51
|
Furube E, Morita M, Miyata S. Characterization of neural stem cells and their progeny in the sensory circumventricular organs of adult mouse. Cell Tissue Res 2015; 362:347-65. [PMID: 25994374 DOI: 10.1007/s00441-015-2201-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 04/11/2015] [Indexed: 01/19/2023]
Abstract
Although evidence has accumulated that neurogenesis and gliogenesis occur in the subventricular zone (SVZ) and subgranular zone (SGZ) of adult mammalian brains, recent studies indicate the presence of neural stem cells (NSCs) in adult brains, particularly the circumventricular regions. In the present study, we aimed to determine characterization of NSCs and their progenitor cells in the sensory circumventricular organs (CVOs), including organum vasculosum of the lamina terminalis, subfornical organ, and area postrema of adult mouse. There were two types of NSCs: tanycyte-like ependymal cells and astrocyte-like cells. Astrocyte-like NSCs proliferated slowly and oligodendrocyte progenitor cells (OPCs) and neural progenitor cells (NPCs) actively divided. Molecular marker protein expression of NSCs and their progenitor cells were similar to those reported in the SVZ and SGZ, except that astrocyte-like NSCs expressed S100β. These circumventricular NSCs possessed the capacity to give rise to oligodendrocytes and sparse numbers of neurons and astrocytes in the sensory CVOs and adjacent brain regions. The inhibition of vascular endothelial growth factor (VEGF) signaling by using a VEGF receptor-associated tyrosine kinase inhibitor AZD2171 largely suppressed basal proliferation of OPCs. A single systemic administration of lipopolysaccharide attenuated proliferation of OPCs and induced remarkable proliferation of microglia. The present study indicates that sensory circumventricular NSCs provide new neurons and glial cells in the sensory CVOs and adjacent brain regions.
Collapse
Affiliation(s)
- Eriko Furube
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, 606-8585, Japan
| | - Mitsuhiro Morita
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan
| | - Seiji Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, 606-8585, Japan.
| |
Collapse
|
52
|
Developmental expression of vascular endothelial growth factor receptor 3 and vascular endothelial growth factor C in forebrain. Neuroscience 2015; 303:544-57. [PMID: 25943477 DOI: 10.1016/j.neuroscience.2015.04.063] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 04/25/2015] [Accepted: 04/27/2015] [Indexed: 01/19/2023]
Abstract
Increased understanding of the neurovascular niche suggests that development of the central nervous system (CNS) and its vasculature is coordinated through shared regulatory factors. These include the vascular endothelial growth factor (VEGF) family, reported to promote neuroproliferation and neuroprotection in addition to angiogenesis via its receptors VEGFR1-3. VEGFR3, a mediator of lymphangiogenesis, is expressed in murine and rat brain from early gestation, has been associated with neural progenitors and neurons (Choi et al., 2010) and oligodendroglia (Le Bras et al., 2006) in the developing cortex and is reported to mediate adult neurogenesis in the subventricular zone (SVZ) (Calvo et al., 2011). The early expression pattern of VEGFR3 protein and its cellular associations has not as yet been comprehensively reported. We describe the temporal expression of VEGFR3 protein at a cellular level and its close association with its VEGFC ligand, determined by double-labeling immunohistochemistry in the developing rat brain from embryonic day (E) 13 to postnatal day (P) 23. We found high expression of VEGFR3 in the ventricular zone and along radial glia in early gestation in association with neural stem cells and neuroblasts. Similar expression patterns were seen in the immature olfactory bulb and optic cup. In later development we found less expression by neural progenitors in proliferative regions including the SVZ and dentate gyrus of the hippocampus. In contrast, VEGFR3 expression increased with development in the cortex in neurons and astrocytes, and appeared in the emerging population of oligodendroglial progenitors. High expression in ventricular ependyma, choroid plexus and pigmented retinal epithelium was noted from E18. VEGFC ligand was found in association with VEGFR3 throughout development, with highest expression in embryonic stages. Our findings suggest an important role for VEGFC/VEGFR3 signaling in neuronal proliferation in early forebrain development, and ongoing functions with niche neurogenesis, glial and ependymal function in the maturing postnatal brain.
Collapse
|
53
|
Bhuiyan M, Kim JC, Hwang SN, Lee MY, Kim S. Ischemic tolerance is associated with VEGF-C and VEGFR-3 signaling in the mouse hippocampus. Neuroscience 2015; 290:90-102. [DOI: 10.1016/j.neuroscience.2015.01.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 10/24/2022]
|
54
|
|
55
|
Han J, Calvo CF, Kang TH, Baker KL, Park JH, Parras C, Levittas M, Birba U, Pibouin-Fragner L, Fragner P, Bilguvar K, Duman RS, Nurmi H, Alitalo K, Eichmann AC, Thomas JL. Vascular endothelial growth factor receptor 3 controls neural stem cell activation in mice and humans. Cell Rep 2015; 10:1158-72. [PMID: 25704818 PMCID: PMC4685253 DOI: 10.1016/j.celrep.2015.01.049] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 12/12/2014] [Accepted: 01/20/2015] [Indexed: 01/17/2023] Open
Abstract
Neural stem cells (NSCs) continuously produce new neurons within the adult mammalian hippocampus. NSCs are typically quiescent but activated to self-renew or differentiate into neural progenitor cells. The molecular mechanisms of NSC activation remain poorly understood. Here, we show that adult hippocampal NSCs express vascular endothelial growth factor receptor (VEGFR) 3 and its ligand VEGF-C, which activates quiescent NSCs to enter the cell cycle and generate progenitor cells. Hippocampal NSC activation and neurogenesis are impaired by conditional deletion of Vegfr3 in NSCs. Functionally, this is associated with compromised NSC activation in response to VEGF-C and physical activity. In NSCs derived from human embryonic stem cells (hESCs), VEGF-C/VEGFR3 mediates intracellular activation of AKT and ERK pathways that control cell fate and proliferation. These findings identify VEGF-C/VEGFR3 signaling as a specific regulator of NSC activation and neurogenesis in mammals.
Collapse
Affiliation(s)
- Jinah Han
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Charles-Félix Calvo
- Université Pierre and Marie Curie-Paris 6, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; INSERM/CNRS U-1127/UMR-7225, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France
| | - Tae Hyuk Kang
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Kasey L Baker
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - June-Hee Park
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Carlos Parras
- Université Pierre and Marie Curie-Paris 6, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; INSERM/CNRS U-1127/UMR-7225, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France
| | - Marine Levittas
- Université Pierre and Marie Curie-Paris 6, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; INSERM/CNRS U-1127/UMR-7225, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France
| | - Ulrick Birba
- Université Pierre and Marie Curie-Paris 6, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; INSERM/CNRS U-1127/UMR-7225, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France
| | - Laurence Pibouin-Fragner
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Pascal Fragner
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Kaya Bilguvar
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Harri Nurmi
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland
| | - Anne C Eichmann
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510-3221, USA.
| | - Jean-Léon Thomas
- Université Pierre and Marie Curie-Paris 6, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; INSERM/CNRS U-1127/UMR-7225, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, 75013 Paris, France; Department of Neurology, Yale University School of Medicine, New Haven, CT 06510-3221, USA.
| |
Collapse
|
56
|
Shin YJ, Riew TR, Park JH, Pak HJ, Lee MY. Expression of vascular endothelial growth factor-C (VEGF-C) and its receptor (VEGFR-3) in the glial reaction elicited by human mesenchymal stem cell engraftment in the normal rat brain. J Histochem Cytochem 2014; 63:170-80. [PMID: 25473093 DOI: 10.1369/0022155414564218] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To determine whether vascular endothelial growth factor-C (VEGF-C) and its receptor (VEGFR-3) are involved in the glial reaction elicited by transplanted mesenchymal stem cells (MSCs), we examined the cellular localization of VEGF-C and VEGFR-3 proteins in the striatum of adult normal rats that received bone marrow-derived human MSCs. The MSC grafts were infiltrated with activated microglia/macrophages and astrocytes over a 2-week period post-transplantation, which appeared to parallel the loss of transplanted MSCs. VEGF-C/VEGFR-3 was expressed in activated microglia/macrophages recruited to the graft site, where the induction of VEGF-C protein was rather late compared with that of its receptor. VEGF-C protein was absent or very weak on day 3, whereas VEGFR-3 immunoreactivity was evident within the first three days. Furthermore, within three days, VEGF-C could be detected in the brain macrophages localized immediately adjacent to the needle track. At the same time, almost all the brain macrophages in both regions expressed VEGFR-3. Reactive astrocytes at the graft site expressed VEGFR-3, but not VEGF-C. These data demonstrated the characteristic time- and cell-dependent expression patterns for VEGF-C and VEGFR-3 within the engrafted brain tissue, suggesting that they may contribute to neuroinflammation in MSC transplantation, possibly through the recruitment and/or activation of microglia/macrophages and astrogliosis.
Collapse
Affiliation(s)
- Yoo-Jin Shin
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea (YJS, TRR, JHP, HJP, MYL)
| | - Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea (YJS, TRR, JHP, HJP, MYL)
| | - Joo-Hee Park
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea (YJS, TRR, JHP, HJP, MYL)
| | - Ha-Jin Pak
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea (YJS, TRR, JHP, HJP, MYL)
| | - Mun-Yong Lee
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea (YJS, TRR, JHP, HJP, MYL)
| |
Collapse
|
57
|
Sundar SJ, Hsieh JK, Manjila S, Lathia JD, Sloan A. The role of cancer stem cells in glioblastoma. Neurosurg Focus 2014; 37:E6. [DOI: 10.3171/2014.9.focus14494] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recurrence in glioblastoma is nearly universal, and its prognosis remains dismal despite significant advances in treatment over the past decade. Glioblastoma demonstrates considerable intratumoral phenotypic and molecular heterogeneity and contains a population of cancer stem cells that contributes to tumor propagation, maintenance, and treatment resistance. Cancer stem cells are functionally defined by their ability to self-renew and to differentiate, and they constitute the diverse hierarchy of cells composing a tumor. When xenografted into an appropriate host, they are capable of tumorigenesis. Given the critical role of cancer stem cells in the pathogenesis of glioblastoma, research into their molecular and phenotypic characteristics is a therapeutic priority. In this review, the authors discuss the evolution of the cancer stem cell model of tumorigenesis and describe the specific role of cancer stem cells in the pathogenesis of glioblastoma and their molecular and microenvironmental characteristics. They also discuss recent clinical investigations into targeted therapies against cancer stem cells in the treatment of glioblastoma.
Collapse
Affiliation(s)
| | - Jason K. Hsieh
- 1Case Western Reserve University School of Medicine
- 2Cleveland Clinic Lerner College of Medicine
| | - Sunil Manjila
- 3Department of Neurological Surgery, University Hospitals Case Medical Center
| | - Justin D. Lathia
- 2Cleveland Clinic Lerner College of Medicine
- 4Department of Cellular & Molecular Medicine, Lerner Research Institute, Cleveland Clinic; and
- 5Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Andrew Sloan
- 1Case Western Reserve University School of Medicine
- 3Department of Neurological Surgery, University Hospitals Case Medical Center
- 5Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
58
|
Armendáriz BG, Masdeu MDM, Soriano E, Ureña JM, Burgaya F. The diverse roles and multiple forms of focal adhesion kinase in brain. Eur J Neurosci 2014; 40:3573-90. [DOI: 10.1111/ejn.12737] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 08/25/2014] [Indexed: 02/04/2023]
Affiliation(s)
- Beatriz G. Armendáriz
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| | - Maria del Mar Masdeu
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| | - Eduardo Soriano
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| | - Jesús M. Ureña
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| | - Ferran Burgaya
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| |
Collapse
|
59
|
Dimou L, Götz M. Glial cells as progenitors and stem cells: new roles in the healthy and diseased brain. Physiol Rev 2014; 94:709-37. [PMID: 24987003 DOI: 10.1152/physrev.00036.2013] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The diverse functions of glial cells prompt the question to which extent specific subtypes may be devoted to a specific function. We discuss this by reviewing one of the most recently discovered roles of glial cells, their function as neural stem cells (NSCs) and progenitor cells. First we give an overview of glial stem and progenitor cells during development; these are the radial glial cells that act as NSCs and other glial progenitors, highlighting the distinction between the lineage of cells in vivo and their potential when exposed to a different environment, e.g., in vitro. We then proceed to the adult stage and discuss the glial cells that continue to act as NSCs across vertebrates and others that are more lineage-restricted, such as the adult NG2-glia, the most frequent progenitor type in the adult mammalian brain, that remain within the oligodendrocyte lineage. Upon certain injury conditions, a distinct subset of quiescent astrocytes reactivates proliferation and a larger potential, clearly demonstrating the concept of heterogeneity with distinct subtypes of, e.g., astrocytes or NG2-glia performing rather different roles after brain injury. These new insights not only highlight the importance of glial cells for brain repair but also their great potential in various aspects of regeneration.
Collapse
Affiliation(s)
- Leda Dimou
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University, Munich, Germany; Institute for Stem Cell Research, HelmholtzZentrum, Neuherberg, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Magdalena Götz
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University, Munich, Germany; Institute for Stem Cell Research, HelmholtzZentrum, Neuherberg, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
60
|
Vascular endothelial growth factors A and C are induced in the SVZ following neonatal hypoxia-ischemia and exert different effects on neonatal glial progenitors. Transl Stroke Res 2014; 4:158-70. [PMID: 23565129 DOI: 10.1007/s12975-012-0213-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Episodes of neonatal hypoxia-ischemia (H-I) are strongly associated with cerebral palsy and a wide spectrum of other neurological deficits in children. Two key processes required to repair damaged organs are to amplify the number of precursors capable of regenerating damaged cells and to direct their differentiation towards the cell types that need to be replaced. Since hypoxia induces vascular endothelial growth factor (VEGF) production, it is logical to predict that VEGFs are key mediators of tissue repair after H-I injury. The goal of this study was to test the hypothesis that certain VEGF isoforms increase during recovery from neonatal H-I and that they would differentially affect the proliferation and differentiation of subventricular zone (SVZ) progenitors. During the acute recovery period from H-I both VEGF-A and VEGF-C were transiently induced in the SVZ, which correlated with an increase in SVZ blood vessel diameter. These growth factors were produced by glial progenitors, astrocytes and to a lesser extent, microglia. VEGF-A promoted the production of astrocytes from SVZ glial progenitors while VEGF-C stimulated the proliferation of both early and late oligodendrocyte progenitors, which was abolished by blocking the VEGFR-3. Altogether, these results provide new insights into the signals that coordinate the reactive responses of the progenitors in the SVZ to neonatal H-I. Our studies further suggest that therapeutics that extend VEGF-C production and/or agonists that stimulate the VEGFR-3 will promote oligodendrocyte progenitor cell development to enhance myelination after perinatal brain injury.
Collapse
|
61
|
Hennekam syndrome can be caused by FAT4 mutations and be allelic to Van Maldergem syndrome. Hum Genet 2014; 133:1161-7. [DOI: 10.1007/s00439-014-1456-y] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/15/2014] [Indexed: 11/27/2022]
|
62
|
Guérit S, Allain AE, Léon C, Cazenave W, Ferrara N, Branchereau P, Bikfalvi A. VEGF modulates synaptic activity in the developing spinal cord. Dev Neurobiol 2014; 74:1110-22. [DOI: 10.1002/dneu.22187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 04/25/2014] [Indexed: 01/31/2023]
Affiliation(s)
- Sylvaine Guérit
- Université Bordeaux; Angiogenesis and Tumor Microenvironment Laboratory; F-33615 Pessac Cedex France
- INSERM; Angiogenesis and Tumor Microenvironment Laboratory; F-33615 Pessac Cedex France
| | - Anne-Emilie Allain
- Université Bordeaux, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287; F-33615 Pessac Cedex France
- CNRS; Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287; F-33615 Pessac Cedex France
| | - Céline Léon
- Université Bordeaux; Angiogenesis and Tumor Microenvironment Laboratory; F-33615 Pessac Cedex France
- INSERM; Angiogenesis and Tumor Microenvironment Laboratory; F-33615 Pessac Cedex France
| | - William Cazenave
- Université Bordeaux, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287; F-33615 Pessac Cedex France
- CNRS; Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287; F-33615 Pessac Cedex France
| | | | - Pascal Branchereau
- Université Bordeaux, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287; F-33615 Pessac Cedex France
- CNRS; Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287; F-33615 Pessac Cedex France
| | - Andréas Bikfalvi
- Université Bordeaux; Angiogenesis and Tumor Microenvironment Laboratory; F-33615 Pessac Cedex France
- INSERM; Angiogenesis and Tumor Microenvironment Laboratory; F-33615 Pessac Cedex France
| |
Collapse
|
63
|
Pronto-Laborinho AC, Pinto S, de Carvalho M. Roles of vascular endothelial growth factor in amyotrophic lateral sclerosis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:947513. [PMID: 24987705 PMCID: PMC4022172 DOI: 10.1155/2014/947513] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/24/2014] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal devastating neurodegenerative disorder, involving progressive degeneration of motor neurons in spinal cord, brainstem, and motor cortex. Riluzole is the only drug approved in ALS but it only confers a modest improvement in survival. In spite of a high number of clinical trials no other drug has proved effectiveness. Recent studies support that vascular endothelial growth factor (VEGF), originally described as a key angiogenic factor, also plays a key role in the nervous system, including neurogenesis, neuronal survival, neuronal migration, and axon guidance. VEGF has been used in exploratory clinical studies with promising results in ALS and other neurological disorders. Although VEGF is a very promising compound, translating the basic science breakthroughs into clinical practice is the major challenge ahead. VEGF-B, presenting a single safety profile, protects motor neurons from degeneration in ALS animal models and, therefore, it will be particularly interesting to test its effects in ALS patients. In the present paper the authors make a brief description of the molecular properties of VEGF and its receptors and review its different features and therapeutic potential in the nervous system/neurodegenerative disease, particularly in ALS.
Collapse
Affiliation(s)
- Ana Catarina Pronto-Laborinho
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Instituto de Medicina Molecular (IMM), Translational Clinical Physiology Unit, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Susana Pinto
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Instituto de Medicina Molecular (IMM), Translational Clinical Physiology Unit, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Mamede de Carvalho
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Instituto de Medicina Molecular (IMM), Translational Clinical Physiology Unit, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Department of Neurosciences, Hospital Santa Maria, Centro Hospitalar Lisboa Norte, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| |
Collapse
|
64
|
Medina-Rodríguez EM, Arenzana FJ, Bribián A, de Castro F. Protocol to isolate a large amount of functional oligodendrocyte precursor cells from the cerebral cortex of adult mice and humans. PLoS One 2013; 8:e81620. [PMID: 24303061 PMCID: PMC3841116 DOI: 10.1371/journal.pone.0081620] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/24/2013] [Indexed: 01/09/2023] Open
Abstract
During development, oligodendrocytes are generated from oligodendrocyte precursor cells (OPCs), a cell type that is a significant proportion of the total cells (3-8%) in the adult central nervous system (CNS) of both rodents and humans. Adult OPCs are responsible for the spontaneous remyelination that occurs in demyelinating diseases like Multiple Sclerosis (MS) and they constitute an interesting source of cells for regenerative therapy in such conditions. However, there is little data regarding the neurobiology of adult OPCs isolated from mice since an efficient method to isolate them has yet to be established. We have designed a protocol to obtain viable adult OPCs from the cerebral cortex of different mouse strains and we have compared its efficiency with other well-known methods. In addition, we show that this protocol is also useful to isolate functional OPCs from human brain biopsies. Using this method we can isolate primary cortical OPCs in sufficient quantities so as to be able to study their survival, maturation and function, and to facilitate an evaluation of their utility in myelin repair.
Collapse
Affiliation(s)
| | | | - Ana Bribián
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Toledo, Spain
- * E-mail:
| |
Collapse
|
65
|
Kwon HB, Fukuhara S, Asakawa K, Ando K, Kashiwada T, Kawakami K, Hibi M, Kwon YG, Kim KW, Alitalo K, Mochizuki N. The parallel growth of motoneuron axons with the dorsal aorta depends on Vegfc/Vegfr3 signaling in zebrafish. Development 2013; 140:4081-90. [PMID: 24046321 PMCID: PMC3913045 DOI: 10.1242/dev.091702] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Blood vessels and neurons grow often side by side. However, the molecular and cellular mechanisms underlying their parallel development remain unclear. Here, we report that a subpopulation of secondary motoneurons extends axons ventrally outside of the neural tubes and rostrocaudally as a fascicle beneath the dorsal aorta (DA) in zebrafish. We tried to clarify the mechanism by which these motoneuron axons grow beneath the DA and found that Vegfc in the DA and Vegfr3 in the motoneurons were essential for the axon growth. Forced expression of either Vegfc in arteries or Vegfr3 in motoneurons resulted in enhanced axon growth of motoneurons over the DA. Both vegfr3 morphants and vegfc morphants lost the alignment of motoneuron axons with DA. In addition, forced expression of two mutant forms of Vegfr3 in motoneurons, potentially trapping endogenous Vegfc, resulted in failure of growth of motoneuron axons beneath the DA. Finally, a vegfr3 mutant fish lacked the motoneuron axons beneath the DA. Collectively, Vegfc from the preformed DA guides the axon growth of secondary motoneurons.
Collapse
Affiliation(s)
- Hyouk-Bum Kwon
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H. The brain tumor microenvironment. Glia 2013; 60:502-14. [PMID: 22379614 DOI: 10.1002/glia.21264] [Citation(s) in RCA: 289] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
High-grade brain tumors are heterogeneous with respect to the composition of bona fide tumor cells and with respect to a range of intermingling parenchymal cells. Glioblastomas harbor multiple cell types, some with increased tumorigenicity and stem cell-like capacity. The stem-like cells maybe the cells of origin for tumor relapse. However, the tumor-associated parenchymal cells such as vascular cells,microglia, peripheral immune cells, and neural precursor cells also play a vital role in controlling the course of pathology.In this review, we describe the multiple interactions of bulk glioma cells and glioma stem cells with parenchymal cell populations and highlight the pathological impact as well as signaling pathways known for these types of cell-cell communication. The tumor-vasculature not only nourishes glioblastomas, but also provides a specialized niche for these stem-like cells. In addition, microglial cells,which can contribute up to 30% of a brain tumor mass,play a role in glioblastoma cell invasion. Moreover, non-neoplastic astrocytes can be converted into a reactive phenotype by the glioma microenvironment and can then secrete a number of factors which influences tumor biology. The young brain may have the capacity to inhibit gliomagenesis by the endogenous neural precursor cells, which secrete tumor suppressive factors. The factors, pathways, and interactions described in this review provide a new prospective on the cell biology of primary brain tumors, which may ultimately generate new treatment modalities. However, our picture of the multiple interactions between parenchymal and tumor cells is still incomplete.
Collapse
Affiliation(s)
- Nikki A Charles
- Brain Tumor Center and Department of Neurosurgery, Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | |
Collapse
|
67
|
Keifer OP, O'Connor DM, Boulis NM. Gene and protein therapies utilizing VEGF for ALS. Pharmacol Ther 2013; 141:261-71. [PMID: 24177067 DOI: 10.1016/j.pharmthera.2013.10.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/04/2013] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that is usually fatal within 2-5years. Unfortunately, the only treatment currently available is riluzole, which has a limited efficacy. As a redress, there is an expanding literature focusing on other potential treatments. One such potential treatment option utilizes the vascular endothelial growth factor (VEGF) family, which includes factors that are primarily associated with angiogenesis but are now increasingly recognized to have neurotrophic effects. Reduced expression of a member of this family, VEGF-A, in mice results in neurodegeneration similar to that of ALS, while treatment of animal models of ALS with either VEGF-A gene therapy or VEGF-A protein has yielded positive therapeutic outcomes. These basic research findings raise the potential for a VEGF therapy to be translated to the clinic for the treatment of ALS. This review covers the VEGF family, its receptors and neurotrophic effects as well as VEGF therapy in animal models of ALS and advances towards clinical trials.
Collapse
Affiliation(s)
- Orion P Keifer
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, United States
| | - Deirdre M O'Connor
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, United States
| | - Nicholas M Boulis
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, United States.
| |
Collapse
|
68
|
Miyamoto N, Pham LDD, Seo JH, Kim KW, Lo EH, Arai K. Crosstalk between cerebral endothelium and oligodendrocyte. Cell Mol Life Sci 2013; 71:1055-66. [PMID: 24132511 DOI: 10.1007/s00018-013-1488-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/12/2013] [Accepted: 09/30/2013] [Indexed: 01/19/2023]
Abstract
It is now relatively well accepted that the cerebrovascular system does not merely provide inert pipes for blood delivery to the brain. Cerebral endothelial cells may compose an embedded bunker of trophic factors that contribute to brain homeostasis and function. Recent findings suggest that soluble factors from cerebral endothelial cells nourish neighboring cells, such as neurons and astrocytes. Although data are strongest in supporting mechanisms of endothelial-neuron and/or endothelial-astrocyte trophic coupling, it is likely that similar interactions also exist between cerebral endothelial cells and oligodendrocyte lineage cells. In this mini-review, we summarize current advances in the field of endothelial-oligodendrocyte trophic coupling. These endothelial-oligodendrocyte interactions may comprise the oligovascular niche to maintain their cellular functions and sustain ongoing angiogenesis/oligodendrogenesis. Importantly, it should be noted that the cell-cell interactions are not static-the trophic coupling is disturbed under acute phase after brain injury, but would be recovered in the chronic phase to promote brain remodeling and repair. Oligodendrocyte lineage cells play critical roles in white matter function, and under pathological conditions, oligodendrocyte dysfunction lead to white matter damage. Therefore, a deeper understanding of the mechanisms of endothelial-oligodendrocyte trophic coupling may lead to new therapeutic approaches for white matter-related diseases, such as stroke or vascular dementia.
Collapse
Affiliation(s)
- Nobukazu Miyamoto
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, MGH East 149-2401, Charlestown, MA, 02129, USA
| | | | | | | | | | | |
Collapse
|
69
|
Chaitanya GV, Omura S, Sato F, Martinez NE, Minagar A, Ramanathan M, Guttman BW, Zivadinov R, Tsunoda I, Alexander JS. Inflammation induces neuro-lymphatic protein expression in multiple sclerosis brain neurovasculature. J Neuroinflammation 2013; 10:125. [PMID: 24124909 PMCID: PMC3854084 DOI: 10.1186/1742-2094-10-125] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/24/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is associated with ectopic lymphoid follicle formation. Podoplanin+ (lymphatic marker) T helper17 (Th17) cells and B cell aggregates have been implicated in the formation of tertiary lymphoid organs (TLOs) in MS and experimental autoimmune encephalitis (EAE). Since podoplanin expressed by Th17 cells in MS brains is also expressed by lymphatic endothelium, we investigated whether the pathophysiology of MS involves inductions of lymphatic proteins in the inflamed neurovasculature. METHODS We assessed the protein levels of lymphatic vessel endothelial hyaluronan receptor and podoplanin, which are specific to the lymphatic system and prospero-homeobox protein-1, angiopoietin-2, vascular endothelial growth factor-D, vascular endothelial growth factor receptor-3, which are expressed by both lymphatic endothelium and neurons. Levels of these proteins were measured in postmortem brains and sera from MS patients, in the myelin proteolipid protein (PLP)-induced EAE and Theiler's murine encephalomyelitis virus (TMEV) induced demyelinating disease (TMEV-IDD) mouse models and in cell culture models of inflamed neurovasculature. RESULTS AND CONCLUSIONS Intense staining for LYVE-1 was found in neurons of a subset of MS patients using immunohistochemical approaches. The lymphatic protein, podoplanin, was highly expressed in perivascular inflammatory lesions indicating signaling cross-talks between inflamed brain vasculature and lymphatic proteins in MS. The profiles of these proteins in MS patient sera discriminated between relapsing remitting MS from secondary progressive MS and normal patients. The in vivo findings were confirmed in the in vitro cell culture models of neuroinflammation.
Collapse
Affiliation(s)
- Ganta Vijay Chaitanya
- Department of Molecular & Cellular Physiology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Seiichi Omura
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Fumitaka Sato
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Nicholas E Martinez
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Alireza Minagar
- Department of Neurology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Murali Ramanathan
- Department of Neurology, State University of New York, Buffalo, NY, USA
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY, USA
| | | | - Robert Zivadinov
- Department of Neurology, State University of New York, Buffalo, NY, USA
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, NY, USA
| | - Ikuo Tsunoda
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Jonathan S Alexander
- Department of Molecular & Cellular Physiology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| |
Collapse
|
70
|
Annenkov A. Receptor tyrosine kinase (RTK) signalling in the control of neural stem and progenitor cell (NSPC) development. Mol Neurobiol 2013; 49:440-71. [PMID: 23982746 DOI: 10.1007/s12035-013-8532-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 08/09/2013] [Indexed: 01/04/2023]
Abstract
Important developmental responses are elicited in neural stem and progenitor cells (NSPC) by activation of the receptor tyrosine kinases (RTK), including the fibroblast growth factor receptors, epidermal growth factor receptor, platelet-derived growth factor receptors and insulin-like growth factor receptor (IGF1R). Signalling through these RTK is necessary and sufficient for driving a number of developmental processes in the central nervous system. Within each of the four RTK families discussed here, receptors are activated by sets of ligands that do not cross-activate receptors of the other three families, and therefore, their activation can be independently regulated by ligand availability. These RTK pathways converge on a conserved core of signalling molecules, but differences between the receptors in utilisation of signalling molecules and molecular adaptors for intracellular signal propagation become increasingly apparent. Intracellular inhibitors of RTK signalling are widely involved in the regulation of developmental signalling in NSPC and often determine developmental outcomes of RTK activation. In addition, cellular responses of NSPC to the activation of a given RTK may be significantly modulated by signal strength. Cellular propensity to respond also plays a role in developmental outcomes of RTK signalling. In combination, these mechanisms regulate the balance between NSPC maintenance and differentiation during development and in adulthood. Attribution of particular developmental responses of NSPC to specific pathways of RTK signalling becomes increasingly elucidated. Co-activation of several RTK in developing NSPC is common, and analysis of co-operation between their signalling pathways may advance knowledge of RTK role in NSPC development.
Collapse
Affiliation(s)
- Alexander Annenkov
- Bone and Joint Research Unit, William Harvey Research Institute, Bart's and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK,
| |
Collapse
|
71
|
Koch S, Claesson-Welsh L. Signal transduction by vascular endothelial growth factor receptors. Cold Spring Harb Perspect Med 2013; 2:a006502. [PMID: 22762016 DOI: 10.1101/cshperspect.a006502] [Citation(s) in RCA: 629] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vascular endothelial growth factors (VEGFs) are master regulators of vascular development and of blood and lymphatic vessel function during health and disease in the adult. It is therefore important to understand the mechanism of action of this family of five mammalian ligands, which act through three receptor tyrosine kinases (RTKs). In addition, coreceptors like neuropilins (NRPs) and integrins associate with the ligand/receptor signaling complex and modulate the output. Therapeutics to block several of the VEGF signaling components have been developed with the aim to halt blood vessel formation, angiogenesis, in diseases that involve tissue growth and inflammation, such as cancer. In this review, we outline the current information on VEGF signal transduction in relation to blood and lymphatic vessel biology.
Collapse
Affiliation(s)
- Sina Koch
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, 751 85 Uppsala, Sweden
| | | |
Collapse
|
72
|
Chauvet S, Burk K, Mann F. Navigation rules for vessels and neurons: cooperative signaling between VEGF and neural guidance cues. Cell Mol Life Sci 2013; 70:1685-703. [PMID: 23475066 PMCID: PMC11113827 DOI: 10.1007/s00018-013-1278-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/22/2022]
Abstract
Many organs, such as lungs, nerves, blood and lymphatic vessels, consist of complex networks that carry flows of information, gases, and nutrients within the body. The morphogenetic patterning that generates these organs involves the coordinated action of developmental signaling cues that guide migration of specialized cells. Precision guidance of endothelial tip cells by vascular endothelial growth factors (VEGFs) is well established, and several families of neural guidance molecules have been identified to exert guidance function in both the nervous and the vascular systems. This review discusses recent advances in VEGF research, focusing on the emerging role of neural guidance molecules as key regulators of VEGF function during vascular development and on the novel role of VEGFs in neural cell migration and nerve wiring.
Collapse
Affiliation(s)
- Sophie Chauvet
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Campus de Luminy Case 908, 13288 Marseille Cedex 9, France
| | - Katja Burk
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Campus de Luminy Case 908, 13288 Marseille Cedex 9, France
| | - Fanny Mann
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Campus de Luminy Case 908, 13288 Marseille Cedex 9, France
| |
Collapse
|
73
|
Carmeliet P, Ruiz de Almodovar C, Carmen RDA. VEGF ligands and receptors: implications in neurodevelopment and neurodegeneration. Cell Mol Life Sci 2013; 70:1763-78. [PMID: 23475071 PMCID: PMC11113464 DOI: 10.1007/s00018-013-1283-7] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/15/2022]
Abstract
Intensive research in the last decade shows that the prototypic angiogenic factor vascular endothelial growth factor (VEGF) can have direct effects in neurons and modulate processes such as neuronal migration, axon outgrowth, axon guidance and neuronal survival. Depending on the neuronal cell type and the process, VEGF seems to exert these effects by signaling via different receptors. It is also becoming clear that other VEGF ligands such as VEGF-B, -C and -D can act in various neuronal cell types as well. Moreover, apart from playing a role in physiological conditions, VEGF and VEGF-B have been related to different neurological disorders. We give an update on how VEGF controls different processes during neurodevelopment as well as on its role in several neurodegenerative disorders. We also discuss recent findings demonstrating that other VEGF ligands influence processes such as neurogenesis and dendrite arborization and participate in neurodegeneration.
Collapse
Affiliation(s)
- Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, K.U.Leuven, 3000, Leuven, Belgium.
| | | | | |
Collapse
|
74
|
Interactions between VEGFR and Notch signaling pathways in endothelial and neural cells. Cell Mol Life Sci 2013; 70:1779-92. [PMID: 23479133 DOI: 10.1007/s00018-013-1312-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 02/25/2013] [Accepted: 02/25/2013] [Indexed: 12/25/2022]
Abstract
Notch cell interaction mechanism governs cell fate decisions in many different cell contexts throughout the lifetime of all Metazoan species. It links the fate of one cell to that of its neighbors through cell-to-cell contacts, and binding of Notch receptors expressed on one cell to their membrane bound ligands on an adjacent cell. Environmental cues, such as growth factors and extracellular matrix molecules, superimpose a dynamic regulation on this canonical Notch signaling pathway. In this review, we will focus on Notch signaling in the vertebrate vascular and nervous systems and examine its role in angiogenesis, neurogenesis, and neurovascular interactions. We will also highlight the molecular relationships of the Notch pathway with vascular endothelial growth factors (VEGFs) and their high-affinity tyrosine kinase VEGF receptors, key regulators of both angiogenesis and neurogenesis.
Collapse
|
75
|
Wittko-Schneider IM, Schneider FT, Plate KH. Brain homeostasis: VEGF receptor 1 and 2-two unequal brothers in mind. Cell Mol Life Sci 2013; 70:1705-25. [PMID: 23475067 PMCID: PMC3632714 DOI: 10.1007/s00018-013-1279-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/15/2022]
Abstract
Vascular endothelial growth factors (VEGFs), initially thought to act specifically on the vascular system, exert trophic effects on neural cells during development and adulthood. Therefore, the VEGF system serves as a promising therapeutic target for brain pathologies, but its simultaneous action on vascular cells paves the way for harmful side effects. To circumvent these deleterious effects, many studies have aimed to clarify whether VEGFs directly affect neural cells or if the effects are mediated secondarily via other cell types, like vascular cells. A great number of reports have shown the expression and function of VEGF receptors (VEGFRs), mainly VEGFR-1 and -2, in neural cells, where VEGFR-2 has been described as the major mediator of VEGF-A signals. This review aims to summarize and compare the divergent roles of VEGFR-1 and -2 during CNS development and homeostasis.
Collapse
Affiliation(s)
- Ina M Wittko-Schneider
- Neuroscience Center, Institute of Neurology (Edinger Institute), Goethe University Medical School, Heinrich-Hoffmann Strasse 7, 60528, Frankfurt, Germany.
| | | | | |
Collapse
|
76
|
Induction of vascular endothelial growth factor receptor-3 expression in perivascular cells of the ischemic core following focal cerebral ischemia in rats. Acta Histochem 2013; 115:170-7. [PMID: 22771250 DOI: 10.1016/j.acthis.2012.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 06/16/2012] [Accepted: 06/18/2012] [Indexed: 01/02/2023]
Abstract
Vascular endothelial growth factor receptor (VEGFR)-3, a receptor for VEGF-C and VEGF-D, has recently been reported to be induced within vessel-like structures in the ischemic brain. The purpose of the present study was to characterize and define further the cellular phenotypes of vascular-associated cells that manifest induced VEGFR-3 expression in a rat model of ischemic stroke. Vessel-associated cells expressing VEGFR-3 were found to be perivascular astrocytes in the peri-infarct region, whereas in the ischemic core, where astrocytes had virtually disappeared, induction of VEGFR-3 mRNA and protein was still prominent in vascular structures 3-7 days after reperfusion. VEGFR-3 and nestin expression were colocated in almost all cells associated with the vasculature in the ischemic core, and most (~82%) of the VEGFR-3/nestin double-labeled cells were proliferative. A subpopulation of these VEGFR-3-expressing cells appeared to be included in two immunophenotypically distinct perivascular cells: NG2-positive pericytes and ED2- or OX6-perivascular macrophages. However, most of these cells did not show markers for vasculature-associated cell types such as endothelial cells, microglia/macrophages, and smooth muscle cells. Thus, our data indicated that vasculature-associated VEGFR-3-expressing cells in the ischemic core may represent a heterogeneous population of cells with functional diversity, rather than a uniform cell type.
Collapse
|
77
|
Nieto-Estévez V, Pignatelli J, Araúzo-Bravo MJ, Hurtado-Chong A, Vicario-Abejón C. A global transcriptome analysis reveals molecular hallmarks of neural stem cell death, survival, and differentiation in response to partial FGF-2 and EGF deprivation. PLoS One 2013; 8:e53594. [PMID: 23308259 PMCID: PMC3538603 DOI: 10.1371/journal.pone.0053594] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 12/03/2012] [Indexed: 11/19/2022] Open
Abstract
Neurosphere cell culture is a commonly used model to study the properties and potential applications of neural stem cells (NSCs). However, standard protocols to culture NSCs have yet to be established, and the mechanisms underlying NSC survival and maintenance of their undifferentiated state, in response to the growth factors FGF-2 and EGF are not fully understood. Using cultures of embryonic and adult olfactory bulb stem cells (eOBSCs and aOBSCs), we analyzed the consequences of FGF-2 and EGF addition at different intervals on proliferation, cell cycle progression, cell death and differentiation, as well as on global gene expression. As opposed to cultures supplemented daily, addition of FGF-2 and EGF every 4 days significantly reduced the neurosphere volume and the total number of cells in the spheres, mainly due to increased cell death. Moreover, partial FGF-2 and EGF deprivation produced an increase in OBSC differentiation during the proliferative phase. These changes were more evident in aOBSC than eOBSC cultures. Remarkably, these effects were accompanied by a significant upregulation in the expression of endogenous Fgf-2 and genes involved in cell death and survival (Cryab), lipid catabolic processes (Pla2g7), cell adhesion (Dscaml1), cell differentiation (Dscaml1, Gpr17, S100b, Ndrg2) and signal transduction (Gpr17, Ndrg2). These findings support that a daily supply of FGF-2 and EGF is critical to maintain the viability and the undifferentiated state of NSCs in culture, and they reveal novel molecular hallmarks of NSC death, survival and the initiation of differentiation.
Collapse
Affiliation(s)
- Vanesa Nieto-Estévez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jaime Pignatelli
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Marcos J. Araúzo-Bravo
- Laboratory of Computational Biology and Bioinformatics, Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Anahí Hurtado-Chong
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carlos Vicario-Abejón
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- * E-mail:
| |
Collapse
|
78
|
Eichmann A, Thomas JL. Molecular parallels between neural and vascular development. Cold Spring Harb Perspect Med 2013; 3:a006551. [PMID: 23024177 DOI: 10.1101/cshperspect.a006551] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The human central nervous system (CNS) features a network of ~400 miles of blood vessels that receives >20% of the body's cardiac output and uses most of its blood glucose. Many human diseases, including stroke, retinopathy, and cancer, are associated with the biology of CNS blood vessels. These vessels originate from extrinsic cell populations, including endothelial cells and pericytes that colonize the CNS and interact with glia and neurons to establish the blood-brain barrier and control cerebrovascular exchanges. Neurovascular interactions also play important roles in adult neurogenic niches, which harbor a unique population of neural stem cells that are intimately associated with blood vessels. We here review the cellular and molecular mechanisms required to establish the CNS vascular network, with a special focus on neurovascular interactions and the functions of vascular endothelial growth factors.
Collapse
Affiliation(s)
- Anne Eichmann
- Center for Interdisciplinary Research in Biology, CNRS/UMR 7241-INSERM U1050, Collège de France, 75005 Paris, France.
| | | |
Collapse
|
79
|
Tsigelny IF, Kouznetsova VL, Baitaluk M, Changeux JP. A hierarchical coherent-gene-group model for brain development. GENES BRAIN AND BEHAVIOR 2012; 12:147-65. [DOI: 10.1111/gbb.12005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 05/30/2012] [Accepted: 11/15/2012] [Indexed: 12/28/2022]
Affiliation(s)
| | | | | | - J.-P. Changeux
- Department of Neuroscience, Collège de France & URA CNRS 2182; Institut Pasteur; Paris Cedex; France
| |
Collapse
|
80
|
Porter KI, Southey BR, Sweedler JV, Rodriguez-Zas SL. First survey and functional annotation of prohormone and convertase genes in the pig. BMC Genomics 2012; 13:582. [PMID: 23153308 PMCID: PMC3499383 DOI: 10.1186/1471-2164-13-582] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 06/22/2012] [Indexed: 11/18/2022] Open
Abstract
Background The pig is a biomedical model to study human and livestock traits. Many of these traits are controlled by neuropeptides that result from the cleavage of prohormones by prohormone convertases. Only 45 prohormones have been confirmed in the pig. Sequence homology can be ineffective to annotate prohormone genes in sequenced species like the pig due to the multifactorial nature of the prohormone processing. The goal of this study is to undertake the first complete survey of prohormone and prohormone convertases genes in the pig genome. These genes were functionally annotated based on 35 gene expression microarray experiments. The cleavage sites of prohormone sequences into potentially active neuropeptides were predicted. Results We identified 95 unique prohormone genes, 2 alternative calcitonin-related sequences, 8 prohormone convertases and 1 cleavage facilitator in the pig genome 10.2 assembly and trace archives. Of these, 11 pig prohormone genes have not been reported in the UniProt, UniGene or Gene databases. These genes are intermedin, cortistatin, insulin-like 5, orexigenic neuropeptide QRFP, prokineticin 2, prolactin-releasing peptide, parathyroid hormone 2, urocortin, urocortin 2, urocortin 3, and urotensin 2-related peptide. In addition, a novel neuropeptide S was identified in the pig genome correcting the previously reported pig sequence that is identical to the rabbit sequence. Most differentially expressed prohormone genes were under-expressed in pigs experiencing immune challenge relative to the un-challenged controls, in non-pregnant relative to pregnant sows, in old relative to young embryos, and in non-neural relative to neural tissues. The cleavage prediction based on human sequences had the best performance with a correct classification rate of cleaved and non-cleaved sites of 92% suggesting that the processing of prohormones in pigs is similar to humans. The cleavage prediction models did not find conclusive evidence supporting the production of the bioactive neuropeptides urocortin 2, urocortin 3, torsin family 2 member A, tachykinin 4, islet amyloid polypeptide, and calcitonin receptor-stimulating peptide 2 in the pig. Conclusions The present genomic and functional characterization supports the use of the pig as an effective animal model to gain a deeper understanding of prohormones, prohormone convertases and neuropeptides in biomedical and agricultural research.
Collapse
Affiliation(s)
- Kenneth I Porter
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
81
|
Stanton MJ, Dutta S, Zhang H, Polavaram NS, Leontovich AA, Hönscheid P, Sinicrope FA, Tindall DJ, Muders MH, Datta K. Autophagy control by the VEGF-C/NRP-2 axis in cancer and its implication for treatment resistance. Cancer Res 2012; 73:160-71. [PMID: 23149913 DOI: 10.1158/0008-5472.can-11-3635] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A major contributor to cancer mortality is recurrence and subsequent metastatic transformation following therapeutic intervention. Therefore, in order to develop new treatment modalities and improve the efficacy of current ones, it is important to understand the molecular mechanisms that promote resistance to therapy in cancer cells. One pathway contributing to therapy resistance is autophagy, a self-digestive process that can eliminate unnecessary or damaged organelles to protect cancer cells from death. We have found that the VEGF-C/NRP-2 axis is involved in the activation of autophagy, which helps cancer cell survival following treatment. Inhibition of mTOR complex 1 activity by this axis is the underlying mechanism for the activation of autophagy. Furthermore, we identified two VEGF-C/NRP-2-regulated genes, LAMP-2 and WDFY-1, that have previously been suggested to participate in autophagy and vesicular trafficking. Upregulation of WDFY-1 following VEGF-C or NRP-2 depletion contributes to cytotoxic drug-mediated cell death. Together, these data suggest a link between the VEGF-C/NRP-2 axis and cancer cell survival despite the presence of chemotherapy-induced stress. Effective targeting of this pathway may lead to the development of new cancer therapies.
Collapse
Affiliation(s)
- Marissa J Stanton
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Tillo M, Ruhrberg C, Mackenzie F. Emerging roles for semaphorins and VEGFs in synaptogenesis and synaptic plasticity. Cell Adh Migr 2012; 6:541-6. [PMID: 23076132 PMCID: PMC3547901 DOI: 10.4161/cam.22408] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Synapse formation, maintenance and plasticity are critical for the correct function of the nervous system and its target organs. During development, these processes enable the establishment of appropriate neural circuits. During adulthood, they allow adaptation to both physiological and environmental changes. In this review, we discuss emerging roles for two families of classical axon and vascular guidance cues in synaptogenesis and synaptic plasticity, the semaphorins and the vascular endothelial growth factors (VEGFs). Their contribution to synapse formation and function add a new facet to the spectrum of overlapping and complementary roles for these molecules in development, adulthood and disease.
Collapse
Affiliation(s)
- Miguel Tillo
- Institute of Ophthalmology, University College London, London, UK
| | | | | |
Collapse
|
83
|
Plexin A3 is involved in semaphorin 3F-mediated oligodendrocyte precursor cell migration. Neurosci Lett 2012; 530:127-32. [PMID: 23063687 DOI: 10.1016/j.neulet.2012.09.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 09/22/2012] [Accepted: 09/27/2012] [Indexed: 11/21/2022]
Abstract
Class 3 semaphorins are expressed in the neurodevelopmental or damage repair phase of the central nervous system (CNS). They play an important role in guiding axon growth and directing cell migration, including the migration of oligodendrocyte precursor cells (OPCs). As co-receptors for semaphorin 3F(sema3F), the expression and role of neuropilin-2 (NRP2) and plexin A3 in OPC migration are unclear. Using RT-PCR, Western blot analysis, and immunofluorescence, we demonstrated that primary OPCs and immature oligodendrocytes from neonatal rats express NRP2 and plexin A3. After transfection with NRP2 siRNA and plexin A3 siRNA, the number of migrating OPCs attracted to sema3F remarkably decreased. These results suggest that plexin A3 is expressed in OPCs and immature oligodendrocytes and is involved in OPC migration.
Collapse
|
84
|
Abstract
The neurovascular unit provides a conceptual framework for investigating the pathophysiology of how brain cells die after stroke, brain injury, and neurodegeneration. Emerging data now suggest that this concept can be further extended. Cell-cell signaling between neuronal, glial, and vascular elements in the brain not only mediates the mechanisms of acute injury, but integrated responses in these same elements may also be required for recovery as the entire neurovascular unit attempts to reorganize and remodel. Understanding the common signals and substrates of this transition between acute injury and delayed repair in the neurovascular unit may reveal useful paradigms for augmenting neuronal, glial, and vascular plasticity in damaged and diseased brain.
Collapse
Affiliation(s)
- Changhong Xing
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Boston, USA
| | | | | | | | | |
Collapse
|
85
|
Park JM, Shin YJ, Cho JM, Choi JY, Jeun SS, Cha JH, Lee MY. Upregulation of vascular endothelial growth factor receptor-3 in the spinal cord of Lewis rats with experimental autoimmune encephalomyelitis. J Histochem Cytochem 2012; 61:31-44. [PMID: 22983493 DOI: 10.1369/0022155412462975] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We investigated the spatiotemporal expression of vascular endothelial growth factor receptor-3 (VEGFR-3) in the spinal cord of Lewis rats with experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. VEGFR-3 mRNA and protein were constitutively expressed in gray matter neurons and in a few white matter astrocytes. Induction of VEGFR-3 occurred predominantly in perivascular infiltrated macrophages in the spinal cord white matter during the inductive phase of EAE. VEGFR-3 expression was also induced in activated microglial cells in the gray and white matter, mainly in the peak phase. In addition, reactive astrocytes in the white matter, but not in the gray matter, expressed VEGFR-3 as disease severity increased. These data suggest that VEGFR-3 is involved in the recruitment of monocytic macrophages and in glial reactions during EAE.
Collapse
Affiliation(s)
- Jang-Mi Park
- Department of Anatomy, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
86
|
Kudo Y, Iizuka S, Yoshida M, Nguyen PT, Siriwardena SBSM, Tsunematsu T, Ohbayashi M, Ando T, Hatakeyama D, Shibata T, Koizumi K, Maeda M, Ishimaru N, Ogawa I, Takata T. Periostin directly and indirectly promotes tumor lymphangiogenesis of head and neck cancer. PLoS One 2012; 7:e44488. [PMID: 22952986 PMCID: PMC3431354 DOI: 10.1371/journal.pone.0044488] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 08/03/2012] [Indexed: 11/24/2022] Open
Abstract
Background Metastasis to regional lymph nodes via lymphatic vessels plays a key role in cancer progression. Tumor lymphangiogenesis is known to promote lymphatic metastasis, and vascular endothelial growth factor C (VEGF-C) is a critical activator of tumor lymphangiogenesis during the process of metastasis. We previously identified periostin as an invasion- and angiogenesis-promoting factor in head and neck squamous cell carcinoma (HNSCC). In this study, we discovered a novel role for periostin in tumor lymphangiogenesis. Methods and Findings Periostin overexpression upregulated VEGF-C mRNA expression in HNSCC cells. By using conditioned media from periostin-overexpressing HNSCC cells, we examined tube formation of lymphatic endothelial cells. Conditioned media from periostin-overexpressing cells promoted tube formation. To know the correlation between periostin and VEGF-C, we compared Periostin expression with VEGF-C expression in 54 HNSCC cases by immunohistochemistry. Periostin expression was correlated well with VEGF-C expression in HNSCC cases. Moreover, correlation between periostin and VEGF-C secretion was observed in serum from HNSCC patients. Interestingly, periostin itself promoted tube formation of lymphatic endothelial cells independently of VEGF-C. Periostin-promoted lymphangiogenesis was mediated by Src and Akt activity. Indeed possible correlation between periostin and lymphatic status in periostin-overexpressing xenograft tumors and HNSCC cases was observed. Conclusions Our findings suggest that periostin itself as well as periostin-induced upregulation of VEGF-C may promote lymphangiogenesis. We suggest that periostin may be a marker for prediction of malignant behaviors in HNSCC and a potential target for future therapeutic intervention to obstruct tumoral lymphatic invasion and lymphangiogenesis in HNSCC patients.
Collapse
Affiliation(s)
- Yasusei Kudo
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
- * E-mail: (YK); (TT)
| | - Shinji Iizuka
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Maki Yoshida
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Phuong Thao Nguyen
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Samadarani B. S. M. Siriwardena
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Takaaki Tsunematsu
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Mariko Ohbayashi
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Toshinori Ando
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Daijiro Hatakeyama
- Department of Oral and Maxillofacial Sciences, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Toshiyuki Shibata
- Department of Oral and Maxillofacial Sciences, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Keiichi Koizumi
- Department of Kampo Diagnostic, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Masahiro Maeda
- Department of Research and Development, Immuno-Biological Laboratories, Co., Ltd., Fujioka-shi, Gunma, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Ikuko Ogawa
- Center of Oral Clinical Examination, Hiroshima University Hospital, Hiroshima, Japan
| | - Takashi Takata
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
- * E-mail: (YK); (TT)
| |
Collapse
|
87
|
Ramani P, Nash R, Radevsky L, Patel A, Luckett M, Rogers C. VEGF-C, VEGF-D and VEGFR-3 expression in peripheral neuroblastic tumours. Histopathology 2012; 61:1006-16. [PMID: 22804730 DOI: 10.1111/j.1365-2559.2012.04307.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS More than 50% of neuroblastomas (NBs) present with haematogenous and/or lymphatic metastasis; however, little is known about the clinicopathological significance in NBs of the key lymphangiogenesis growth factors vascular endothelial growth factor (VEGF)-C and VEGF-D and the receptor VEGFR-3. METHODS AND RESULTS Ninety-three NBs and nine ganglioneuromas (GNs) were immunostained for VEGF-C, VEGF-D and VEGFR-3. VEGF-C and VEGF-D were present in 76% and 82% of the NBs, respectively. There was no significant difference in VEGF-C expression between NBs and GNs. VEGF-D expression was significantly higher in NBs compared with GNs and in MYCN-amplified NBs. VEGFR-3 tumoral cell expression (VEGFR-3c), present in 48% of the NBs, was significantly higher in NBs from children ≥ 18 months at presentation and those belonging to a high-risk group. VEGFR-3 lymphovascular density was increased significantly in NBs compared with GNs and in NBs associated with adverse clinicopathological and biological factors. Lymphovascular invasion, assessed in VEGFR-3-stained vessels, was present in ∼50% of NBs. Cox regression analyses demonstrated that VEGFR-3c expression was associated with a significantly shorter event-free survival and that its effect was independent of the important pathological variable, mitosis-karyorrhexis index. CONCLUSIONS VEGF-D and VEGFR-3 up-regulation support tumour progression in NB and VEGFR-3c may provide a useful prognostic marker in NBs.
Collapse
Affiliation(s)
- Pramila Ramani
- School of Cellular and Molecular Medicine, University of Bristol, School of Medical Sciences, University Walk, Bristol, UK.
| | | | | | | | | | | |
Collapse
|
88
|
In vivo imaging of lymphatic vessels in development, wound healing, inflammation, and tumor metastasis. Proc Natl Acad Sci U S A 2012; 109:6223-8. [PMID: 22474390 DOI: 10.1073/pnas.1115542109] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Lymphatic vessel growth or lymphangiogenesis occurs during embryonic development and wound healing and plays an important role in tumor metastasis and inflammatory diseases. However, the possibility of noninvasive detection and quantification of lymphangiogenesis has been lacking. Here, we present the Vegfr3(EGFPLuc) mouse model, where an EGFP-luciferase fusion protein, expressed under the endogenous transcriptional control of the Vegfr3 gene, allows the monitoring of physiological and pathological lymphangiogenesis in vivo. We show tracking of lymphatic vessel development during embryogenesis as well as lymphangiogenesis induced by specific growth factors, during wound healing and in contact hypersensitivity (CHS)--induced inflammation where we also monitor down-regulation of lymphangiogenesis by the glucocorticoid dexamethasone. Importantly, the Vegfr3-reporter allowed us to tracking tumor-induced lymphangiogenesis at the tumor periphery and in lymph nodes in association with the metastatic process. This is the first reporter mouse model for luminescence imaging of lymphangiogenesis. It should provide an important tool for studying the involvement of lymphangiogenesis in pathological processes.
Collapse
|
89
|
Pham LDD, Hayakawa K, Seo JH, Nguyen MN, Som AT, Lee BJ, Guo S, Kim KW, Lo EH, Arai K. Crosstalk between oligodendrocytes and cerebral endothelium contributes to vascular remodeling after white matter injury. Glia 2012; 60:875-81. [PMID: 22392631 DOI: 10.1002/glia.22320] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 02/14/2012] [Indexed: 12/11/2022]
Abstract
After stroke and brain injury, cortical gray matter recovery involves mechanisms of neurovascular matrix remodeling. In white matter, however, the mechanisms of recovery remain unclear. In this study, we demonstrate that oligodendrocytes secrete matrix metalloproteinase-9 (MMP-9), which accelerates the angiogenic response after white matter injury. In primary oligodendrocyte cultures, treatment with the proinflammatory cytokine interleukin-1β (IL-1β) induced an upregulation and secretion of MMP-9. Conditioned media from IL-1β-stimulated oligodendrocytes significantly amplified matrigel tube formation in brain endothelial cells, indicating that MMP-9 from oligodendrocytes can promote angiogenesis in vitro. Next, we asked whether similar signals and substrates operate after white matter injury in vivo. Focal white matter injury and demyelination was induced in mice via stereotactic injection of lysophosphatidylcholine into corpus callosum. Western blot analysis showed that IL-1β expression was increased in damaged white matter. Immunostaining demonstrated MMP-9 signals in myelin-associated oligodendrocytic basic protein-positive oligodendrocytes. Treatment with an IL-1β-neutralizing antibody suppressed the MMP-9 response in oligodendrocytes. Finally, we confirmed that the broad spectrum MMP inhibitor GM6001 inhibited angiogenesis around the injury area in this white matter injury model. In gray matter, a neurovascular niche promotes cortical recovery after brain injury. Our study suggests that an analogous oligovascular niche may mediate recovery in white matter.
Collapse
Affiliation(s)
- Loc-Duyen D Pham
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Hayakawa K, Seo JH, Pham LDD, Miyamoto N, Som AT, Guo S, Kim KW, Lo EH, Arai K. Cerebral endothelial derived vascular endothelial growth factor promotes the migration but not the proliferation of oligodendrocyte precursor cells in vitro. Neurosci Lett 2012; 513:42-6. [PMID: 22342913 DOI: 10.1016/j.neulet.2012.02.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 01/31/2012] [Accepted: 02/01/2012] [Indexed: 11/17/2022]
Abstract
In gray matter, cerebral endothelium is known to provide trophic support for neighboring cells such as neurons. However, signaling from cerebral endothelium to white matter cells remains to be elucidated. Here, we show that vascular endothelial growth factor (VEGF-A) secreted from cerebral endothelial cells promotes the migration but not the proliferation of oligodendrocyte precursor cells (OPCs). Cultured OPCs were obtained from newborn rat cortex, and treatment with conditioned culture media of cerebral endothelial cells increased the OPC proliferation and migration. Importantly, co-treatment with anti-neutralizing antibody for Flk-1 (VEGF-receptor2) inhibited OPC movement but did not affect OPC propagation. Western blot and flow cytometry analyses confirmed that our cultured cerebral endothelial cells produced VEGF-A and our cultured OPCs expressed Flk-1. Taken together, our current data suggest that cerebral endothelium is supportive for oligodendrocyte lineage cells and VEGF-A may participate in the endothelium-OPC cell-cell signaling. This phenomenon may be important for white matter homeostasis.
Collapse
Affiliation(s)
- Kazuhide Hayakawa
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Neuroprotection of VEGF-expression neural stem cells in neonatal cerebral palsy rats. Behav Brain Res 2012; 230:108-15. [PMID: 22342488 DOI: 10.1016/j.bbr.2012.01.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/09/2012] [Accepted: 01/12/2012] [Indexed: 01/18/2023]
Abstract
Cerebral palsy (CP) is a very common neural system development disorder that can cause physical disability in human. Here, we studied the neuroprotective effect of vascular endothelial growth factor (VEGF)-transfected neural stem cells (NSCs) in newborn rats with cerebral palsy (CP). Seven-day-old Sprague-Dawley rats were randomly divided into four groups: sham operation (control group), PBS transplantation (PBS group), VEGF+NSCs transplantation (transgene NSCs group) and NSCs transplantation groups (NSCs group). PBS, Transgene NSCs and NSCs groups respectively received stereotactic injections of PBS, lentiviral vector (pGC-FU-VEGF) infected NSCs or a NSCs suspension in the left sensory-motor cortex 3 days after CP model was established. The NSCs activity, their impacts on neural cell growth and apoptosis, brain development and animal behaviors were examined on the animals up to age 35-days. As expected, unilateral carotid artery occlusion plus hypoxia (cerebral palsy model) resulted in severe neural developmental disorders, including slowed growth, increased in cortical neuron apoptosis, decreased cerebral cortex micro-vessel density and retarded behavior developments. Transplantation of NSCs not only resulted in increases in VEGF protein expression in rat brains, but also largely prevented the behavioral defects and brain tissue pathology that resulted from cerebral palsy procedure, with animals received VEGF transfected NSCs always being marginally better than these received un-transfected cells. In conclusion, NSCs transplantation can partially prevent/slow down the brain damages that are associated with CP in the newborn rats, suggesting a new possible strategy for CP treatment.
Collapse
|
92
|
|
93
|
Persano L, Rampazzo E, Della Puppa A, Pistollato F, Basso G. The three-layer concentric model of glioblastoma: cancer stem cells, microenvironmental regulation, and therapeutic implications. ScientificWorldJournal 2011; 11:1829-41. [PMID: 22125441 PMCID: PMC3217608 DOI: 10.1100/2011/736480] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 09/29/2011] [Indexed: 12/15/2022] Open
Abstract
Tumors arising in the central nervous system are thought to
originate from a sub-population of cells named cancer stem cells
(CSCs) or tumor initiating cells (TICs) that possess an immature
phenotype, combined with self-renewal and chemotherapy resistance
capacity. Moreover, in the last years, these cells have been
identified in particular brain tumor niches fundamental for
supporting their characteristics. In this paper, we report studies
from many authors demonstrating that hypoxia or the so called
“hypoxic niche” plays a crucial role in controlling CSC molecular
and phenotypic profile. We recently investigated the relationship
existing between Glioblastoma (GBM) stem cells and their niche,
defining the theory of three-concentric layers model for GBM mass.
According to this model, GBM stem cells reside preferentially
within the hypoxic core of the tumour mass, while more
differentiated cells are mainly localized along the peripheral and
vascularized part of the tumour. This GBM model provides
explanation of the effects mediated by the tumour microenvironment
on the phenotypic and molecular regulation of GBM stem cells,
describing their spatial distribution in the tumor bulk. Moreover,
we discuss the possible clinical implications of the creation of
this model for future GBM patient management and novel therapeutic
strategies development.
Collapse
Affiliation(s)
- Luca Persano
- Oncohematology Laboratory, Department of Paediatrics, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | | | | | | | | |
Collapse
|
94
|
Vascular endothelial growth factor regulates the migration of oligodendrocyte precursor cells. J Neurosci 2011; 31:10666-70. [PMID: 21775609 DOI: 10.1523/jneurosci.1944-11.2011] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Originally identified as an angiogenic factor, vascular endothelial growth factor (VEGF-A) is now known to play multiple roles in the CNS, including the direct regulation of neuronal and astrocytic functions. Here, we ask whether VEGF-A can also have a novel role in white matter by modulating oligodendrocyte precursor cells (OPCs). OPCs were cultured from rat neonatal cortex. Expression of VEGF-receptor2/KDR/Flk-1 was confirmed with Western blot and immunostaining. VEGF-A did not affect proliferation or differentiation in OPC cultures, but VEGF-A promoted OPC migration in a concentration-dependent manner. Consistent with this migration phenotype, VEGF-A-treated OPCs showed reorganization of actin cytoskeleton in leading-edge processes. VEGF-A-induced migration and actin reorganization were inhibited by an anti-Flk-1 receptor-blocking antibody. Mechanistically, VEGF-A induced binding of focal adhesion kinase (FAK) with paxillin. The FAK inhibitor PF573228 reduced VEGF-A-induced OPC migration. VEGF-A signaling also evoked a transient rise in reactive oxygen species (ROS), and OPC migration was increased when antioxidants were removed from the culture media. Our findings demonstrate that VEGF-A can induce OPC migration via an ROS- and FAK-dependent mechanism, and suggest a novel role for VEGF-A in white-matter maintenance and homeostasis.
Collapse
|
95
|
Zhang CQ, Shu HF, Yin Q, An N, Xu SL, Yin JB, Song YC, Liu SY, Yang H. Expression and cellular distribution of vascular endothelial growth factor-C system in cortical tubers of the tuberous sclerosis complex. Brain Pathol 2011; 22:205-18. [PMID: 21767323 DOI: 10.1111/j.1750-3639.2011.00519.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Cortical tubers are malformations of cortical development in patients with tuberous sclerosis complex (TSC), and highly associated with pediatric intractable epilepsy. Recent evidence has shown that signaling mediated through vascular endothelial growth factor-C (VEGF-C) and its receptors, VEGFR-2 and VEGFR-3, has direct effects on both neurons and glial cells. To understand the potential role of VEGF-C system in the pathogenesis of cortical tubers, we investigated the expression patterns of VEGF-C signaling in cortical tubers compared with age-matched normal control cortex (CTX). We found that VEGF-C, VEGFR-2 and VEGFR-3 were clearly upregulated in tubers at both the mRNA and protein levels, compared with CTX. The in situ hybridization and immunostaining results demonstrated that VEGF-C, VEGFR-2 and VEGFR-3 were highly expressed in dysplastic neurons (DNs), giant cells (GCs) and reactive astrocytes within tubers. Most DNs/GCs expressing VEGF-C and its receptors co-labeled with neuronal rather than astrocytic markers, suggesting a neuronal lineage. In addition, protein levels of Akt-1, p-Bad and ERK1/2, the important downstream factors of the VEGF-C pathway, were significantly increased in cortical tubers, indicating involvement of VEGF-C-dependent prosurvival signaling in cortical tubers. Taken together, our results suggest a putative role for the VEGF-C signaling pathway in the pathogenesis of cortical tubers.
Collapse
Affiliation(s)
- Chun-Qing Zhang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
VEGFR-3 controls tip to stalk conversion at vessel fusion sites by reinforcing Notch signalling. Nat Cell Biol 2011; 13:1202-13. [PMID: 21909098 DOI: 10.1038/ncb2331] [Citation(s) in RCA: 235] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 08/03/2011] [Indexed: 11/08/2022]
Abstract
Angiogenesis, the growth of new blood vessels, involves specification of endothelial cells to tip cells and stalk cells, which is controlled by Notch signalling, whereas vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3 have been implicated in angiogenic sprouting. Surprisingly, we found that endothelial deletion of Vegfr3, but not VEGFR-3-blocking antibodies, postnatally led to excessive angiogenic sprouting and branching, and decreased the level of Notch signalling, indicating that VEGFR-3 possesses passive and active signalling modalities. Furthermore, macrophages expressing the VEGFR-3 and VEGFR-2 ligand VEGF-C localized to vessel branch points, and Vegfc heterozygous mice exhibited inefficient angiogenesis characterized by decreased vascular branching. FoxC2 is a known regulator of Notch ligand and target gene expression, and Foxc2(+/-);Vegfr3(+/-) compound heterozygosity recapitulated homozygous loss of Vegfr3. These results indicate that macrophage-derived VEGF-C activates VEGFR-3 in tip cells to reinforce Notch signalling, which contributes to the phenotypic conversion of endothelial cells at fusion points of vessel sprouts.
Collapse
|
97
|
Abstract
VEGFs (vascular endothelial growth factors) control vascular development during embryogenesis and the function of blood vessels and lymphatic vessels in the adult. There are five related mammalian ligands, which act through three receptor tyrosine kinases. Signalling is modulated through neuropilins, which act as VEGF co-receptors. Heparan sulfate and integrins are also important modulators of VEGF signalling. Therapeutic agents that interfere with VEGF signalling have been developed with the aim of decreasing angiogenesis in diseases that involve tissue growth and inflammation, such as cancer. The present review will outline the current understanding and consequent biology of VEGF receptor signalling.
Collapse
|
98
|
Distribution of vascular endothelial growth factor receptor-3/Flt4 mRNA in adult rat central nervous system. J Chem Neuroanat 2011; 42:56-64. [DOI: 10.1016/j.jchemneu.2011.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 05/21/2011] [Accepted: 06/07/2011] [Indexed: 11/20/2022]
|
99
|
Piltonen M, Planken A, Leskelä O, Myöhänen T, Hänninen AL, Auvinen P, Alitalo K, Andressoo JO, Saarma M, Männistö P. Vascular endothelial growth factor C acts as a neurotrophic factor for dopamine neurons in vitro and in vivo. Neuroscience 2011; 192:550-63. [DOI: 10.1016/j.neuroscience.2011.06.084] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 06/16/2011] [Accepted: 06/30/2011] [Indexed: 11/26/2022]
|
100
|
Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H. The brain tumor microenvironment. Glia 2011; 59:1169-80. [PMID: 21446047 DOI: 10.1002/glia.21136] [Citation(s) in RCA: 403] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 12/08/2010] [Indexed: 02/06/2023]
Abstract
High-grade brain tumors are heterogeneous with respect to the composition of bona fide tumors cells and with respect to a range of intermingling parenchymal cells. Glioblastomas harbor multiple cell types, some with increased tumorigenicity and stem cell-like capacity. The stem-like cells may be the cells of origin for tumor relapse. However, the tumor-associated parenchymal cells-such as vascular cells, microglia, peripheral immune cells, and neural precursor cells-also play a vital role in controlling the course of pathology. In this review, we describe the multiple interactions of bulk glioma cells and glioma stem cells with parenchymal cell populations and highlight the pathological impact and signaling pathways known for these types of cell-cell communication. The tumor-vasculature not only nourishes glioblastomas, but also provides a specialized niche for these stem-like cells. In addition, microglial cells, which can contribute up to 30% of a brain tumor mass, play a role in glioblastoma cell invasion. Moreover, non-neoplastic astrocytes can be converted into a reactive phenotype by the glioma microenvironment and can then secrete a number of factors which influences tumor biology. The young brain may have the capacity to inhibit gliomagenesis by the endogenous neural stem and progenitor cells, which secrete tumor suppressive factors. The factors, pathways, and interactions described in this review provide a new prospective on the cell biology of primary brain tumors, which may ultimately generate new treatment modalities. However, our picture of the multiple interactions between parenchymal and tumor cells is still incomplete. © 2011 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Nikki A Charles
- Departments of Surgery (Neurosurgery), Neurology, and Cancer Biology & Genetics, Memorial Sloan-Kettering Cancer Center, New York, USA.
| | | | | | | | | |
Collapse
|