 111:1826-32. [PMID: 15809373 PMCID: PMC2633594 DOI: 10.1161/01.cir.0000160870.93058.dd] [Citation(s) in RCA: 215] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Notch signaling influences binary cell fate decisions in a variety of tissues. The Notch1 receptor is widely expressed during embryogenesis and is essential for embryonic development. Loss of global Notch1 function results in early embryonic lethality, but the cell type responsible for this defect is not known. Here, we identify the endothelium as the primary target tissue affected by Notch1 signaling. METHODS AND RESULTS We generated an endothelium-specific deletion of Notch1 using Tie2Cre and conditional Notch1(flox/flox) mice. Mutant embryos lacking endothelial Notch1 died at approximately embryonic day 10.5 with profound vascular defects in placenta, yolk sac, and embryo proper, whereas heterozygous deletion had no effect. In yolk sacs of mutant embryos, endothelial cells formed a primary vascular plexus indicative of intact vasculogenesis but failed to induce the secondary vascular remodeling required to form a mature network of well-organized large and small blood vessels, which demonstrates a defect in angiogenesis. These vascular defects were also evident in the placenta, where blood vessels failed to invade the placental labyrinth, and in the embryo proper, where defective blood vessel maturation led to pericardial and intersomitic hemorrhage. Enhanced activation of caspase-3 was detected in endothelial and neural cells of mutant mice, which resulted in enhanced apoptotic degeneration of somites and the neural tube. CONCLUSIONS These findings recapitulate the vascular phenotype of global Notch1-/- mutants and indicate an essential cell-autonomous role of Notch1 signaling in the endothelium during vascular development. These results may have important clinical implications with regard to Notch1 signaling in adult angiogenesis.
Collapse
MESH Headings
- Animals
- Apoptosis
- Caspase 3
- Caspases/metabolism
- Embryo Loss
- Embryo, Mammalian
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/embryology
- Endothelium, Vascular/physiology
- Genotype
- Hemorrhage/etiology
- Mice
- Mice, Mutant Strains
- Neovascularization, Physiologic
- Neural Tube Defects/etiology
- Neural Tube Defects/pathology
- Placenta/blood supply
- Receptor, Notch1
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Signal Transduction
- Somites/pathology
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Transcription Factors/physiology
Collapse
Affiliation(s)
- Florian P Limbourg
- Vascular Medicine Research Unit, Brigham & Women's Hospital, and Harvard Medical School, Boston, Mass, USA
| | | | | | | | | | | |
Collapse
|
1131
|
Abstract
Many cancers seem to depend on a small population of 'cancer stem cells' for their continued growth and propagation. The leukaemia stem cell (LSC) was the first such cell to be described. The origins of these cells are controversial, and their biology - like that of their normal-tissue counterpart, the haematopoietic stem cell (HSC) - is still not fully elucidated. However, the LSC is likely to be the most crucial target in the treatment of leukaemias, and a thorough understanding of its biology - particularly of how the LSC differs from the HSC - might allow it to be selectively targeted, improving therapeutic outcome.
Collapse
Affiliation(s)
- Brian J P Huntly
- Brian J. P. Huntly is at the Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | |
Collapse
|
1132
|
Trosko JE, Upham BL. The emperor wears no clothes in the field of carcinogen risk assessment: ignored concepts in cancer risk assessment. Mutagenesis 2005; 20:81-92. [PMID: 15784692 DOI: 10.1093/mutage/gei017] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The following is a position paper challenging the paradigm that 'carcinogen = mutagen', and that the current rodent bioassay to predict risks to human cancers is relevant and useful. Specifically, we review current observations concerning carcinogenesis that might lead to another approach for assessing the identification of human carcinogenic hazards and the risk assessment that chemicals might pose. We give a brief review of the multistage and multimechanism process of cancer in a tissue that involves not only genotoxic but also epigenetic events, and the importance of stem and progenitor cells in the development of cancer. We focus on the often ignored 'epigenetic' effects of carcinogens and the role of cell communication systems in epigenetically altering gene expression that leads to an imbalance of cell proliferation, differentiation and apoptosis in a tissue that can contribute to the cancer process. To draw attention to the fact that the current paradigm and policy to test toxic chemicals is often misleading and incorrect, we discuss how oxidative stress, in spite of the DNA damaging data, most probably contributes to cancer at the epigenetic level. Additionally, we briefly review how this mutagenic concept has greatly diverted attention away from doing research on the lower molecular weight, non-genotoxic, polycyclic aromatic hydrocarbons (PAHs), and how these low molecular weight PAHs are etiologically more relevant to the disease potential of environmental mixtures such as cigarette smoke.
Collapse
Affiliation(s)
- James E Trosko
- National Food Safety Toxicology Center, Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI 48824, USA.
| | | |
Collapse
|
1133
|
Duda DG, Jain RK. Pleiotropy of tissue-specific growth factors: from neurons to vessels via the bone marrow. J Clin Invest 2005; 115:596-8. [PMID: 15765145 PMCID: PMC1052004 DOI: 10.1172/jci24511] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Recent evidence has demonstrated that endothelial-specific growth factors affect the development of apparently unrelated organs and cells. Expanding this evidence further, new findings in this issue of the JCI show that neurotrophic factors can affect neovascularization. Neurotrophic factors achieve proangiogenic effects not only by directly affecting endothelial cells, but also by recruiting hematopoietic precursors. Further understanding of the biology of angiogenic factors, as well as of the function of hematopoietic cells in tissue neovascularization, will lead to improved therapeutic strategies for the treatment of diseases ranging from ischemia to cancer.
Collapse
Affiliation(s)
- Dan G Duda
- Edwin L. Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
1134
|
Kaigler D, Krebsbach PH, West ER, Horger K, Huang YC, Mooney DJ. Endothelial cell modulation of bone marrow stromal cell osteogenic potential. FASEB J 2005; 19:665-7. [PMID: 15677693 DOI: 10.1096/fj.04-2529fje] [Citation(s) in RCA: 182] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In the context of bone development and regeneration, the intimate association of the vascular endothelium with osteogenic cells suggests that endothelial cells (ECs) may directly regulate the differentiation of osteoprogenitor cells. To investigate this question, bone marrow stromal cells (BMSCs) were cultured: in the presence of EC-conditioned medium, on EC extracellular matrix, and in EC cocultures with and without cell contact. RNA and protein were isolated from ECs and analyzed by reverse transcriptase-polymerase chain reaction and Western blotting, respectively, for expression of bone morphogenetic protein 2 (BMP-2). In animal studies, BMSCs and ECs were cotransplanted into severe combined immunodeficient mice on biodegradable polymer matrices, and histomorphometric analysis was performed to determine the extent of new bone and blood vessel formation. ECs significantly increased BMSC osteogenic differentiation in vitro only when cultured in direct contact. ECs expressed BMP-2, and experiments employing interfering RNA inhibition confirmed its production as contributing to the increased BMSC osteogenic differentiation. In vivo, cotransplantation of ECs with BMSCs resulted in greater bone formation than did transplantation of BMSCs alone. These data suggest that ECs function not only to form the microvasculature that delivers nutrients to developing bone but also to modulate the differentiation of osteoprogenitor cells in vitro and in vivo.
Collapse
Affiliation(s)
- Darnell Kaigler
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | |
Collapse
|
1135
|
Bissell MJ, Labarge MA. Context, tissue plasticity, and cancer: are tumor stem cells also regulated by the microenvironment? Cancer Cell 2005; 7:17-23. [PMID: 15652746 PMCID: PMC2933216 DOI: 10.1016/j.ccr.2004.12.013] [Citation(s) in RCA: 211] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mina J Bissell
- Department Cancer Biology, Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, California 94720, USA.
| | | |
Collapse
|
1136
|
Abstract
Postnatal stem cells have been isolated from a variety of tissues and they are highly expected to have potentiality to be utilized for cell-based clinical therapies. Bone marrow stromal stem cells (BMSSCs) derived from bone marrow stromal tissue have been identified as a population of multipotent mesenchymal stem cells that are capable of differentiating into osteoblasts, adipocytes, chondrocytes, muscle cells, and neural cells. The most significant tissue regeneration trait of BMSSCs is their in vivo bone regeneration capability, which has been widely studied for understanding molecular and cellular mechanisms of osteogenesis, and, more importantly, developing into a stem-cell-based therapy. Recent studies further demonstrated that BMSSC-mediated bone regeneration is a promising approach for regenerative medicine in clinical trials. However, there are some fundamental questions that remain to be answered prior to successful utilization of BMSSCs in clinical therapy. For instance, how to maintain stemness of BMSSCs will be a critical issue for developing methodologies to propagate multi-potential stem cells in vitro, in order to allow the development of effective clinical therapies.
Collapse
Affiliation(s)
- Wataru Sonoyama
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
1137
|
Sim FJ, Goldman SA. White matter progenitor cells reside in an oligodendrogenic niche. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2005:61-81. [PMID: 16315609 DOI: 10.1007/3-540-27626-2_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Affiliation(s)
- F J Sim
- Dicision of Cell and Gene Therapy, Department of Neurology, University of Rochester Medical Center, NY 14642, USA
| | | |
Collapse
|
1138
|
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
|
1139
|
Nachman RL, Jaffe EA. Endothelial cell culture: beginnings of modern vascular biology. J Clin Invest 2004; 114:1037-40. [PMID: 15489946 PMCID: PMC522268 DOI: 10.1172/jci23284] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Endothelial cells derived from human umbilical veins were first successfully cultured in vitro in 1973. Weibel-Palade bodies and the von Willebrand factor antigen were used as morphological, immunohistochemical, and functional markers to unequivocally identify the cells. These landmark studies helped initiate the growth of modern vascular biology.
Collapse
Affiliation(s)
- Ralph L Nachman
- Department of Medicine, Weill Medical College of Cornell University, New York, New York, USA
| | | |
Collapse
|
1140
|
Reif A, Schmitt A, Fritzen S, Chourbaji S, Bartsch C, Urani A, Wycislo M, Mössner R, Sommer C, Gass P, Lesch KP. Differential effect of endothelial nitric oxide synthase (NOS-III) on the regulation of adult neurogenesis and behaviour. Eur J Neurosci 2004; 20:885-95. [PMID: 15305857 DOI: 10.1111/j.1460-9568.2004.03559.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Although it has been postulated that adult neurogenesis, i.e. the generation of functional neurons from progenitor cells in the mammalian brain, is involved in both the pathogenesis of depressive disorders and the therapeutic effect of antidepressant drugs, its regulation is still poorly understood. Nitric oxide, a gaseous messenger molecule, represents a possible modulating agent as it is involved in learning and memory formation as well as synapto- and morphogenesis. Here we investigated whether adult neurogenesis is altered in mice lacking endothelial nitric oxide synthase (NOS-III). Compared to wild-type littermates, NOS-III-deficient mice showed a significant reduction in neuronal progenitor cell proliferation in the dentate gyrus, suggesting a role for NOS-III in the stimulation of neuroneogenesis. NeuN, beta-III-tubulin and GFAP double-immunolabelling demonstrated that proliferating progenitor cells differentiate preferentially into neurons but not into astrocytes. However, when the survival rate of newly formed cells was examined no difference between wild-type and NOS-III knockout mice was found, suggesting that NOS-III selectively exerts its effects on the proliferation of progenitor cells. This might be mediated by a decrease in vascular endothelial growth factor (VEGF) transcripts in the hippocampus of knockout animals. At the behavioural level, while NOS-III knockout mice displayed better and faster learning in a learned helplessness paradigm, no depression-like behaviours were observed. In conclusion, our results indicated that NOS-III is involved in the proliferation of neuronal progenitor cells, although behavioural analysis does not provide evidence for a pro-depressive effect of reduced neuroneogenesis.
Collapse
Affiliation(s)
- Andreas Reif
- Molecular and Clinical Psychobiology, Department of Psychiatry and Psychotherapy, Julius-Maximilians-University Würzburg, Füchsleinstr. 15, D-97080 Würzburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1141
|
|
1142
|
Yasuhara T, Shingo T, Date I. The Potential Role of Vascular Endothelial Growth Factor in the Central Nervous System. Rev Neurosci 2004; 15:293-307. [PMID: 15526553 DOI: 10.1515/revneuro.2004.15.4.293] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
During the past decade, vascular endothelial growth factor (VEGF) has been widely investigated, and reported to have pleiotropic functions in the central nervous system (CNS) and its supporting physiological environment. VEGF is involved in not only such well-known functions as angiogenesis, accentuation of vessel permeability, and glial proliferation, but also more recently acknowledged functions such as neuroprotection and even neurogenesis itself. Most recently, the neurogenesis function has attracted much attention, and a number of research groups have taken up the challenge of elucidating this activity. In keeping with this trend, our knowledge of VEGF receptors has increased, and certain suggestions concerning the mechanisms of neuroprotection have come to light in the course of the ongoing work, though at times what the researchers had to work with was only a tiny percent of the signal transduction of VEGF. Together with flt-1 (VEGF receptor 1) and flk-1 (VEGF receptor 2), neuropilin (NP) is frequently described as being involved in the neuroprotective effects of VEGF. In this review, both the direct and indirect neuroprotective effects of VEGF, including various signaling pathways as well as the neurogenesis induced by this factor, are discussed in the context of the newly emerging insights into the biological mechanisms of VEGF and closely related, interacting molecules.
Collapse
Affiliation(s)
- Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan.
| | | | | |
Collapse
|