1
|
Friess D, Brauer S, Pöysti A, Choudhury C, Harris L. Tools to study neural and glioma stem cell quiescence. Trends Neurosci 2024; 47:736-748. [PMID: 39191628 DOI: 10.1016/j.tins.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Quiescence is a prolonged but reversible state of cell-cycle arrest that is an adaptive feature of most adult stem cell populations. In the brain, quiescence helps to protect adult neural stem cells from stress and supports lifelong neurogenesis. Unfortunately however, entry into a quiescent or a slow-cycling state is also a malignant feature of brain cancer stem cells. In glioblastoma, where the process has been best characterised, quiescent glioma stem cells preferentially survive chemoradiation, and after therapy, reactivate to regrow the tumour and drive recurrence. In this Review, we discuss the in vitro and in vivo models that have been developed for studying neural stem cell quiescence and how these tools may be used to deepen biological understanding and to develop novel therapies targeting quiescent glioma stem cells.
Collapse
Affiliation(s)
- Dana Friess
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia; The University of Queensland, Brisbane, School of Biomedical Sciences, QLD, 4067, Australia
| | - Stephanie Brauer
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia; Queensland University of Technology, School of Biomedical Sciences, QLD, 4059, Australia
| | - Anni Pöysti
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, WC1E 6DD London, UK
| | - Chandra Choudhury
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia; The University of Queensland, Brisbane, School of Biomedical Sciences, QLD, 4067, Australia
| | - Lachlan Harris
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia; The University of Queensland, Brisbane, School of Biomedical Sciences, QLD, 4067, Australia; Queensland University of Technology, School of Biomedical Sciences, QLD, 4059, Australia.
| |
Collapse
|
2
|
Amodeo V, Davies T, Martinez-Segura A, Clements MP, Ragdale HS, Bailey A, Dos Santos MS, MacRae JI, Mokochinski J, Kramer H, Garcia-Diaz C, Gould AP, Marguerat S, Parrinello S. Diet suppresses glioblastoma initiation in mice by maintaining quiescence of mutation-bearing neural stem cells. Dev Cell 2023; 58:836-846.e6. [PMID: 37084728 PMCID: PMC10618406 DOI: 10.1016/j.devcel.2023.03.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 12/09/2021] [Accepted: 03/28/2023] [Indexed: 04/23/2023]
Abstract
Glioblastoma is thought to originate from neural stem cells (NSCs) of the subventricular zone that acquire genetic alterations. In the adult brain, NSCs are largely quiescent, suggesting that deregulation of quiescence maintenance may be a prerequisite for tumor initiation. Although inactivation of the tumor suppressor p53 is a frequent event in gliomagenesis, whether or how it affects quiescent NSCs (qNSCs) remains unclear. Here, we show that p53 maintains quiescence by inducing fatty-acid oxidation (FAO) and that acute p53 deletion in qNSCs results in their premature activation to a proliferative state. Mechanistically, this occurs through direct transcriptional induction of PPARGC1a, which in turn activates PPARα to upregulate FAO genes. Dietary supplementation with fish oil containing omega-3 fatty acids, natural PPARα ligands, fully restores quiescence of p53-deficient NSCs and delays tumor initiation in a glioblastoma mouse model. Thus, diet can silence glioblastoma driver mutations, with important implications for cancer prevention.
Collapse
Affiliation(s)
- Valeria Amodeo
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | - Timothy Davies
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | - Amalia Martinez-Segura
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Melanie P Clements
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | | | - Andrew Bailey
- The Francis Crick Institute, 1 Midland Road, London NW1 1AA, UK
| | | | - James I MacRae
- The Francis Crick Institute, 1 Midland Road, London NW1 1AA, UK
| | - Joao Mokochinski
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Holger Kramer
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Claudia Garcia-Diaz
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | - Alex P Gould
- The Francis Crick Institute, 1 Midland Road, London NW1 1AA, UK
| | - Samuel Marguerat
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Simona Parrinello
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK.
| |
Collapse
|
3
|
Quaresima S, Istiaq A, Jono H, Cacci E, Ohta K, Lupo G. Assessing the Role of Ependymal and Vascular Cells as Sources of Extracellular Cues Regulating the Mouse Ventricular-Subventricular Zone Neurogenic Niche. Front Cell Dev Biol 2022; 10:845567. [PMID: 35450289 PMCID: PMC9016221 DOI: 10.3389/fcell.2022.845567] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Neurogenesis persists in selected regions of the adult mouse brain; among them, the ventricular-subventricular zone (V-SVZ) of the lateral ventricles represents a major experimental paradigm due to its conspicuous neurogenic output. Postnatal V-SVZ neurogenesis is maintained by a resident population of neural stem cells (NSCs). Although V-SVZ NSCs are largely quiescent, they can be activated to enter the cell cycle, self-renew and generate progeny that gives rise to olfactory bulb interneurons. These adult-born neurons integrate into existing circuits to modify cognitive functions in response to external stimuli, but cells shed by V-SVZ NSCs can also reach injured brain regions, suggesting a latent regenerative potential. The V-SVZ is endowed with a specialized microenvironment, which is essential to maintain the proliferative and neurogenic potential of NSCs, and to preserve the NSC pool from exhaustion by finely tuning their quiescent and active states. Intercellular communication is paramount to the stem cell niche properties of the V-SVZ, and several extracellular signals acting in the niche milieu have been identified. An important part of these signals comes from non-neural cell types, such as local vascular cells, ependymal and glial cells. Understanding the crosstalk between NSCs and other niche components may aid therapeutic approaches for neuropathological conditions, since neurodevelopmental disorders, age-related cognitive decline and neurodegenerative diseases have been associated with dysfunctional neurogenic niches. Here, we review recent advances in the study of the complex interactions between V-SVZ NSCs and their cellular niche. We focus on the extracellular cues produced by ependymal and vascular cells that regulate NSC behavior in the mouse postnatal V-SVZ, and discuss the potential implication of these molecular signals in pathological conditions.
Collapse
Affiliation(s)
- Sabrina Quaresima
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Arif Istiaq
- Department of Stem Cell Biology, Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirofumi Jono
- Department of Pharmacy, Kumamoto University Hospital, Kumamoto, Japan
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Emanuele Cacci
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Kunimasa Ohta
- Department of Stem Cell Biology, Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
- *Correspondence: Kunimasa Ohta, ; Giuseppe Lupo,
| | - Giuseppe Lupo
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- *Correspondence: Kunimasa Ohta, ; Giuseppe Lupo,
| |
Collapse
|
4
|
Ichinose M, Suzuki N, Wang T, Kobayashi H, Vrbanac L, Ng JQ, Wright JA, Lannagan TRM, Gieniec KA, Lewis M, Ando R, Enomoto A, Koblar S, Thomas P, Worthley DL, Woods SL. The BMP antagonist gremlin 1 contributes to the development of cortical excitatory neurons, motor balance and fear responses. Development 2021; 148:269258. [PMID: 34184027 PMCID: PMC8313862 DOI: 10.1242/dev.195883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 06/15/2021] [Indexed: 12/13/2022]
Abstract
Bone morphogenetic protein (BMP) signaling is required for early forebrain development and cortical formation. How the endogenous modulators of BMP signaling regulate the structural and functional maturation of the developing brain remains unclear. Here, we show that expression of the BMP antagonist Grem1 marks committed layer V and VI glutamatergic neurons in the embryonic mouse brain. Lineage tracing of Grem1-expressing cells in the embryonic brain was examined by administration of tamoxifen to pregnant Grem1creERT; Rosa26LSLTdtomato mice at 13.5 days post coitum (dpc), followed by collection of embryos later in gestation. In addition, at 14.5 dpc, bulk mRNA-seq analysis of differentially expressed transcripts between FACS-sorted Grem1-positive and -negative cells was performed. We also generated Emx1-cre-mediated Grem1 conditional knockout mice (Emx1-Cre;Grem1flox/flox) in which the Grem1 gene was deleted specifically in the dorsal telencephalon. Grem1Emx1cKO animals had reduced cortical thickness, especially layers V and VI, and impaired motor balance and fear sensitivity compared with littermate controls. This study has revealed new roles for Grem1 in the structural and functional maturation of the developing cortex. Summary: The BMP antagonist Grem1 is expressed by committed deep-layer glutamatergic neurons in the embryonic mouse cortex. Grem1 conditional knockout mice display cortical and behavioral abnormalities.
Collapse
Affiliation(s)
- Mari Ichinose
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Nobumi Suzuki
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Tongtong Wang
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Hiroki Kobayashi
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia.,Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Laura Vrbanac
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Jia Q Ng
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Josephine A Wright
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Tamsin R M Lannagan
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Krystyna A Gieniec
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Martin Lewis
- Department of Psychiatry, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5001, Australia.,Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Ryota Ando
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Simon Koblar
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Paul Thomas
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Daniel L Worthley
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Susan L Woods
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, SA 5000, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| |
Collapse
|
5
|
Li KN, Tumbar T. Hair follicle stem cells as a skin-organizing signaling center during adult homeostasis. EMBO J 2021; 40:e107135. [PMID: 33880808 PMCID: PMC8167365 DOI: 10.15252/embj.2020107135] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/16/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Stem cells are the essential source of building blocks for tissue homeostasis and regeneration. Their behavior is dictated by both cell-intrinsic cues and extrinsic cues from the microenvironment, known as the stem cell niche. Interestingly, recent work began to demonstrate that hair follicle stem cells (HFSCs) are not only passive recipients of signals from the surroundings, but also actively send out signals to modulate the organization and function of their own niches. Here, we discuss recent findings, and briefly refer to the old, on the interaction of HFSCs and their niches with the emphasis on the outwards signals from HFSCs toward their niches. We also highlight recent technology advancements that further promote our understanding of HFSC niches. Taken together, the HFSCs emerge as a skin-organizing center rich in signaling output for niche remodeling during various stages of adult skin homeostasis. The intricate crosstalk between HFSCs and their niches adds important insight to skin biology that will inform clinical and bioengineering fields aiming to build complete and functional 3D organotypic cultures for skin replacement therapies.
Collapse
Affiliation(s)
- Kefei Nina Li
- Molecular Biology and GeneticsCornell UniversityIthacaNYUSA
| | | |
Collapse
|
6
|
Winkelman MA, Koppes AN, Koppes RA, Dai G. Bioengineering the neurovascular niche to study the interaction of neural stem cells and endothelial cells. APL Bioeng 2021; 5:011507. [PMID: 33688617 PMCID: PMC7932757 DOI: 10.1063/5.0027211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
The ability of mammalian neural stem cells (NSCs) to self-renew and differentiate throughout adulthood has made them ideal to study neurogenesis and attractive candidates for neurodegenerative disease therapies. In the adult mammalian brain, NSCs are maintained in the neurovascular niche (NVN) where they are found near the specialized blood vessels, suggesting that brain endothelial cells (BECs) are prominent orchestrators of NSC fate. However, most of the current knowledge of the mammalian NVN has been deduced from nonhuman studies. To circumvent the challenges of in vivo studies, in vitro models have been developed to better understand the reciprocal cellular mechanisms of human NSCs and BECs. This review will cover the current understanding of mammalian NVN biology, the effects of endothelial cell-derived signals on NSC fate, and the in vitro models developed to study the interactions between NSCs and BECs.
Collapse
Affiliation(s)
- Max A Winkelman
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| | | | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| |
Collapse
|
7
|
Omiya H, Yamaguchi S, Watanabe T, Kuniya T, Harada Y, Kawaguchi D, Gotoh Y. BMP signaling suppresses Gemc1 expression and ependymal differentiation of mouse telencephalic progenitors. Sci Rep 2021; 11:613. [PMID: 33436697 PMCID: PMC7804439 DOI: 10.1038/s41598-020-79610-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 11/18/2020] [Indexed: 01/29/2023] Open
Abstract
The lateral ventricles of the adult mammalian brain are lined by a single layer of multiciliated ependymal cells, which generate a flow of cerebrospinal fluid through directional beating of their cilia as well as regulate neurogenesis through interaction with adult neural stem cells. Ependymal cells are derived from a subset of embryonic neural stem-progenitor cells (NPCs, also known as radial glial cells) that becomes postmitotic during the late embryonic stage of development. Members of the Geminin family of transcriptional regulators including GemC1 and Mcidas play key roles in the differentiation of ependymal cells, but it remains largely unclear what extracellular signals regulate these factors and ependymal differentiation during embryonic and early-postnatal development. We now show that the levels of Smad1/5/8 phosphorylation and Id1/4 protein expression-both of which are downstream events of bone morphogenetic protein (BMP) signaling-decline in cells of the ventricular-subventricular zone in the mouse lateral ganglionic eminence in association with ependymal differentiation. Exposure of postnatal NPC cultures to BMP ligands or to a BMP receptor inhibitor suppressed and promoted the emergence of multiciliated ependymal cells, respectively. Moreover, treatment of embryonic NPC cultures with BMP ligands reduced the expression level of the ependymal marker Foxj1 and suppressed the emergence of ependymal-like cells. Finally, BMP ligands reduced the expression levels of Gemc1 and Mcidas in postnatal NPC cultures, whereas the BMP receptor inhibitor increased them. Our results thus implicate BMP signaling in suppression of ependymal differentiation from NPCs through regulation of Gemc1 and Mcidas expression during embryonic and early-postnatal stages of mouse telencephalic development.
Collapse
Affiliation(s)
- Hanae Omiya
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Shima Yamaguchi
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Tomoyuki Watanabe
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Takaaki Kuniya
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Yujin Harada
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Daichi Kawaguchi
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Yukiko Gotoh
- grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan ,grid.26999.3d0000 0001 2151 536XInternational Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| |
Collapse
|
8
|
Abstract
All organisms growing beyond the oxygen diffusion limit critically depend on a functional vasculature for survival. Yet blood vessels are far more than passive, uniform conduits for oxygen and nutrient supply. A remarkable organotypic heterogeneity is brought about by tissue-specific differentiated endothelial cells (lining the blood vessels' lumen) and allows blood vessels to deal with organ-specific demands for homeostasis. On the flip side, when blood vessels go awry, they promote life-threatening diseases characterized by endothelial cells inappropriately adopting an angiogenic state (eg, tumor vascularization) or becoming dysfunctional (eg, diabetic microvasculopathies), calling respectively for antiangiogenic therapies and proangiogenic/vascular regenerative strategies. In solid tumors, despite initial enthusiasm, growth factor-based (mostly anti-VEGF [vascular endothelial growth factor]) antiangiogenic therapies do not sufficiently live up to the expectations in terms of efficiency and patient survival, in part, due to intrinsic and acquired therapy resistance. Tumors cunningly deploy alternative growth factors than the ones targeted by the antiangiogenic therapies to reinstigate angiogenesis or revert to other ways of securing blood flow, independently of the targeted growth factors. In trying to alleviate tissue ischemia and to repair dysfunctional or damaged endothelium, local in-tissue administration of (genes encoding) proangiogenic factors or endothelial (stem) cells harnessing regenerative potential have been explored. Notwithstanding evaluation in clinical trials, these approaches are often hampered by dosing issues and limited half-life or local retention of the administered agents. Here, without intending to provide an all-encompassing historical overview, we focus on some recent advances in understanding endothelial cell behavior in health and disease and identify novel molecular players and concepts that could eventually be considered for therapeutic targeting.
Collapse
Affiliation(s)
- Guy Eelen
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.)
| | - Lucas Treps
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.)
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China (X.L., P.C.)
| | - Peter Carmeliet
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.).,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China (X.L., P.C.)
| |
Collapse
|
9
|
Pasquier J, Ghiabi P, Chouchane L, Razzouk K, Rafii S, Rafii A. Angiocrine endothelium: from physiology to cancer. J Transl Med 2020; 18:52. [PMID: 32014047 PMCID: PMC6998193 DOI: 10.1186/s12967-020-02244-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/28/2020] [Indexed: 02/08/2023] Open
Abstract
The concept of cancer as a cell-autonomous disease has been challenged by the wealth of knowledge gathered in the past decades on the importance of tumor microenvironment (TM) in cancer progression and metastasis. The significance of endothelial cells (ECs) in this scenario was initially attributed to their role in vasculogenesis and angiogenesis that is critical for tumor initiation and growth. Nevertheless, the identification of endothelial-derived angiocrine factors illustrated an alternative non-angiogenic function of ECs contributing to both physiological and pathological tissue development. Gene expression profiling studies have demonstrated distinctive expression patterns in tumor-associated endothelial cells that imply a bilateral crosstalk between tumor and its endothelium. Recently, some of the molecular determinants of this reciprocal interaction have been identified which are considered as potential targets for developing novel anti-angiocrine therapeutic strategies.
Collapse
Affiliation(s)
- Jennifer Pasquier
- Nice Breast Institute, 57 bld de la Californie, 06000, Nice, France.
- Stem Cell & Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar.
| | - Pegah Ghiabi
- Stem Cell & Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Lotfi Chouchane
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, 10065, USA
- Laboratory of Genetic Medicine and Immunology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Kais Razzouk
- Nice Breast Institute, 57 bld de la Californie, 06000, Nice, France
| | - Shahin Rafii
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Arash Rafii
- Nice Breast Institute, 57 bld de la Californie, 06000, Nice, France
- Stem Cell & Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| |
Collapse
|
10
|
Distinct Molecular Signatures of Quiescent and Activated Adult Neural Stem Cells Reveal Specific Interactions with Their Microenvironment. Stem Cell Reports 2018; 11:565-577. [PMID: 29983386 PMCID: PMC6092681 DOI: 10.1016/j.stemcr.2018.06.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 06/06/2018] [Accepted: 06/06/2018] [Indexed: 12/31/2022] Open
Abstract
Deciphering the mechanisms that regulate the quiescence of adult neural stem cells (NSCs) is crucial for the development of therapeutic strategies based on the stimulation of their endogenous regenerative potential in the damaged brain. We show that LeXbright cells sorted from the adult mouse subventricular zone exhibit all the characteristic features of quiescent NSCs. Indeed, they constitute a subpopulation of slowly dividing cells that is able to enter the cell cycle to regenerate the irradiated niche. Comparative transcriptomic analyses showed that they express hallmarks of NSCs but display a distinct molecular signature from activated NSCs (LeX+EGFR+ cells). Particularly, numerous membrane receptors are expressed on quiescent NSCs. We further revealed a different expression pattern of Syndecan-1 between quiescent and activated NSCs and demonstrated its role in the proliferation of activated NSCs. Our data highlight the central role of the stem cell microenvironment in the regulation of quiescence in adult neurogenic niches. Transcriptome analysis reveals molecular hallmarks of activated and quiescent NSCs Data resource of putative markers and/or regulators of NSC quiescence Quiescent NSCs integrate various signals from the microenvironment Syndecan-1 is involved in proliferation of NSCs
Collapse
|
11
|
Crouch EE, Doetsch F. FACS isolation of endothelial cells and pericytes from mouse brain microregions. Nat Protoc 2018; 13:738-751. [PMID: 29565899 DOI: 10.1038/nprot.2017.158] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The vasculature is emerging as a key contributor to brain function during neurodevelopment and in mature physiological and pathological states. The brain vasculature itself also exhibits regional heterogeneity, highlighting the need to develop approaches for purifying cells from different microregions. Previous approaches for isolation of endothelial cells and pericytes have predominantly required transgenic mice and large amounts of tissue, and have resulted in impure populations. In addition, the prospective purification of brain pericytes has been complicated by the fact that widely used pericyte markers are also expressed by other cell types in the brain. Here, we describe the detailed procedures for simultaneous isolation of pure populations of endothelial cells and pericytes directly from adult mouse brain microregions using fluorescence-activated cell sorting (FACS) with antibodies against CD31 (endothelial cells) and CD13 (pericytes). This protocol is scalable and takes ∼5 h, including microdissection of the region of interest, enzymatic tissue dissociation, immunostaining, and FACS. This protocol allows the isolation of brain vascular cells from any mouse strain under diverse conditions; these cells can be used for multiple downstream applications, including in vitro and in vivo experiments, and transcriptomic, proteomic, metabolomic, epigenomic, and single-cell analysis.
Collapse
Affiliation(s)
- Elizabeth E Crouch
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
12
|
Lebaschi A, Nakagawa Y, Wada S, Cong GT, Rodeo SA. Tissue-specific endothelial cells: a promising approach for augmentation of soft tissue repair in orthopedics. Ann N Y Acad Sci 2018; 1410:44-56. [PMID: 29265420 DOI: 10.1111/nyas.13575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/12/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022]
Abstract
Biologics are playing an increasingly significant role in the practice of modern medicine and surgery in general and orthopedics in particular. Cell-based approaches are among the most important and widely used modalities in orthopedic biologics, with mesenchymal stem cells and other multi/pluripotent cells undergoing evaluation in numerous preclinical and clinical studies. On the other hand, fully differentiated endothelial cells (ECs) have been found to perform critical roles in homeostasis of visceral tissues through production of an adaptive panel of so-called "angiocrine factors." This newly discovered function of ECs renders them excellent candidates for novel approaches in cell-based biologics. Here, we present a review of the role of ECs and angiocrine factors in some visceral tissues, followed by an overview of current cell-based approaches and a discussion of the potential applications of ECs in soft tissue repair.
Collapse
Affiliation(s)
- Amir Lebaschi
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Yusuke Nakagawa
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Susumu Wada
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Guang-Ting Cong
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Scott A Rodeo
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York.,Sports Medicine and Shoulder Service, Hospital for Special Surgery, New York, New York
| |
Collapse
|
13
|
Wen Y, Yang H, Liu Y, Liu Q, Wang A, Ding Y, Jin Z. Evaluation of BMMSCs-EPCs sheets for repairing alveolar bone defects in ovariectomized rats. Sci Rep 2017; 7:16568. [PMID: 29185450 PMCID: PMC5707386 DOI: 10.1038/s41598-017-16404-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/13/2017] [Indexed: 01/14/2023] Open
Abstract
The aim of this paper is to investigate the effect that bone marrow mesenchymal stem cells (BMMSCs) - endothelial progenitor cells (EPCs), BMMSCs and EPCs sheets have on repairing maxillary alveolar defects in ovariectomized (OVX) rats. In this study, after identification using multi-lineage differentiation and flow cytometry, BMMSCs and EPCs were isolated from female rats. The BMMSCs-EPCs, BMMSCs and EPCs sheets were detected by hematoxylin-eosin (H&E) staining, alkaline phosphatase (ALP) staining and qRT-PCR. Defects were created in maxillary alveoli and repaired with BMMSCs-EPCs, BMMSCs and EPCs sheets in OVX rats. The repair effects were determined by histological staining and micro-CT analysis at 2, 4 and 8 weeks after implantation. We aim to clarify whether BMMSCs-EPCs sheets are more effective in repairing alveolar bone defects than are BMMSCs and EPCs sheets in OVX rats. The results show that the osteogenic potential and the effect of bone repair are greater in the BMMSCs-EPCs sheet group and that this group has a higher ability to repair alveolar bone defects in OVX rats. These results suggest that BMMSCs-EPCs sheets have potential in clinical applications for treating humans with osteoporosis.
Collapse
Affiliation(s)
- Yi Wen
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, 145 Changle West Road, Xi'an, 710032, China
| | - Hongxu Yang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, the Fourth Military Medical University, 145 Changle West Road, Xi'an, 710032, China
| | - Yanli Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and Emergency, School of Stomatology, the Fourth Military Medical University, 145 Changle West Road, Xi'an, 710032, China
| | - Qian Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, 145 Changle West Road, Xi'an, 710032, China
| | - Axian Wang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, 145 Changle West Road, Xi'an, 710032, China
| | - Yin Ding
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, 145 Changle West Road, Xi'an, 710032, China
| | - Zuolin Jin
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, 145 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
14
|
Azizoglu DB, Cleaver O. Blood vessel crosstalk during organogenesis-focus on pancreas and endothelial cells. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 5:598-617. [PMID: 27328421 DOI: 10.1002/wdev.240] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/23/2016] [Accepted: 04/16/2016] [Indexed: 01/02/2023]
Abstract
Blood vessels form a highly branched, interconnected, and largely stereotyped network of tubes that sustains every organ and tissue in vertebrates. How vessels come to take on their particular architecture, or how they are 'patterned,' and in turn, how they influence surrounding tissues are fundamental questions of organogenesis. Decades of work have begun to elucidate how endothelial progenitors arise and home to precise locations within tissues, integrating attractive and repulsive cues to build vessels where they are needed. Conversely, more recent findings have revealed an exciting facet of blood vessel interaction with tissues, where vascular cells provide signals to developing organs and progenitors therein. Here, we discuss the exchange of reciprocal signals between endothelial cells and neighboring tissues during embryogenesis, with a special focus on the developing pancreas. Understanding the mechanisms driving both sides of these interactions will be crucial to the development of therapies, from improving organ regeneration to efficient production of cell based therapies. Specifically, elucidating the interface of the vasculature with pancreatic lineages, including endocrine cells, will instruct approaches such as generation of replacement beta cells for Type I diabetes. WIREs Dev Biol 2016, 5:598-617. doi: 10.1002/wdev.240 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- D Berfin Azizoglu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
15
|
The Neurogenic Potential of Astrocytes Is Regulated by Inflammatory Signals. Mol Neurobiol 2015; 53:3724-3739. [PMID: 26138449 PMCID: PMC4937102 DOI: 10.1007/s12035-015-9296-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023]
Abstract
Although the adult brain contains neural stem cells (NSCs) that generate new neurons throughout life, these astrocyte-like populations are restricted to two discrete niches. Despite their terminally differentiated phenotype, adult parenchymal astrocytes can re-acquire NSC-like characteristics following injury, and as such, these ‘reactive’ astrocytes offer an alternative source of cells for central nervous system (CNS) repair following injury or disease. At present, the mechanisms that regulate the potential of different types of astrocytes are poorly understood. We used in vitro and ex vivo astrocytes to identify candidate pathways important for regulation of astrocyte potential. Using in vitro neural progenitor cell (NPC)-derived astrocytes, we found that exposure of more lineage-restricted astrocytes to either tumor necrosis factor alpha (TNF-α) (via nuclear factor-κB (NFκB)) or the bone morphogenetic protein (BMP) inhibitor, noggin, led to re-acquisition of NPC properties accompanied by transcriptomic and epigenetic changes consistent with a more neurogenic, NPC-like state. Comparative analyses of microarray data from in vitro-derived and ex vivo postnatal parenchymal astrocytes identified several common pathways and upstream regulators associated with inflammation (including transforming growth factor (TGF)-β1 and peroxisome proliferator-activated receptor gamma (PPARγ)) and cell cycle control (including TP53) as candidate regulators of astrocyte phenotype and potential. We propose that inflammatory signalling may control the normal, progressive restriction in potential of differentiating astrocytes as well as under reactive conditions and represent future targets for therapies to harness the latent neurogenic capacity of parenchymal astrocytes.
Collapse
|
16
|
Regional and stage-specific effects of prospectively purified vascular cells on the adult V-SVZ neural stem cell lineage. J Neurosci 2015; 35:4528-39. [PMID: 25788671 DOI: 10.1523/jneurosci.1188-14.2015] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Adult neural stem cells reside in specialized niches. In the ventricular-subventricular zone (V-SVZ), quiescent neural stem cells (qNSCs) become activated (aNSCs), and generate transit amplifying cells (TACs), which give rise to neuroblasts that migrate to the olfactory bulb. The vasculature is an important component of the adult neural stem cell niche, but whether vascular cells in neurogenic areas are intrinsically different from those elsewhere in the brain is unknown. Moreover, the contribution of pericytes to the neural stem cell niche has not been defined. Here, we describe a rapid FACS purification strategy to simultaneously isolate primary endothelial cells and pericytes from brain microregions of nontransgenic mice using CD31 and CD13 as surface markers. We compared the effect of purified vascular cells from a neurogenic (V-SVZ) and non-neurogenic brain region (cortex) on the V-SVZ stem cell lineage in vitro. Endothelial and pericyte diffusible signals from both regions differentially promote the proliferation and neuronal differentiation of qNSCs, aNSCs, and TACs. Unexpectedly, diffusible cortical signals had the most potent effects on V-SVZ proliferation and neurogenesis, highlighting the intrinsic capacity of non-neurogenic vasculature to support stem cell behavior. Finally, we identify PlGF-2 as an endothelial-derived mitogen that promotes V-SVZ cell proliferation. This purification strategy provides a platform to define the functional and molecular contribution of vascular cells to stem cell niches and other brain regions under different physiological and pathological states.
Collapse
|
17
|
Morell M, Tsan YC, O'Shea KS. Inducible expression of noggin selectively expands neural progenitors in the adult SVZ. Stem Cell Res 2015; 14:79-94. [DOI: 10.1016/j.scr.2014.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 10/09/2014] [Accepted: 11/06/2014] [Indexed: 12/29/2022] Open
|
18
|
Abstract
Previous studies have shown that the Hippo pathway effector yes-associated protein (YAP) plays an important role in maintaining stem cell proliferation. However, the precise molecular mechanism of YAP in regulating murine embryonic neural stem cells (NSCs) remains largely unknown. Here, we show that bone morphogenetic protein-2 (BMP2) treatment inhibited the proliferation of mouse embryonic NSCs, that YAP was critical for mouse NSC proliferation, and that BMP2 treatment-induced inhibition of mouse NSC proliferation was abrogated by YAP knockdown, indicating that the YAP protein mediates the inhibitory effect of BMP2 signaling. Additionally, we found that BMP2 treatment reduced YAP nuclear translocation, YAP-TEAD interaction, and YAP-mediated transactivation. BMP2 treatment inhibited YAP/TEAD-mediated Cyclin D1 (ccnd1) expression, and knockdown of ccnd1 abrogated the BMP2-mediated inhibition of mouse NSC proliferation. Mechanistically, we found that Smad1/4, effectors of BMP2 signaling, competed with YAP for the interaction with TAED1 and inhibited YAP's cotranscriptional activity. Our data reveal mechanistic cross talk between BMP2 signaling and the Hippo-YAP pathway in murine NSC proliferation, which may be exploited as a therapeutic target in neurodegenerative diseases and aging.
Collapse
|
19
|
Ottone C, Krusche B, Whitby A, Clements M, Quadrato G, Pitulescu ME, Adams RH, Parrinello S. Direct cell-cell contact with the vascular niche maintains quiescent neural stem cells. Nat Cell Biol 2014; 16:1045-56. [PMID: 25283993 PMCID: PMC4298702 DOI: 10.1038/ncb3045] [Citation(s) in RCA: 258] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/28/2014] [Indexed: 02/07/2023]
Abstract
The vasculature is a prominent component of the subventricular zone neural stem cell niche. Although quiescent neural stem cells physically contact blood vessels at specialised endfeet, the significance of this interaction is not understood. In contrast, it is well established that vasculature-secreted soluble factors promote lineage progression of committed progenitors. Here we specifically investigated the role of cell-cell contact-dependent signalling in the vascular niche. Unexpectedly, we find that direct cell-cell interactions with endothelial cells enforces quiescence and promotes stem cell identity. Mechanistically, endothelial ephrinB2 and Jagged1 mediate these effects by suppressing cell-cycle entry downstream of mitogens and inducing stemness genes to jointly inhibit differentiation. In vivo, endothelial-specific ablation of either of the genes which encode these proteins, Efnb2 and Jag1 respectively, aberrantly activates quiescent stem cells, resulting in depletion. Thus, we identify the vasculature as a critical niche compartment for stem cell maintenance, furthering our understanding of how anchorage to the niche maintains stem cells within a pro-differentiative microenvironment.
Collapse
Affiliation(s)
- Cristina Ottone
- Cell Interactions and Cancer Group, MRC Clinical Sciences Centre, Imperial College London, Du Cane Road London W12 0NN, UK
| | - Benjamin Krusche
- Cell Interactions and Cancer Group, MRC Clinical Sciences Centre, Imperial College London, Du Cane Road London W12 0NN, UK
| | - Ariadne Whitby
- Cell Interactions and Cancer Group, MRC Clinical Sciences Centre, Imperial College London, Du Cane Road London W12 0NN, UK
| | - Melanie Clements
- Cell Interactions and Cancer Group, MRC Clinical Sciences Centre, Imperial College London, Du Cane Road London W12 0NN, UK
| | - Giorgia Quadrato
- Hertie Institute for Brain Research, University of Tuebingen, Tuebingen D-72076, Germany
| | - Mara E Pitulescu
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and Faculty of Medicine, University of Muenster, Muenster D-48149, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and Faculty of Medicine, University of Muenster, Muenster D-48149, Germany
| | - Simona Parrinello
- Cell Interactions and Cancer Group, MRC Clinical Sciences Centre, Imperial College London, Du Cane Road London W12 0NN, UK
| |
Collapse
|
20
|
Abstract
An important hallmark of many adult stem cell niches is their proximity to the vasculature in vivo, a feature common to neural stem cells (NSCs), mesenchymal stem cells (MSCs) from bone marrow, adipose, and other tissues, hematopoietic stem cells (HSCs), and many tumor stem cells. This review summarizes key studies supporting the vasculature's instructive role in adult stem cell niches, and the putative underlying molecular mechanisms by which blood vessels in these niches exert control over progenitor cell fates. The importance of the perivascular niche for pathology, notably tumor metastasis and dormancy, is also highlighted. Finally, the implications of the perivascular regulation of stem and progenitor cells on biomaterial design and the impact on future research directions are discussed.
Collapse
Affiliation(s)
- Andrew J Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
21
|
Role of angiogenesis in bone repair. Arch Biochem Biophys 2014; 561:109-17. [PMID: 25034215 DOI: 10.1016/j.abb.2014.07.006] [Citation(s) in RCA: 262] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 07/01/2014] [Accepted: 07/08/2014] [Indexed: 12/25/2022]
Abstract
Bone vasculature plays a vital role in bone development, remodeling and homeostasis. New blood vessel formation is crucial during both primary bone development as well as fracture repair in adults. Both bone repair and bone remodeling involve the activation and complex interaction between angiogenic and osteogenic pathways. Interestingly studies have demonstrated that angiogenesis precedes the onset of osteogenesis. Indeed reduced or inadequate blood flow has been linked to impaired fracture healing and old age related low bone mass disorders such as osteoporosis. Similarly the slow penetration of host blood vessels in large engineered bone tissue grafts has been cited as one of the major hurdle still impeding current bone construction engineering strategies. This article reviews the current knowledge elaborating the importance of vascularization during bone healing and remodeling, and the current therapeutic strategies being adapted to promote and improve angiogenesis.
Collapse
|
22
|
Haus DL, Nguyen HX, Gold EM, Kamei N, Perez H, Moore HD, Anderson AJ, Cummings BJ. CD133-enriched Xeno-Free human embryonic-derived neural stem cells expand rapidly in culture and do not form teratomas in immunodeficient mice. Stem Cell Res 2014; 13:214-26. [PMID: 25082219 PMCID: PMC5675021 DOI: 10.1016/j.scr.2014.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 06/14/2014] [Accepted: 06/30/2014] [Indexed: 12/17/2022] Open
Abstract
Common methods for the generation of human embryonic-derived neural stem cells (hNSCs) result in cells with potentially compromised safety profiles due to maintenance of cells in conditions containing non-human proteins (e.g. in bovine serum or on mouse fibroblast feeders). Additionally, sufficient expansion of resulting hNSCs for scaling out or up in a clinically relevant time frame has proven to be difficult. Here, we report a strategy that produces hNSCs in completely “Xeno-Free” culture conditions. Furthermore, we have enriched the hNSCs for the cell surface marker CD133 via magnetic sorting, which has led to an increase in the expansion rate and neuronal fate specification of the hNSCs in vitro. Critically, we have also confirmed neural lineage specificity upon sorted hNSC transplantation into the immunodeficient NOD-scid mouse brain. The future use or adaptation of these protocols has the potential to better facilitate the advancement of pre-clinical strategies from the bench to the bedside.
Collapse
Affiliation(s)
- Daniel L Haus
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine, CA 92697-1750, USA
| | - Hal X Nguyen
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, CA 92697-1750, USA; UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697-1750, USA
| | - Eric M Gold
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine, CA 92697-1750, USA
| | - Noriko Kamei
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, CA 92697-1750, USA; UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697-1750, USA
| | - Harvey Perez
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, CA 92697-1750, USA; UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697-1750, USA
| | - Harry D Moore
- Centre for Stem Cell Biology, University of Sheffield, Sheffield S10 2TN, UK; Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Aileen J Anderson
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, CA 92697-1750, USA; Physical and Medical Rehabilitation, University of California, Irvine, CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine, CA 92697-1750, USA; UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697-1750, USA
| | - Brian J Cummings
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, CA 92697-1750, USA; Physical and Medical Rehabilitation, University of California, Irvine, CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine, CA 92697-1750, USA; UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697-1750, USA.
| |
Collapse
|
23
|
Agis-Balboa RC, Fischer A. Generating new neurons to circumvent your fears: the role of IGF signaling. Cell Mol Life Sci 2014; 71:21-42. [PMID: 23543251 PMCID: PMC11113432 DOI: 10.1007/s00018-013-1316-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 02/12/2013] [Accepted: 03/04/2013] [Indexed: 12/13/2022]
Abstract
Extinction of fear memory is a particular form of cognitive function that is of special interest because of its involvement in the treatment of anxiety and mood disorders. Based on recent literature and our previous findings (EMBO J 30(19):4071-4083, 2011), we propose a new hypothesis that implies a tight relationship among IGF signaling, adult hippocampal neurogenesis and fear extinction. Our proposed model suggests that fear extinction-induced IGF2/IGFBP7 signaling promotes the survival of neurons at 2-4 weeks old that would participate in the discrimination between the original fear memory trace and the new safety memory generated during fear extinction. This is also called "pattern separation", or the ability to distinguish similar but different cues (e.g., context). To understand the molecular mechanisms underlying fear extinction is therefore of great clinical importance.
Collapse
Affiliation(s)
- R C Agis-Balboa
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Grisebach Str. 5, 37077, Göttingen, Germany,
| | | |
Collapse
|
24
|
Adult neural stem cells and their niche: a dynamic duo during homeostasis, regeneration, and aging. Curr Opin Neurobiol 2013; 23:935-42. [PMID: 24090877 DOI: 10.1016/j.conb.2013.09.004] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 09/03/2013] [Indexed: 12/27/2022]
Abstract
Stem cells persist in specialized niches in the adult mammalian brain. Emerging findings highlight the complexity and heterogeneity of different compartments in the niche, as well as the presence of local signaling microdomains. Stem cell quiescence and activation are regulated not only by anchorage to the niche and diffusible signals, but also by biophysical properties, including fluid dynamics. Importantly, the adult neural stem cell niche integrates both local and systemic changes, reflecting the physiological state of the organism. Moreover niche signaling is bidirectional, with stem cells and their progeny and niche cells dynamically interacting with each other during homeostasis, regeneration and aging.
Collapse
|
25
|
Hegarty SV, O'Keeffe GW, Sullivan AM. BMP-Smad 1/5/8 signalling in the development of the nervous system. Prog Neurobiol 2013; 109:28-41. [PMID: 23891815 DOI: 10.1016/j.pneurobio.2013.07.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 07/16/2013] [Accepted: 07/16/2013] [Indexed: 02/07/2023]
Abstract
The transcription factors, Smad1, Smad5 and Smad8, are the pivotal intracellular effectors of the bone morphogenetic protein (BMP) family of proteins. BMPs and their receptors are expressed in the nervous system (NS) throughout its development. This review focuses on the actions of Smad 1/5/8 in the developing NS. The mechanisms by which these Smad proteins regulate the induction of the neuroectoderm, the central nervous system (CNS) primordium, and finally the neural crest, which gives rise to the peripheral nervous system (PNS), are reviewed herein. We describe how, following neural tube closure, the most dorsal aspect of the tube becomes a signalling centre for BMPs, which directs the pattern of the development of the dorsal spinal cord (SC), through the action of Smad1, Smad5 and Smad8. The direct effects of Smad 1/5/8 signalling on the development of neuronal and non-neuronal cells from various neural progenitor cell populations are then described. Finally, this review discusses the neurodevelopmental abnormalities associated with the knockdown of Smad 1/5/8.
Collapse
Affiliation(s)
- Shane V Hegarty
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | | | | |
Collapse
|
26
|
Pineda JR, Daynac M, Chicheportiche A, Cebrian-Silla A, Sii Felice K, Garcia-Verdugo JM, Boussin FD, Mouthon MA. Vascular-derived TGF-β increases in the stem cell niche and perturbs neurogenesis during aging and following irradiation in the adult mouse brain. EMBO Mol Med 2013; 5:548-62. [PMID: 23526803 PMCID: PMC3628106 DOI: 10.1002/emmm.201202197] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/18/2012] [Accepted: 12/20/2012] [Indexed: 01/20/2023] Open
Abstract
Neurogenesis decreases during aging and following cranial radiotherapy, causing a progressive cognitive decline that is currently untreatable. However, functional neural stem cells remained present in the subventricular zone of high dose-irradiated and aged mouse brains. We therefore investigated whether alterations in the neurogenic niches are perhaps responsible for the neurogenesis decline. This hypothesis was supported by the absence of proliferation of neural stem cells that were engrafted into the vascular niches of irradiated host brains. Moreover, we observed a marked increase in TGF-β1 production by endothelial cells in the stem cell niche in both middle-aged and irradiated mice. In co-cultures, irradiated brain endothelial cells induced the apoptosis of neural stem/progenitor cells via TGF-β/Smad3 signalling. Strikingly, the blockade of TGF-β signalling in vivo using a neutralizing antibody or the selective inhibitor SB-505124 significantly improved neurogenesis in aged and irradiated mice, prevented apoptosis and increased the proliferation of neural stem/progenitor cells. These findings suggest that anti-TGF-β-based therapy may be used for future interventions to prevent neurogenic collapse following radiotherapy or during aging.
Collapse
Affiliation(s)
- Jose R Pineda
- CEA DSV iRCM SCSR, Laboratoire de Radiopathologie, Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Chen J, Sun W, Zheng Y, Xiong H, Cai Y. Bone morphogenetic protein 4, inhibitor of differentiation 1, and epidermal growth factor receptor regulate the survival of cochlear sensory epithelial cells. J Neurosci Res 2013; 91:515-26. [DOI: 10.1002/jnr.23175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 09/06/2012] [Accepted: 10/13/2012] [Indexed: 12/11/2022]
|
28
|
Porlan E, Perez-Villalba A, Delgado AC, Ferrón SR. Paracrine regulation of neural stem cells in the subependymal zone. Arch Biochem Biophys 2012; 534:11-9. [PMID: 23073070 DOI: 10.1016/j.abb.2012.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 09/27/2012] [Accepted: 10/05/2012] [Indexed: 12/31/2022]
Abstract
Stem cells maintain their self-renewal and multipotency capacities through a self-organizing network of transcription factors and intracellular pathways activated by extracellular signaling from the microenvironment or "niche" in which they reside in vivo. In the adult mammalian brain new neurons continue to be generated throughout life of the organisms and this lifelong process of neurogenesis is supported by a reservoir of neural stem cells in the germinal regions. The discovery of adult neurogenesis in the mammalian brain has sparked great interest in defining the conditions that guide neural stem cell (NSC) maintenance and differentiation into the great variety of neuronal and glial subtypes. Here we review current knowledge regarding the paracrine regulation provided by the components of the niche and its function, focusing on the main germinal region of the adult central nervous system (CNS), the subependymal zone (SEZ).
Collapse
Affiliation(s)
- Eva Porlan
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Spain
| | | | | | | |
Collapse
|
29
|
Gene expression profiling of neural stem cells and their neuronal progeny reveals IGF2 as a regulator of adult hippocampal neurogenesis. J Neurosci 2012; 32:3376-87. [PMID: 22399759 DOI: 10.1523/jneurosci.4248-11.2012] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neural stem cells (NSCs) generate neurons throughout life in the hippocampal dentate gyrus (DG). How gene expression signatures differ among NSCs and immature neurons remains largely unknown. We isolated NSCs and their progeny in the adult DG using transgenic mice expressing a GFP reporter under the control of the Sox2 promoter (labeling NSCs) and transgenic mice expressing a DsRed reporter under the control of the doublecortin (DCX) promoter (labeling immature neurons). Transcriptome analyses revealed distinct gene expression profiles between NSCs and immature neurons. Among the genes that were expressed at significantly higher levels in DG NSCs than in immature neurons was the growth factor insulin-like growth factor 2 (IGF2). We show that IGF2 selectively controls proliferation of DG NSCs in vitro and in vivo through AKT-dependent signaling. Thus, by gene expression profiling of NSCs and their progeny, we have identified IGF2 as a novel regulator of adult neurogenesis.
Collapse
|
30
|
Wang YZ, Plane JM, Jiang P, Zhou CJ, Deng W. Concise review: Quiescent and active states of endogenous adult neural stem cells: identification and characterization. Stem Cells 2011; 29:907-12. [PMID: 21557389 DOI: 10.1002/stem.644] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The adult mammalian central nervous system (CNS) lacks the capacity for regeneration, making it a highly sought-after topic for researchers. The identification of neural stem cells (NSCs) in the adult CNS wiped out a long-held dogma that the adult brain contains a set number of neurons and is incapable of replacing them. The discovery of adult NSCs (aNSCs) stoked the fire for researchers who dream of brain self-repair. Unfortunately, the quiescent nature and limited plasticity of aNSCs diminish their regenerative potential. Recent studies evaluating aNSC plasticity under pathological conditions indicate that a switch from quiescent to active aNSCs in neurogenic regions plays an important role in both repairing the damaged tissue and preserving progenitor pools. Here, we summarize the most recent findings and present questions about characterizing the active and quiescent aNSCs in major neurogenic regions, and factors for maintaining their active and quiescent states, hoping to outline an emerging view for promoting the endogenous aNSC-based regeneration.
Collapse
Affiliation(s)
- Ya-Zhou Wang
- Cellular and Molecular Biology Laboratory, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | | | | | | | | |
Collapse
|
31
|
DLK1 Promotes Neurogenesis of Human and Mouse Pluripotent Stem Cell-Derived Neural Progenitors Via Modulating Notch and BMP Signalling. Stem Cell Rev Rep 2011; 8:459-71. [DOI: 10.1007/s12015-011-9298-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
32
|
Markiewicz I, Sypecka J, Domanska-Janik K, Wyszomirski T, Lukomska B. Cellular environment directs differentiation of human umbilical cord blood-derived neural stem cells in vitro. J Histochem Cytochem 2011; 59:289-301. [PMID: 21378283 DOI: 10.1369/0022155410397997] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cord blood-derived neural stem cells (NSCs) are proposed as an alternative cell source to repair brain damage upon transplantation. However, there is a lack of data showing how these cells are driven to generate desired phenotypes by recipient nervous tissue. Previous research indicates that local environment provides signals driving the fate of stem cells. To investigate the impact of these local cues interaction, the authors used a model of cord blood-derived NSCs co-cultured with different rat brain-specific primary cultures, creating the neural-like microenvironment conditions in vitro. Neuronal and astro-, oligo-, and microglia cell cultures were obtained by the previously described methods. The CMFDA-labeled neural stem cells originated from, non-transformed human umbilical cord blood cell line (HUCB-NSCs) established in a laboratory. The authors show that the close vicinity of astrocytes and oligodendrocytes promotes neuronal differentiation of HUCB-NSCs, whereas postmitotic neurons induce oligodendrogliogenesis of these cells. In turn, microglia or endothelial cells do not favor any phenotypes of their neural commitment. Studies have confirmed that HUCB-NSCs can read cues from the neurogenic microenvironment, attaining features of neurons, astrocytes, or oligodendrocytes. The specific responses of neurally committed cord blood-derived cells, reported in this work, are very much similar to those described previously for NSCs derived from other "more typical" sources. This further proves their genuine neural nature. Apart from having a better insight into the neurogenesis in the adult brain, these findings might be important when predicting cord blood cell derivative behavior after their transplantation for neurological disorders.
Collapse
Affiliation(s)
- Inga Markiewicz
- Neurorepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | | | | | | | |
Collapse
|
33
|
Goldberg JS, Hirschi KK. Diverse roles of the vasculature within the neural stem cell niche. Regen Med 2010; 4:879-97. [PMID: 19903006 DOI: 10.2217/rme.09.61] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
An interdependent relationship between the vascular and nervous systems begins during the earliest stages of development and persists through the mammalian lifespan. Accordingly, the process of adult neurogenesis involves the coordinated response of both systems to maintain a specialized microenvironment (niche) that tips the scale towards maintenance or regeneration, as needed. Understanding the nature and regulation of this balance will provide a foundation on which the potential for molecular- and stem cell-based therapies can be developed to treat prevalent CNS diseases and disorders. The vasculature is cited as a prominent feature within the adult subventricular zone and subgranular zone, known adult neural stem cell niches, helping to retain neural stem and progenitor cell potential. The vascular compartment within the neural stem cell niche has the unique opportunity to not only regulate neural stem and progenitor cells through direct contact with, and paracrine signaling from, endothelial and mural cells that make up blood vessels, but also integrates systemic signals into the local microenvironment via distribution of soluble factors from blood circulation to regulate stem cell niche behavior. Understanding the intricate role that the vasculature plays to influence neural stem cells in the context of niche regulation will help to bridge the gap from bench to bedside for the development of regeneration-based therapies for the CNS.
Collapse
Affiliation(s)
- Joshua S Goldberg
- Baylor College of Medicine, Department of Pediatrics & Molecular, Houston, TX 77030, USA
| | | |
Collapse
|
34
|
Butler JM, Kobayashi H, Rafii S. Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat Rev Cancer 2010; 10:138-46. [PMID: 20094048 PMCID: PMC2944775 DOI: 10.1038/nrc2791] [Citation(s) in RCA: 440] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The precise mechanisms whereby anti-angiogenesis therapy blocks tumour growth or causes vascular toxicity are unknown. We propose that endothelial cells establish a vascular niche that promotes tumour growth and tissue repair not only by delivering nutrients and O2 but also through an 'angiocrine' mechanism by producing stem and progenitor cell-active trophogens. Identification of endothelial-derived instructive angiocrine factors will allow direct tumour targeting, while diminishing the unwanted side effects associated with the use of anti-angiogenic agents.
Collapse
Affiliation(s)
- Jason M Butler
- Hideki Kobayashi and Shahin Rafii are at the Howard Hughes Medical Institute, Ansary Stem Cell Institute, Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | |
Collapse
|
35
|
Rozen N, Bick T, Bajayo A, Shamian B, Schrift-Tzadok M, Gabet Y, Yayon A, Bab I, Soudry M, Lewinson D. Transplanted blood-derived endothelial progenitor cells (EPC) enhance bridging of sheep tibia critical size defects. Bone 2009; 45:918-24. [PMID: 19665064 DOI: 10.1016/j.bone.2009.07.085] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 07/28/2009] [Accepted: 07/29/2009] [Indexed: 12/16/2022]
Abstract
The angiogenic events that accompany bone regeneration function as a "limiting factor" and are the primary regulatory mechanisms that direct the healing process. The general aim of this study was to test whether blood-derived progenitor cells that have endothelial characteristics (EPC), when applied to a large segmental defect, would promote bone regeneration. We established a critical-sized gap platform in sheep tibiae. Our model system takes advantage of the physiological wound healing process that occurs during the first two weeks following injury, and results in the gap being filled with scar tissue. EPC were expanded ex-vivo and 2 x 10(7) cells/0.2 ml were implanted into a wedged-shaped canal excavated in the fibrotic scar tissue. Sham treated sheep served as controls. Bone regeneration was followed every two weeks for three months by X-ray radiography. At the end of the experimental period, the regenerating segments were subjected to micro-computed tomographic (microCT) analysis. While minimal bone formation was detected in sham-treated sheep, six out of seven autologous EPC-transplanted sheep showed initial mineralization already by 2 weeks and complete bridging by 8-12 weeks post EPC transplantation. Histology of gaps 12 weeks post sham treatment showed mostly fibrotic scar tissue. On the contrary, EPC transplantation led to formation of dense and massive woven bone all throughout the defect. The results of this preclinical study open new therapeutic opportunities for the treatment of large scale bone injuries.
Collapse
Affiliation(s)
- Nimrod Rozen
- Department of Orthopaedic Surgery, Ha'emek Medical Center, Afula 18101, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Annenkov A. The insulin-like growth factor (IGF) receptor type 1 (IGF1R) as an essential component of the signalling network regulating neurogenesis. Mol Neurobiol 2009; 40:195-215. [PMID: 19714501 DOI: 10.1007/s12035-009-8081-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 08/14/2009] [Indexed: 02/07/2023]
Abstract
The insulin-like growth factor receptor type 1 (IGF1R) signalling pathway is activated in the mammalian nervous system from early developmental stages. Its major effect on developing neural cells is to promote their growth and survival. This pathway can integrate its action with signalling pathways of growth and morphogenetic factors that induce cell fate specification and selective expansion of specified neural cell subsets. This suggests that during developmental and adult neurogenesis cellular responses to many signalling factors, including ligands of Notch, sonic hedgehog, fibroblast growth factor family members, ligands of the epidermal growth factor receptor, bone morphogenetic proteins and Wingless and Int-1, may be modified by co-activation of the IGF1R. Modulation of cell migration is another possible role that IGF1R activation may play in neurogenesis. Here, I briefly overview neurogenesis and discuss a role for IGF1R-mediated signalling in the developing and mature nervous system with emphasis on crosstalk between the signalling pathways of the IGF1R and other factors regulating neural cell development and migration. Studies on neural as well as on non-neural cells are highlighted because it may be interesting to test in neurogenic paradigms some of the models based on the information obtained in studies on non-neural cell types.
Collapse
Affiliation(s)
- Alexander Annenkov
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, UK.
| |
Collapse
|
37
|
Abstract
Stem cells are multipotent cells that can give rise to a differentiated progeny as well as self-renew. The balanced coordination of these two stem cell fates is essential for embryonic development and tissue homeostasis in the adult. Perturbed stem cell function contributes significantly to a variety of pathological conditions, eg impaired self-renewal capacity due to cellular senescence contributes to ageing, and degenerative diseases or impaired stem cell differentiation by oncogenic mutations contribute to cancer formation. This review focuses on the molecular mechanisms involved in regulating the normal function of neural stem cells in the adult mammalian brain and on the involvement of these cells in brain pathology.
Collapse
Affiliation(s)
- G Yadirgi
- Institute of Cell and Molecular Science, St. Bartholomew's and the London School of Medicine and Dentistry, London, UK
| | | |
Collapse
|
38
|
Kusumoto KI, Parton A, Barnes D. Mitogen limitation and bone morphogenetic protein-4 promote neurogenesis in SFME cells, an EGF-dependent neural stem cell line. In Vitro Cell Dev Biol Anim 2008; 45:55-61. [PMID: 19057972 DOI: 10.1007/s11626-008-9153-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Accepted: 10/07/2008] [Indexed: 10/21/2022]
Abstract
Serum-free mouse embryo (SFME) cells are an epidermal growth factor (EGF)-dependent established line derived from brains of 16-d-old Balb/c mouse embryos. SFME cells grow indefinitely in serum-free medium without replicative senescence, chromosomal abnormalities, or malignant transformation. SFME cells express nestin, a neural stem cell marker, under serum-free conditions. Exposure to serum or transforming growth factor beta (TGF-beta) leads to a marked increase in differentiation toward the astrocytic lineage with expression of glial fibrillary acidic protein and other astrocyte markers. In this study, we show that treatment of SFME cells with bone morphogenetic protein-4 (BMP-4), another member of the TGF-beta family, led to differentiation toward a neuronal lineage under conditions of low mitogenic stimulation (0.5 ng/mL) by EGF and fibroblast growth factor. Maximum mitogenic stimulation with 50 ng/mL EGF blocked the BMP-4 effect on neuronal differentiation, but did not block TGF-beta-induced expression of markers of the astrocytic lineage. BMP-4 treatment also enhanced the activity of the neuron-specific enolase (NSE) promoter in SFME-NSE-lacZ cells that carry the gene for bacterial beta-galactosidase under the control of the NSE promoter. Extended BMP-4 treatment caused SFME cells to express a neuronal phenotype synthesizing gamma-aminobutyric acid. These results indicate that SFME cells have the capacity to generate both neurons and astrocytes in vitro, which resemble the behavior of EGF-dependent multipotential stem cells in the central nervous system, and establish a relationship between effects of BMP-4 and degree of mitogenic stimulation by other peptide growth factors.
Collapse
Affiliation(s)
- Ken-ichi Kusumoto
- Department of Biological Chemistry, Biotechnology and Food Research Institute, Fukuoka Industrial Technology Center, Kurume, Fukuoka 8390861, Japan
| | | | | |
Collapse
|
39
|
Maishi N, Annan DA, Kikuchi H, Hida Y, Hida K. An antiestrogen-binding protein in human tissues. Cancers (Basel) 1983; 11:cancers11101511. [PMID: 31600937 PMCID: PMC6826555 DOI: 10.3390/cancers11101511] [Citation(s) in RCA: 68] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/22/2022] Open
Abstract
Although nonsteroidal antiestrogens of the triphenylethylene type are generally considered to act through the estrogen receptor, some observations suggest that estrogen target tissues may also contain a binding protein specific for these compounds. The data so far reported, however, are also consistent with ligand-induced changes in conformation or in the state of aggregation of the estrogen receptor. The studies reported here demonstrate the existence of a protein in human myometrial cytosol which binds 1-[4-(2-dimethylaminoethoxy)phenyl]1,2-diphenylbut-1(Z)-ene ([3H]tamoxifen) with high affinity (Kd = 2.3 X 10(-9) M). This protein exhibits striking specificity for nonsteroidal antiestrogens. Estradiol competes weakly for bound [3H]tamoxifen, while other estrogens and nonestrogenic steroid hormones do not compete at all. Sedimentation analysis and molecular sieve chromatography indicate that the antiestrogen-binding protein is a larger species than the estrogen receptor and elutes from DEAE-Sephacel at a lower KCl concentration (0.03 M) than the estrogen receptor (0.15 M). Differential thermal stability of the estrogen receptor and the antiestrogen-binding protein was demonstrable in the absence of added ligand. The antiestrogen-binding protein was ubiquitous, being present in many tissues where estrogen receptor was undetectable. These findings support the separate existence of an antiestrogen-binding protein.
Collapse
Affiliation(s)
- Nako Maishi
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
| | - Dorcas A Annan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
| | - Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo 060-8636, Japan.
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan.
| | - Kyoko Hida
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
| |
Collapse
|