1
|
Liu S, Ren J, Hu Y, Zhou F, Zhang L. TGFβ family signaling in human stem cell self-renewal and differentiation. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:26. [PMID: 39604763 PMCID: PMC11602941 DOI: 10.1186/s13619-024-00207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Human stem cells are undifferentiated cells with the capacity for self-renewal and differentiation into distinct cell lineages, playing important role in the development and maintenance of diverse tissues and organs. The microenvironment of stem cell provides crucial factors and components that exert significant influence over the determination of cell fate. Among these factors, cytokines from the transforming growth factor β (TGFβ) superfamily, including TGFβ, bone morphogenic protein (BMP), Activin and Nodal, have been identified as important regulators governing stem cell maintenance and differentiation. In this review, we present a comprehensive overview of the pivotal roles played by TGFβ superfamily signaling in governing human embryonic stem cells, somatic stem cells, induced pluripotent stem cells, and cancer stem cells. Furthermore, we summarize the latest research and advancements of TGFβ family in various cancer stem cells and stem cell-based therapy, discussing their potential clinical applications in cancer therapy and regeneration medicine.
Collapse
Affiliation(s)
- Sijia Liu
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiang Ren
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanmei Hu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Long Zhang
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Moreira JF, Solá S. Dynamics of Neurogenic Signals as Biological Switchers of Brain Plasticity. Stem Cell Rev Rep 2024; 20:2032-2044. [PMID: 39259446 PMCID: PMC11554707 DOI: 10.1007/s12015-024-10788-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
The discovery of adult neurogenesis in the middle of the past century is considered one of the most important breakthroughs in neuroscience. Despite its controversial nature, this discovery shaped our concept of neural plasticity, revolutionizing the way we look at our brains. In fact, after the discovery of adult neurogenesis, we started to consider the brain as something even more dynamic and highly adaptable. In neurogenic niches, adult neurogenesis is supported by neural stem cells (NSCs). These cells possess a unique set of characteristics such as being quiescent for long periods while actively sensing and reacting to their surroundings to influence a multitude of processes, including the generation of new neurons and glial cells. Therefore, NSCs can be viewed as sentinels to our brain's homeostasis, being able to replace damaged cells and simultaneously secrete numerous factors that restore regular brain function. In addition, it is becoming increasingly evident that NSCs play a central role in memory formation and consolidation. In this review, we will dissect how NSCs influence their surroundings through paracrine and autocrine types of action. We will also depict the mechanism of action of each factor. Finally, we will describe how NSCs integrate different and often opposing signals to guide their fate.
Collapse
Affiliation(s)
- João F Moreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
3
|
Marcy G, Foucault L, Babina E, Capeliez T, Texeraud E, Zweifel S, Heinrich C, Hernandez-Vargas H, Parras C, Jabaudon D, Raineteau O. Single-cell analysis of the postnatal dorsal V-SVZ reveals a role for Bmpr1a signaling in silencing pallial germinal activity. SCIENCE ADVANCES 2023; 9:eabq7553. [PMID: 37146152 PMCID: PMC10162676 DOI: 10.1126/sciadv.abq7553] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The ventricular-subventricular zone (V-SVZ) is the largest neurogenic region of the postnatal forebrain, containing neural stem cells (NSCs) that emerge from both the embryonic pallium and subpallium. Despite of this dual origin, glutamatergic neurogenesis declines rapidly after birth, while GABAergic neurogenesis persists throughout life. We performed single-cell RNA sequencing of the postnatal dorsal V-SVZ for unraveling the mechanisms leading to pallial lineage germinal activity silencing. We show that pallial NSCs enter a state of deep quiescence, characterized by high bone morphogenetic protein (BMP) signaling, reduced transcriptional activity and Hopx expression, while in contrast, subpallial NSCs remain primed for activation. Induction of deep quiescence is paralleled by a rapid blockade of glutamatergic neuron production and differentiation. Last, manipulation of Bmpr1a demonstrates its key role in mediating these effects. Together, our results highlight a central role of BMP signaling in synchronizing quiescence induction and blockade of neuronal differentiation to rapidly silence pallial germinal activity after birth.
Collapse
Affiliation(s)
- Guillaume Marcy
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
- Univ Lyon, Université Claude Bernard Lyon 1, Bioinformatic Platform of the Labex Cortex, 69008 Lyon, France
| | - Louis Foucault
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Elodie Babina
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Timothy Capeliez
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Emeric Texeraud
- Univ Lyon, Université Claude Bernard Lyon 1, Bioinformatic Platform of the Labex Cortex, 69008 Lyon, France
| | - Stefan Zweifel
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Christophe Heinrich
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Hector Hernandez-Vargas
- Cancer Research Centre of Lyon (CRCL), INSERM U 1052, CNRS UMR 5286, UCBL1, Université de Lyon, Centre Léon Bérard, 28 rue Laennec, 69373 Lyon Cedex 08, France
| | - Carlos Parras
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- Clinic of Neurology, Geneva University Hospital, Geneva, Switzerland
| | - Olivier Raineteau
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| |
Collapse
|
4
|
Identification of the Time Period during Which BMP Signaling Regulates Proliferation of Neural Progenitor Cells in Zebrafish. Int J Mol Sci 2023; 24:ijms24021733. [PMID: 36675251 PMCID: PMC9863262 DOI: 10.3390/ijms24021733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling regulates neural induction, neuronal specification, and neuronal differentiation. However, the role of BMP signaling in neural progenitors remains unclear. This is because interruption of BMP signaling before or during neural induction causes severe effects on subsequent neural developmental processes. To examine the role of BMP signaling in the development of neural progenitors in zebrafish, we bypassed the effect of BMP signaling on neural induction and suppressed BMP signaling at different time points during gastrulation using a temporally controlled transgenic line carrying a dominant-negative form of Bmp receptor type 1aa and a chemical inhibitor of BMP signaling, DMH1. Inhibiting BMP signaling from 8 hpf could bypass BMP regulation on neural induction, induce the number of proliferating neural progenitors, and reduce the number of neuronal precursors. Inhibiting BMP signaling upregulates the expression of the Notch downstream gene hairy/E(spl)-related 2 (her2). Inhibiting Notch signaling or knocking down the Her2 function reduced neural progenitor proliferation, whereas inactivating BMP signaling in Notch-Her2 deficient background restored the number of proliferating neural progenitors. These results reveal the time window for the proliferation of neural progenitors during zebrafish development and a fine balance between BMP and Notch signaling in regulating the proliferation of neural progenitor cells.
Collapse
|
5
|
Current Opportunities for Targeting Dysregulated Neurodevelopmental Signaling Pathways in Glioblastoma. Cells 2022; 11:cells11162530. [PMID: 36010607 PMCID: PMC9406959 DOI: 10.3390/cells11162530] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most common and highly lethal type of brain tumor, with poor survival despite advances in understanding its complexity. After current standard therapeutic treatment, including tumor resection, radiotherapy and concomitant chemotherapy with temozolomide, the median overall survival of patients with this type of tumor is less than 15 months. Thus, there is an urgent need for new insights into GBM molecular characteristics and progress in targeted therapy in order to improve clinical outcomes. The literature data revealed that a number of different signaling pathways are dysregulated in GBM. In this review, we intended to summarize and discuss current literature data and therapeutic modalities focused on targeting dysregulated signaling pathways in GBM. A better understanding of opportunities for targeting signaling pathways that influences malignant behavior of GBM cells might open the way for the development of novel GBM-targeted therapies.
Collapse
|
6
|
Morales-Garcia JA, Calleja-Conde J, Lopez-Moreno JA, Alonso-Gil S, Sanz-SanCristobal M, Riba J, Perez-Castillo A. N,N-dimethyltryptamine compound found in the hallucinogenic tea ayahuasca, regulates adult neurogenesis in vitro and in vivo. Transl Psychiatry 2020; 10:331. [PMID: 32989216 PMCID: PMC7522265 DOI: 10.1038/s41398-020-01011-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
N,N-dimethyltryptamine (DMT) is a component of the ayahuasca brew traditionally used for ritual and therapeutic purposes across several South American countries. Here, we have examined, in vitro and vivo, the potential neurogenic effect of DMT. Our results demonstrate that DMT administration activates the main adult neurogenic niche, the subgranular zone of the dentate gyrus of the hippocampus, promoting newly generated neurons in the granular zone. Moreover, these mice performed better, compared to control non-treated animals, in memory tests, which suggest a functional relevance for the DMT-induced new production of neurons in the hippocampus. Interestingly, the neurogenic effect of DMT appears to involve signaling via sigma-1 receptor (S1R) activation since S1R antagonist blocked the neurogenic effect. Taken together, our results demonstrate that DMT treatment activates the subgranular neurogenic niche regulating the proliferation of neural stem cells, the migration of neuroblasts, and promoting the generation of new neurons in the hippocampus, therefore enhancing adult neurogenesis and improving spatial learning and memory tasks.
Collapse
Affiliation(s)
- Jose A. Morales-Garcia
- grid.4711.30000 0001 2183 4846Institute for Biomedical Research “A. Sols” (CSIC-UAM). Arturo Duperier 4, 28029 Madrid, Spain ,grid.413448.e0000 0000 9314 1427Spanish Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), c/ Valderrebollo 5, 28031 Madrid, Spain ,grid.4795.f0000 0001 2157 7667Department of Cellular Biology, School of Medicine, Complutense University of Madrid, Plaza Ramón y Cajal, 28040 Madrid, Spain ,grid.411347.40000 0000 9248 5770Cellular Neurobiology Laboratory, Neurobiology Department, UCS-UCM, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Javier Calleja-Conde
- grid.4795.f0000 0001 2157 7667Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Carretera de Humera, 28223 Madrid, Spain
| | - Jose A. Lopez-Moreno
- grid.4795.f0000 0001 2157 7667Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Carretera de Humera, 28223 Madrid, Spain
| | - Sandra Alonso-Gil
- grid.4711.30000 0001 2183 4846Institute for Biomedical Research “A. Sols” (CSIC-UAM). Arturo Duperier 4, 28029 Madrid, Spain ,grid.413448.e0000 0000 9314 1427Spanish Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), c/ Valderrebollo 5, 28031 Madrid, Spain
| | - Marina Sanz-SanCristobal
- grid.4711.30000 0001 2183 4846Institute for Biomedical Research “A. Sols” (CSIC-UAM). Arturo Duperier 4, 28029 Madrid, Spain ,grid.413448.e0000 0000 9314 1427Spanish Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), c/ Valderrebollo 5, 28031 Madrid, Spain
| | - Jordi Riba
- grid.5012.60000 0001 0481 6099Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, 6229 ER The Netherlands
| | - Ana Perez-Castillo
- Institute for Biomedical Research "A. Sols" (CSIC-UAM). Arturo Duperier 4, 28029, Madrid, Spain. .,Spanish Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), c/ Valderrebollo 5, 28031, Madrid, Spain. .,Cellular Neurobiology Laboratory, Neurobiology Department, UCS-UCM, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain.
| |
Collapse
|
7
|
Vieira de Castro J, Gonçalves CS, Hormigo A, Costa BM. Exploiting the Complexities of Glioblastoma Stem Cells: Insights for Cancer Initiation and Therapeutic Targeting. Int J Mol Sci 2020; 21:ijms21155278. [PMID: 32722427 PMCID: PMC7432229 DOI: 10.3390/ijms21155278] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
The discovery of glioblastoma stem cells (GSCs) in the 2000s revolutionized the cancer research field, raising new questions regarding the putative cell(s) of origin of this tumor type, and partly explaining the highly heterogeneous nature of glioblastoma (GBM). Increasing evidence has suggested that GSCs play critical roles in tumor initiation, progression, and resistance to conventional therapies. The remarkable oncogenic features of GSCs have generated significant interest in better defining and characterizing these cells and determining novel pathways driving GBM that could constitute attractive key therapeutic targets. While exciting breakthroughs have been achieved in the field, the characterization of GSCs is a challenge and the cell of origin of GBM remains controversial. For example, the use of several cell-surface molecular markers to identify and isolate GSCs has been a challenge. It is now widely accepted that none of these markers is, per se, sufficiently robust to distinguish GSCs from normal stem cells. Finding new strategies that are able to more efficiently and specifically target these niches could also prove invaluable against this devastating and therapy-insensitive tumor. In this review paper, we summarize the most relevant findings and discuss emerging concepts and open questions in the field of GSCs, some of which are, to some extent, pertinent to other cancer stem cells.
Collapse
Affiliation(s)
- Joana Vieira de Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Céline S. Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Adília Hormigo
- Department of Neurology, Neurosurgery, Medicine, The Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, NY 10029-6574, USA;
| | - Bruno M. Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
- Correspondence: ; Tel.: +35-1-253-604-872
| |
Collapse
|
8
|
Brodski C, Blaess S, Partanen J, Prakash N. Crosstalk of Intercellular Signaling Pathways in the Generation of Midbrain Dopaminergic Neurons In Vivo and from Stem Cells. J Dev Biol 2019; 7:jdb7010003. [PMID: 30650592 PMCID: PMC6473842 DOI: 10.3390/jdb7010003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Dopamine-synthesizing neurons located in the mammalian ventral midbrain are at the center stage of biomedical research due to their involvement in severe human neuropsychiatric and neurodegenerative disorders, most prominently Parkinson’s Disease (PD). The induction of midbrain dopaminergic (mDA) neurons depends on two important signaling centers of the mammalian embryo: the ventral midline or floor plate (FP) of the neural tube, and the isthmic organizer (IsO) at the mid-/hindbrain boundary (MHB). Cells located within and close to the FP secrete sonic hedgehog (SHH), and members of the wingless-type MMTV integration site family (WNT1/5A), as well as bone morphogenetic protein (BMP) family. The IsO cells secrete WNT1 and the fibroblast growth factor 8 (FGF8). Accordingly, the FGF8, SHH, WNT, and BMP signaling pathways play crucial roles during the development of the mDA neurons in the mammalian embryo. Moreover, these morphogens are essential for the generation of stem cell-derived mDA neurons, which are critical for the modeling, drug screening, and cell replacement therapy of PD. This review summarizes our current knowledge about the functions and crosstalk of these signaling pathways in mammalian mDA neuron development in vivo and their applications in stem cell-based paradigms for the efficient derivation of these neurons in vitro.
Collapse
Affiliation(s)
- Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel.
| | - Sandra Blaess
- Institute of Reconstructive Neurobiology, University of Bonn Medical Center, 53127 Bonn, Germany.
| | - Juha Partanen
- Faculty of Biological and Environmental Sciences, FIN00014-University of Helsinki, P.O. Box 56, Viikinkaari 9, FIN-00014 Helsinki, Finland.
| | - Nilima Prakash
- Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany.
| |
Collapse
|
9
|
BMP and WNT signalling cooperate through LEF1 in the neuronal specification of adult hippocampal neural stem and progenitor cells. Sci Rep 2018; 8:9241. [PMID: 29915186 PMCID: PMC6006330 DOI: 10.1038/s41598-018-27581-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/25/2018] [Indexed: 01/17/2023] Open
Abstract
Neuronal production from neural stem cells persists during adulthood in the subgranular zone of the hippocampal dentate gyrus. Extracellular signals provided by the hippocampal microenvironment regulate the neuronal fate commitment of the stem cell progeny. To date, the identity of those signals and their crosstalk has been only partially resolved. Here we show that adult rat hippocampal neural stem and progenitor cells (AH-NSPCs) express receptors for bone morphogenetic proteins (BMPs) and that the BMP/P-Smad pathway is active in AH-NSPCs undergoing differentiation towards the neuronal lineage. In vitro, exposure to the BMP2 and BMP4 ligands is sufficient to increase neurogenesis from AH-NSPCs in a WNT dependent manner while decreasing oligodendrogenesis. Moreover, BMP2/4 and WNT3A, a key regulator of adult hippocampal neurogenesis, cooperate to further enhance neuronal production. Our data point to a mechanistic convergence of the BMP and WNT pathways at the level of the T-cell factor/lymphoid enhancer factor gene Lef1. Altogether, we provide evidence that BMP signalling is an important regulator for the neuronal fate specification of AH-NSPCs cultures and we show that it significantly cooperates with the previously described master regulator of adult hippocampal neurogenesis, the WNT signalling pathway.
Collapse
|
10
|
Yamamoto H, Kurachi M, Naruse M, Shibasaki K, Ishizaki Y. BMP4 signaling in NPCs upregulates Bcl-xL to promote their survival in the presence of FGF-2. Biochem Biophys Res Commun 2018; 496:588-593. [DOI: 10.1016/j.bbrc.2018.01.090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 01/13/2018] [Indexed: 12/17/2022]
|
11
|
BMP/SMAD Pathway Promotes Neurogenesis of Midbrain Dopaminergic Neurons In Vivo and in Human Induced Pluripotent and Neural Stem Cells. J Neurosci 2018; 38:1662-1676. [PMID: 29321139 DOI: 10.1523/jneurosci.1540-17.2018] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 12/28/2017] [Accepted: 01/02/2018] [Indexed: 01/08/2023] Open
Abstract
The embryonic formation of midbrain dopaminergic (mDA) neurons in vivo provides critical guidelines for the in vitro differentiation of mDA neurons from stem cells, which are currently being developed for Parkinson's disease cell replacement therapy. Bone morphogenetic protein (BMP)/SMAD inhibition is routinely used during early steps of stem cell differentiation protocols, including for the generation of mDA neurons. However, the function of the BMP/SMAD pathway for in vivo specification of mammalian mDA neurons is virtually unknown. Here, we report that BMP5/7-deficient mice (Bmp5-/-; Bmp7-/-) lack mDA neurons due to reduced neurogenesis in the mDA progenitor domain. As molecular mechanisms accounting for these alterations in Bmp5-/-; Bmp7-/- mutants, we have identified expression changes of the BMP/SMAD target genes MSX1/2 (msh homeobox 1/2) and SHH (sonic hedgehog). Conditionally inactivating SMAD1 in neural stem cells of mice in vivo (Smad1Nes) hampered the differentiation of progenitor cells into mDA neurons by preventing cell cycle exit, especially of TH+SOX6+ (tyrosine hydroxylase, SRY-box 6) and TH+GIRK2+ (potassium voltage-gated channel subfamily-J member-6) substantia nigra neurons. BMP5/7 robustly increased the in vitro differentiation of human induced pluripotent stem cells and induced neural stem cells to mDA neurons by up to threefold. In conclusion, we have identified BMP/SMAD signaling as a novel critical pathway orchestrating essential steps of mammalian mDA neurogenesis in vivo that balances progenitor proliferation and differentiation. Moreover, we demonstrate the potential of BMPs to improve the generation of stem-cell-derived mDA neurons in vitro, highlighting the importance of sequential BMP/SMAD inhibition and activation in this process.SIGNIFICANCE STATEMENT We identify bone morphogenetic protein (BMP)/SMAD signaling as a novel essential pathway regulating the development of mammalian midbrain dopaminergic (mDA) neurons in vivo and provide insights into the molecular mechanisms of this process. BMP5/7 regulate MSX1/2 (msh homeobox 1/2) and SHH (sonic hedgehog) expression to direct mDA neurogenesis. Moreover, the BMP signaling component SMAD1 controls the differentiation of mDA progenitors, particularly to substantia nigra neurons, by directing their cell cycle exit. Importantly, BMP5/7 increase robustly the differentiation of human induced pluripotent and induced neural stem cells to mDA neurons. BMP/SMAD are routinely inhibited in initial stages of stem cell differentiation protocols currently being developed for Parkinson's disease cell replacement therapies. Therefore, our findings on opposing roles of the BMP/SMAD pathway during in vitro mDA neurogenesis might improve these procedures significantly.
Collapse
|
12
|
Morales-García JA, de la Fuente Revenga M, Alonso-Gil S, Rodríguez-Franco MI, Feilding A, Perez-Castillo A, Riba J. The alkaloids of Banisteriopsis caapi, the plant source of the Amazonian hallucinogen Ayahuasca, stimulate adult neurogenesis in vitro. Sci Rep 2017; 7:5309. [PMID: 28706205 PMCID: PMC5509699 DOI: 10.1038/s41598-017-05407-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/07/2017] [Indexed: 11/10/2022] Open
Abstract
Banisteriopsis caapi is the basic ingredient of ayahuasca, a psychotropic plant tea used in the Amazon for ritual and medicinal purposes, and by interested individuals worldwide. Animal studies and recent clinical research suggests that B. caapi preparations show antidepressant activity, a therapeutic effect that has been linked to hippocampal neurogenesis. Here we report that harmine, tetrahydroharmine and harmaline, the three main alkaloids present in B. caapi, and the harmine metabolite harmol, stimulate adult neurogenesis in vitro. In neurospheres prepared from progenitor cells obtained from the subventricular and the subgranular zones of adult mice brains, all compounds stimulated neural stem cell proliferation, migration, and differentiation into adult neurons. These findings suggest that modulation of brain plasticity could be a major contribution to the antidepressant effects of ayahuasca. They also expand the potential application of B. caapi alkaloids to other brain disorders that may benefit from stimulation of endogenous neural precursor niches.
Collapse
Affiliation(s)
- Jose A Morales-García
- Instituto de Investigaciones Biomédicas (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain.,Departamento de Biología Celular, Facultad de Medicina, UCM, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Mario de la Fuente Revenga
- Human Neuropsychopharmacology Research Group. Sant Pau Institute of Biomedical Research (IIB-Sant Pau). Sant Antoni María Claret, 167. 08025, Barcelona, Spain.,Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, 28006, Madrid, Spain.,MFR currently at: Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Sandra Alonso-Gil
- Instituto de Investigaciones Biomédicas (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
| | | | - Amanda Feilding
- The Beckley Foundation, Beckley Park, Oxford, OX3 9SY, United Kingdom
| | - Ana Perez-Castillo
- Instituto de Investigaciones Biomédicas (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain. .,Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain.
| | - Jordi Riba
- Human Neuropsychopharmacology Research Group. Sant Pau Institute of Biomedical Research (IIB-Sant Pau). Sant Antoni María Claret, 167. 08025, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Planta, 028029, Madrid, Spain.
| |
Collapse
|
13
|
Kawaguchi-Niida M, Shibata N, Furuta Y. Smad4 is essential for directional progression from committed neural progenitor cells through neuronal differentiation in the postnatal mouse brain. Mol Cell Neurosci 2017; 83:55-64. [PMID: 28669622 DOI: 10.1016/j.mcn.2017.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/01/2017] [Accepted: 06/25/2017] [Indexed: 01/19/2023] Open
Abstract
Signaling by the TGFβ super-family, consisting of TGFβ/activin- and bone morphogenetic protein (BMP) branch pathways, is involved in the central nervous system patterning, growth, and differentiation during embryogenesis. Neural progenitor cells are implicated in various pathological conditions, such as brain injury, infarction, Parkinson's disease and Alzheimer's disease. However, the roles of TGFβ/BMP signaling in the postnatal neural progenitor cells in the brain are still poorly understood. We examined the functional contribution of Smad4, a key integrator of TGFβ/BMP signaling pathways, to the regulation of neural progenitor cells in the subventricular zone (SVZ). Conditional loss of Smad4 in neural progenitor cells caused an increase in the number of neural stem like cells in the SVZ. Smad4 conditional mutants also exhibited attenuation in neuronal lineage differentiation in the adult brain that led to a deficit in olfactory bulb neurons as well as to a reduction of brain parenchymal volume. SVZ-derived neural stem/progenitor cells from the Smad4 mutant brains yielded increased growth of neurospheres, elevated self-renewal capacity and resistance to differentiation. These results indicate that loss of Smad4 in neural progenitor cells causes defects in progression of neural progenitor cell commitment within the SVZ and subsequent neuronal differentiation in the postnatal mouse brain.
Collapse
Affiliation(s)
- Motoko Kawaguchi-Niida
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan; Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | - Noriyuki Shibata
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yasuhide Furuta
- Animal Resource Development Unit and Genetic Engineering Team, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, Kobe, Japan; Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
Bokara KK, Kim JH, Kim JY, Lee JE. Transfection of arginine decarboxylase gene increases the neuronal differentiation of neural progenitor cells. Stem Cell Res 2016; 17:256-265. [PMID: 27591482 DOI: 10.1016/j.scr.2016.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/26/2016] [Accepted: 08/16/2016] [Indexed: 10/21/2022] Open
Abstract
Growing evidence suggests that the clinical use of neural progenitor cells (NPCs) is hampered by heterogeneity, poor neuronal yield and low survival rate. Recently, we reported that retrovirus-delivered human arginine decarboxylase (hADC) genes improve cell survival against oxidative insult in murine NPCs in vitro. This study investigates whether the induced expression of hADC gene in mNPCs induces any significant change in the cell fate commitment. The evaluation of induced hADC gene function was assessed by knockdown of hADC gene using specific siRNA. The hADC gene delivery triggered higher expression of N-CAM, cell adhesion molecule and MAP-2, neuronal marker. However, the hADC gene knockdown showed downregulation of N-CAM and MAP-2 expression suggesting that hADC gene delivery favors cell fate commitment of mNPCs towards neuronal lineage. Neurite outgrowth was significantly longer in the hADC infected cells. The neurotrophic signal, BDNF aided in the neuronal commitment, differentiation, and maturation of hADC-mNPCs through PI3K and ERK1/2 activation. The induction of neuron-like differentiation is believed to be regulated by the expression of GSK-3β and Wnt/β-catenin signaling pathways. Our findings suggest that hADC gene delivery favors cell fate commitment of mNPCs towards neuronal lineage, bring new advances in the field of neurogenesis and cell therapy.
Collapse
Affiliation(s)
- Kiran Kumar Bokara
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; CSIR-Centre for Cellular and Molecular Biology, Medical Biotechnology Complex, ANNEXE II, Uppal Road, Uppal, Hyderabad 500007, India.
| | - Jae Hwan Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| | - Jae Young Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; BK 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
15
|
Hover LD, Owens P, Munden AL, Wang J, Chambless LB, Hopkins CR, Hong CC, Moses HL, Abel TW. Bone morphogenetic protein signaling promotes tumorigenesis in a murine model of high-grade glioma. Neuro Oncol 2015; 18:928-38. [PMID: 26683138 DOI: 10.1093/neuonc/nov310] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 11/14/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Improved therapies for high-grade glioma (HGG) are urgently needed as the median survival for grade IV gliomas is only 15 months. Bone morphogenetic protein (BMP) signaling plays critical and complex roles in many types of cancer, including glioma, with most of the recently published work focusing on BMP-mediated regulation of glioma stem cells (GSCs). We hypothesized that BMP signaling may be an important modulator of tumorigenic properties in glioma cells outside of the GSC compartment. METHODS We used a human HGG tissue microarray and performed immunohistochemistry for phospho-Smads1,5,8. To examine the role of BMP signaling in tumorigenic astrocytes, transgenic mice were used to delete the BMP type IA receptor (Bmpr1a) and generate astrocytes transformed with oncogenic Ras and homozygous deletion of p53. The cells were transplanted orthotopically into immunocompetent adult host mice. RESULTS First we established that BMP signaling is active within the vast majority of HGG tumor cells. Mice implanted with BMPR1a-knockout transformed astrocytes showed an increase in median survival compared with mice that received BMPR1a-intact transformed astrocytes (52.5 vs 16 days). In vitro analysis showed that deletion of BMPR1a in oncogenic astrocytes resulted in decreased proliferation, decreased invasion, decreased migration, and increased expression of stemness markers. In addition, inhibition of BMP signaling in murine cells and astrocytoma cells with a small molecule BMP receptor kinase inhibitor resulted in similar tumor suppressive effects in vitro. CONCLUSION BMP inhibition may represent a viable therapeutic approach in adult HGG.
Collapse
Affiliation(s)
- Laura D Hover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (L.D.H., T.W.A.); Department of Cancer Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee (P.O., H.L.M.); Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee (A.M.); Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (J.W., L.C.); Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (C.H.); Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center (C.C.H.); Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee (C.C.H.)
| | - Philip Owens
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (L.D.H., T.W.A.); Department of Cancer Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee (P.O., H.L.M.); Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee (A.M.); Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (J.W., L.C.); Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (C.H.); Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center (C.C.H.); Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee (C.C.H.)
| | - Alexander L Munden
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (L.D.H., T.W.A.); Department of Cancer Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee (P.O., H.L.M.); Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee (A.M.); Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (J.W., L.C.); Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (C.H.); Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center (C.C.H.); Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee (C.C.H.)
| | - Jialiang Wang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (L.D.H., T.W.A.); Department of Cancer Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee (P.O., H.L.M.); Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee (A.M.); Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (J.W., L.C.); Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (C.H.); Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center (C.C.H.); Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee (C.C.H.)
| | - Lola B Chambless
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (L.D.H., T.W.A.); Department of Cancer Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee (P.O., H.L.M.); Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee (A.M.); Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (J.W., L.C.); Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (C.H.); Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center (C.C.H.); Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee (C.C.H.)
| | - Corey R Hopkins
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (L.D.H., T.W.A.); Department of Cancer Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee (P.O., H.L.M.); Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee (A.M.); Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (J.W., L.C.); Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (C.H.); Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center (C.C.H.); Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee (C.C.H.)
| | - Charles C Hong
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (L.D.H., T.W.A.); Department of Cancer Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee (P.O., H.L.M.); Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee (A.M.); Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (J.W., L.C.); Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (C.H.); Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center (C.C.H.); Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee (C.C.H.)
| | - Harold L Moses
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (L.D.H., T.W.A.); Department of Cancer Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee (P.O., H.L.M.); Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee (A.M.); Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (J.W., L.C.); Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (C.H.); Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center (C.C.H.); Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee (C.C.H.)
| | - Ty W Abel
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (L.D.H., T.W.A.); Department of Cancer Biology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee (P.O., H.L.M.); Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee (A.M.); Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee (J.W., L.C.); Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (C.H.); Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center (C.C.H.); Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee (C.C.H.); Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee (C.C.H.)
| |
Collapse
|
16
|
Choe Y, Pleasure SJ, Mira H. Control of Adult Neurogenesis by Short-Range Morphogenic-Signaling Molecules. Cold Spring Harb Perspect Biol 2015; 8:a018887. [PMID: 26637286 DOI: 10.1101/cshperspect.a018887] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Adult neurogenesis is dynamically regulated by a tangled web of local signals emanating from the neural stem cell (NSC) microenvironment. Both soluble and membrane-bound niche factors have been identified as determinants of adult neurogenesis, including morphogens. Here, we review our current understanding of the role and mechanisms of short-range morphogen ligands from the Wnt, Notch, Sonic hedgehog, and bone morphogenetic protein (BMP) families in the regulation of adult neurogenesis. These morphogens are ideally suited to fine-tune stem-cell behavior, progenitor expansion, and differentiation, thereby influencing all stages of the neurogenesis process. We discuss cross talk between their signaling pathways and highlight findings of embryonic development that provide a relevant context for understanding neurogenesis in the adult brain. We also review emerging examples showing that the web of morphogens is in fact tightly linked to the regulation of neurogenesis by diverse physiologic processes.
Collapse
Affiliation(s)
- Youngshik Choe
- Department of Neurology, Programs in Neuroscience, Developmental and Stem Cell Biology, UCSF Institute for Regeneration Medicine, San Francisco, California 94158
| | - Samuel J Pleasure
- Department of Neurology, Programs in Neuroscience, Developmental and Stem Cell Biology, UCSF Institute for Regeneration Medicine, San Francisco, California 94158
| | - Helena Mira
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| |
Collapse
|
17
|
Sallustio F, Serino G, Schena FP. Potential Reparative Role of Resident Adult Renal Stem/Progenitor Cells in Acute Kidney Injury. Biores Open Access 2015; 4:326-33. [PMID: 26309808 PMCID: PMC4509615 DOI: 10.1089/biores.2015.0011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human kidney is particularly susceptible to ischemia and toxins with consequential tubular necrosis and activation of inflammatory processes. This process can lead to the acute renal injury, and even if the kidney has a great capacity for regeneration after tubular damage, in several circumstances, the normal renal repair program may not be sufficient to achieve a successful regeneration. Resident adult renal stem/progenitor cells could participate in this repair process and have the potentiality to enhance the renal regenerative mechanism. This could be achieved both directly, by means of their capacity to differentiate and integrate into the renal tissues, and by means of paracrine factors able to induce or improve the renal repair or regeneration. Recent genetic fate-tracing studies indicated that tubular damage is instead repaired by proliferative duplication of epithelial cells, acquiring a transient progenitor phenotype and by fate-restricted clonal cell progeny emerging from different nephron segments. In this review, we discuss about the properties and the reparative characteristics of high regenerative CD133(+)/CD24(+) cells, with a view to a future application of these cells for the treatment of acute renal injury.
Collapse
Affiliation(s)
- Fabio Sallustio
- Department of Emergency and Organ Transplantation, University of Bari , Bari, Italy . ; C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3 , Valenzano, Italy . ; Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento , Lecce-Monteroni, Lecce, Italy
| | - Grazia Serino
- Department of Emergency and Organ Transplantation, University of Bari , Bari, Italy
| | - Francesco Paolo Schena
- C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3 , Valenzano, Italy . ; Schena Foundation, Research Center of Renal Diseases , Bari, Italy
| |
Collapse
|
18
|
Videla Richardson GA, Garcia CP, Roisman A, Slavutsky I, Fernandez Espinosa DD, Romorini L, Miriuka SG, Arakaki N, Martinetto H, Scassa ME, Sevlever GE. Specific Preferences in Lineage Choice and Phenotypic Plasticity of Glioma Stem Cells Under BMP4 and Noggin Influence. Brain Pathol 2015; 26:43-61. [PMID: 25808628 DOI: 10.1111/bpa.12263] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 03/12/2015] [Indexed: 12/23/2022] Open
Abstract
Although BMP4-induced differentiation of glioma stem cells (GSCs) is well recognized, details of the cellular responses triggered by this morphogen are still poorly defined. In this study, we established several GSC-enriched cell lines (GSC-ECLs) from high-grade gliomas. The expansion of these cells as adherent monolayers, and not as floating neurospheres, enabled a thorough study of the phenotypic changes that occurred during their differentiation. Herein, we evaluated GSC-ECLs' behavior toward differentiating conditions by depriving them of growth factors and/or by adding BMP4 at different concentrations. After analyzing cellular morphology, proliferation and lineage marker expression, we determined that GSC-ECLs have distinct preferences in lineage choice, where some of them showed an astrocyte fate commitment and others a neuronal one. We found that this election seems to be dictated by the expression pattern of BMP signaling components present in each GSC-ECL. Additionally, treatment of GSC-ECLs with the BMP antagonist, Noggin, also led to evident phenotypic changes. Interestingly, under certain conditions, some GSC-ECLs adopted an unexpected smooth muscle-like phenotype. As a whole, our findings illustrate the wide differentiation potential of GSCs, highlighting their molecular complexity and paving a way to facilitate personalized differentiating therapies.
Collapse
Affiliation(s)
- Guillermo Agustín Videla Richardson
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Carolina Paola Garcia
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Alejandro Roisman
- Laboratorio de Neuropatología, Departamento de Neuropatología y Biología Molecular, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Irma Slavutsky
- Laboratorio de Neuropatología, Departamento de Neuropatología y Biología Molecular, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Damián Darío Fernandez Espinosa
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Leonardo Romorini
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Santiago Gabriel Miriuka
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Naomi Arakaki
- Laboratorio de Biología Molecular, Departamento de Neuropatología y Biología Molecular, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Horacio Martinetto
- Laboratorio de Genética de Neoplasias Linfoides, Instituto de Medicina Experimental, CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - María Elida Scassa
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Gustavo Emilio Sevlever
- Laboratorio de Biología Molecular, Departamento de Neuropatología y Biología Molecular, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| |
Collapse
|
19
|
Morales-Garcia JA, Alonso-Gil S, Gil C, Martinez A, Santos A, Perez-Castillo A. Phosphodiesterase 7 inhibition induces dopaminergic neurogenesis in hemiparkinsonian rats. Stem Cells Transl Med 2015; 4:564-75. [PMID: 25925836 DOI: 10.5966/sctm.2014-0277] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/09/2015] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Parkinson's disease is characterized by a loss of dopaminergic neurons in a specific brain region, the ventral midbrain. Parkinson's disease is diagnosed when approximately 50% of the dopaminergic neurons of the substantia nigra pars compacta (SNpc) have degenerated and the others are already affected by the disease. Thus, it is conceivable that all therapeutic strategies, aimed at neuroprotection, start too late. Therefore, an urgent medical need exists to discover new pharmacological targets and novel drugs with disease-modifying properties. In this regard, modulation of endogenous adult neurogenesis toward a dopaminergic phenotype might provide a new strategy to target Parkinson's disease by partially ameliorating the dopaminergic cell loss that occurs in this disorder. We have previously shown that a phosphodiesterase 7 (PDE7) inhibitor, S14, exerts potent neuroprotective and anti-inflammatory effects in different rodent models of Parkinson's disease, indicating that this compound could represent a novel therapeutic agent to stop the dopaminergic cell loss that occurs during the progression of the disease. In this report we show that, in addition to its neuroprotective effect, the PDE7 inhibitor S14 is also able to induce endogenous neuroregenerative processes toward a dopaminergic phenotype. We describe a population of actively dividing cells that give rise to new neurons in the SNpc of hemiparkinsonian rats after treatment with S14. In conclusion, our data identify S14 as a novel regulator of dopaminergic neuron generation. SIGNIFICANCE Parkinson's disease is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the ventral midbrain. Currently, no cure and no effective disease-modifying therapy are available for Parkinson's disease; therefore, an urgent medical need exists to discover new pharmacological targets and novel drugs for the treatment of this disorder. The present study reports that an inhibitor of the enzyme phosphodiesterase 7 (S14) induces proliferation in vitro and in vivo of neural stem cells, promoting its differentiation toward a dopaminergic phenotype and therefore enhancing dopaminergic neuron generation. Because this drug is also able to confer neuroprotection of these cells in animal models of Parkinson's disease, S14 holds great promise as a therapeutic new strategy for this disorder.
Collapse
Affiliation(s)
- Jose A Morales-Garcia
- Instituto de Investigaciones Biomédicas, CSIC-UAM, Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Centro de Investigaciones Biológicas (CSIC), Madrid, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, UCM, Madrid, Spain
| | - Sandra Alonso-Gil
- Instituto de Investigaciones Biomédicas, CSIC-UAM, Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Centro de Investigaciones Biológicas (CSIC), Madrid, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, UCM, Madrid, Spain
| | - Carmen Gil
- Instituto de Investigaciones Biomédicas, CSIC-UAM, Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Centro de Investigaciones Biológicas (CSIC), Madrid, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, UCM, Madrid, Spain
| | - Ana Martinez
- Instituto de Investigaciones Biomédicas, CSIC-UAM, Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Centro de Investigaciones Biológicas (CSIC), Madrid, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, UCM, Madrid, Spain
| | - Angel Santos
- Instituto de Investigaciones Biomédicas, CSIC-UAM, Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Centro de Investigaciones Biológicas (CSIC), Madrid, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, UCM, Madrid, Spain
| | - Ana Perez-Castillo
- Instituto de Investigaciones Biomédicas, CSIC-UAM, Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Centro de Investigaciones Biológicas (CSIC), Madrid, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, UCM, Madrid, Spain
| |
Collapse
|
20
|
Hover LD, Abel TW, Owens P. Genomic Analysis of the BMP Family in Glioblastomas. TRANSLATIONAL ONCOGENOMICS 2015; 7:1-9. [PMID: 25987829 PMCID: PMC4406393 DOI: 10.4137/tog.s22256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/22/2015] [Accepted: 01/29/2015] [Indexed: 12/29/2022]
Abstract
Glioblastoma multiforme (GBM) is a grade IV glioma with a median survival of 15 months. Recently,
bone morphogenetic protein (BMP) signaling has been shown to promote survival in xenograft murine
models. To gain a better understanding of the role of BMP signaling in human GBMs, we examined the
genomic alterations of 90 genes associated with BMP signaling in GBM patient samples. We completed
this analysis using publically available datasets compiled through The Cancer Genome Atlas and the
Glioma Molecular Diagnostic Initiative. Here we show how mRNA expression is altered in GBM samples
and how that is associated with patient survival, highlighting both known and novel associations
between BMP signaling and GBM biology.
Collapse
Affiliation(s)
- Laura D Hover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ty W Abel
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Philip Owens
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
21
|
Koszinowski S, Buss K, Kaehlcke K, Krieglstein K. Signaling via the transcriptionally regulated activin receptor 2B is a novel mediator of neuronal cell death during chicken ciliary ganglion development. Int J Dev Neurosci 2015; 41:98-104. [PMID: 25660516 DOI: 10.1016/j.ijdevneu.2015.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 01/30/2015] [Accepted: 01/31/2015] [Indexed: 11/25/2022] Open
Abstract
The TGF-β ligand superfamily members activin A and BMP control important aspects of embryonic neuronal development and differentiation. Both are known to bind to activin receptor subtypes IIA (ActRIIA) and IIB, while in the avian ciliary ganglion (CG), so far only ActRIIA-expression has been described. We show that the expression of ACVR2B, coding for the ActRIIB, is tightly regulated during CG development and the knockdown of ACVR2B expression leads to a deregulation in the execution of neuronal apoptosis and therefore affects ontogenetic programmed cell death in vivo. While the differentiation of choroid neurons was impeded in the knockdown, pointing toward a reduction in activin A-mediated neural differentiation signaling, naturally occurring neuronal cell death in the CG was not prevented by follistatin treatment. Systemic injections of the BMP antagonist noggin, on the other hand, reduced the number of apoptotic neurons to a similar extent as ACVR2B knockdown. We therefore propose a novel pathway in the regulation of CG neuron ontogenetic programmed cell death, which could be mediated by BMP and signals via the ActRIIB.
Collapse
Affiliation(s)
- S Koszinowski
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany; Faculty of Biology, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany.
| | - K Buss
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany
| | - K Kaehlcke
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany
| | - K Krieglstein
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany.
| |
Collapse
|
22
|
Ramasamy S, Yu F, Hong Yu Y, Srivats H, Dawe GS, Ahmed S. NogoR1 and PirB signaling stimulates neural stem cell survival and proliferation. Stem Cells 2015; 32:1636-48. [PMID: 24449409 DOI: 10.1002/stem.1645] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 12/11/2013] [Indexed: 11/07/2022]
Abstract
Neural stem cells (NSCs) and neural progenitors (NPs) in the mammalian neocortex give rise to the main cell types of the nervous system. The biological behavior of these NSCs and NPs is regulated by extracellular niche derived autocrine-paracrine signaling factors on a developmental timeline. Our previous reports [Plos One 2010;5:e15341; J Neurochem 2011;117:565-578] have shown that chondroitin sulfate proteoglycan and ApolipoproteinE are autocrine-paracrine survival factors for NSCs. NogoA, a myelin related protein, is expressed in the cortical ventricular zones where NSCs reside. However, the functional role of Nogo signaling proteins in NSC behavior is not completely understood. In this study, we show that NogoA receptors, NogoR1 and PirB, are expressed in the ventricular zone where NSCs reside between E10.5 and 14.5 but not at E15.5. Nogo ligands stimulate NSC survival and proliferation in a dosage-dependent manner in vitro. NogoR1 and PirB are low and high affinity Nogo receptors, respectively and are responsible for the effects of Nogo ligands on NSC behavior. Inhibition of autocrine-paracrine Nogo signaling blocks NSC survival and proliferation. In NSCs, NogoR1 functions through Rho whereas PirB uses Shp1/2 signaling pathways to control NSC behavior. Taken together, this work suggests that Nogo signaling is an important pathway for survival of NSCs.
Collapse
Affiliation(s)
- Srinivas Ramasamy
- Institute of Medical Biology, 8A Biomedical Grove, #05-37 Immunos, Singapore
| | | | | | | | | | | |
Collapse
|
23
|
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
|
24
|
Gallo V, Deneen B. Glial development: the crossroads of regeneration and repair in the CNS. Neuron 2014; 83:283-308. [PMID: 25033178 DOI: 10.1016/j.neuron.2014.06.010] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2014] [Indexed: 02/07/2023]
Abstract
Given the complexities of the mammalian CNS, its regeneration is viewed as the holy grail of regenerative medicine. Extraordinary efforts have been made to understand developmental neurogenesis, with the hopes of clinically applying this knowledge. CNS regeneration also involves glia, which comprises at least 50% of the cellular constituency of the brain and is involved in all forms of injury and disease response, recovery, and regeneration. Recent developmental studies have given us unprecedented insight into the processes that regulate the generation of CNS glia. Because restorative processes often parallel those found in development, we will peer through the lens of developmental gliogenesis to gain a clearer understanding of the processes that underlie glial regeneration under pathological conditions. Specifically, this review will focus on key signaling pathways that regulate astrocyte and oligodendrocyte development and describe how these mechanisms are reutilized in these populations during regeneration and repair after CNS injury.
Collapse
Affiliation(s)
- Vittorio Gallo
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC 20010, USA.
| | - Benjamin Deneen
- Department of Neuroscience and Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Bond AM, Peng CY, Meyers EA, McGuire T, Ewaleifoh O, Kessler JA. BMP signaling regulates the tempo of adult hippocampal progenitor maturation at multiple stages of the lineage. Stem Cells 2014; 32:2201-14. [PMID: 24578327 DOI: 10.1002/stem.1688] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 01/24/2014] [Accepted: 02/18/2014] [Indexed: 01/15/2023]
Abstract
Novel environmental stimuli, such as running and learning, increase proliferation of adult hippocampal neural stem cells (NSCs) and enlarge the population of new neurons. However, it remains unclear how increased numbers of new neurons can be generated in a time frame far shorter than the time required for proliferating stem cells to generate these neurons. Here, we show that bone morphogenetic protein (BMP) signaling in the subgranular zone regulates the tempo of neural progenitor cell (NPC) maturation by directing their transition between states of quiescence and activation at multiple stages along the lineage. Virally mediated overexpression of BMP4 caused NPC cell cycle exit and slowed the normal maturation of NPCs, resulting in a long-term reduction in neurogenesis. Conversely, overexpression of the BMP inhibitor noggin promoted NPC cell cycle entry and accelerated NPC maturation. Similarly, BMP receptor type 2 (BMPRII) ablation in Ascl1(+) intermediate NPCs accelerated their maturation into neurons. Importantly, ablation of BMPRII in GFAP(+) stem cells accelerated maturation without depleting the NSC pool, indicating that an increased rate of neurogenesis does not necessarily diminish the stem cell population. Thus, inhibition of BMP signaling is a mechanism for rapidly expanding the pool of new neurons in the adult hippocampus by tipping the balance between quiescence/activation of NPCs and accelerating the rate at which they mature into neurons.
Collapse
Affiliation(s)
- Allison M Bond
- Department of Neurology, Northwestern University's Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
26
|
Zhang Z, Lin CCJ. Taking advantage of neural development to treat glioblastoma. Eur J Neurosci 2014; 40:2859-66. [PMID: 24964151 DOI: 10.1111/ejn.12655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/29/2014] [Accepted: 05/11/2014] [Indexed: 01/02/2023]
Abstract
Glioblastoma (GBM) is by far the most common and most malignant primary adult brain tumor (World Health Organization grade IV), containing a fraction of stem-like cells that are highly tumorigenic and multipotent. Recent research has revealed that GBM stem-like cells play important roles in GBM pathogenesis. GBM is thought to arise from genetic anomalies in glial development. Over the past decade, a wide range of studies have shown that several signaling pathways involved in neural development, including basic helix-loop-helix, Wnt-β-catenin, bone morphogenetic proteins-Smads, epidermal growth factor-epidermal growth factor receptor, and Notch, play important roles in GBM pathogenesis. In this review, we highlight the significance of these pathways in the context of developing treatments for GBM. Extrapolating knowledge and concepts from neural development will have significant implications for designing better strategies with which to treat GBM.
Collapse
Affiliation(s)
- Zhiyuan Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, School of Medicine, Nanjing University, Jiangsu Province, China; Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | | |
Collapse
|
27
|
Rahman M, Azari H, Deleyrolle L, Millette S, Zeng H, Reynolds BA. Controlling tumor invasion: bevacizumab and BMP4 for glioblastoma. Future Oncol 2014; 9:1389-96. [PMID: 23980685 DOI: 10.2217/fon.13.96] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Bevacizumab has been reported to result in increased tumor invasion when used to treat malignant glioma. We hypothesized that BMP4 would prevent diffuse tumor infiltration induced by bevacizumab for malignant glioma in a xenograft model. METHODS Human glioblastoma (GBM) tumor cells were implanted in the striatum of immunocompromised mice. The animals were treated with bevacizumab and BMP4. Tumor growth and invasion were measured. RESULTS The bevacizumab-treated mice had increased survival compared with control animals (p = 0.02). BMP4 alone did not result in improved survival (p = 1.0). The bevacizumab (p = 0.006) and bevacizumab plus BMP4 (p = 0.006) groups demonstrated significantly decreased total tumor size compared with control. Tumor invasion was significantly decreased in the bevacizumab (p = 0.005), BMP4 (p = 0.04) alone and bevacizumab plus BMP4 (p = 0.002) groups compared with control. No synergistic effect between bevacizumab and BMP4 was observed. CONCLUSION Bevacizumab treatment did not result in diffuse infiltration of human GBM in a mouse xenograft model. BMP4 did have an independent favorable effect on GBM that was not synergistic with bevacizumab treatment.
Collapse
Affiliation(s)
- Maryam Rahman
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA. n
| | | | | | | | | | | |
Collapse
|
28
|
Xu D, Zhao W, Pan G, Qian M, Zhu X, Liu W, Cai G, Cui Z. Expression of Nemo-like kinase after spinal cord injury in rats. J Mol Neurosci 2014; 52:410-8. [PMID: 24395089 DOI: 10.1007/s12031-013-0191-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 11/20/2013] [Indexed: 12/13/2022]
Abstract
Wnt can induce signal transduction via the canonical pathway, which was involved in many processes in the nervous system. Nemo-like kinase (NLK) acts as a negative regulator of β-catenin/T-cell factor/lymphoid enhancer factor (LEF) and functions downstream of transforming growth factor β-activated kinase-1 in the Wnt signaling pathway. In this study, we performed a spinal cord injury (SCI) test in adult Sprague-Dawley rats and investigated the dynamic changes and role of NLK expression in the spinal cord. Western blot analysis revealed that NLK expression was low in normal spinal cord. It then increased markedly, peaked at 3 days, and declined to basal levels from 5 days after injury. Immunohistochemistry confirmed that NLK immunoactivity was expressed at low levels in gray and white matter under normal conditions and increased prominently in gray matter after the SCI test. Double immunofluorescent staining for NLK, caspase-3, β-catenin, and NeuN (neuronal nuclei) revealed that NLK and β-catenin were markedly increased and colocalized in apoptotic neurons. Coimmunoprecipitation data demonstrated that overexpression of NLK protein reduced β-catenin binding to LEF-1. Our results suggested that NLK was associated with neuronal apoptosis through attenuating the Wnt/β-catenin signaling pathway after SCIs.
Collapse
Affiliation(s)
- Dawei Xu
- Department of Orthopedics, The Second Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Dendrite complexity of sympathetic neurons is controlled during postnatal development by BMP signaling. J Neurosci 2013; 33:15132-44. [PMID: 24048844 DOI: 10.1523/jneurosci.4748-12.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Dendrite development is controlled by the interplay of intrinsic and extrinsic signals affecting initiation, growth, and maintenance of complex dendrites. Bone morphogenetic proteins (BMPs) stimulate dendrite growth in cultures of sympathetic, cortical, and hippocampal neurons but it was unclear whether BMPs control dendrite morphology in vivo. Using a conditional knock-out strategy to eliminate Bmpr1a and Smad4 in immature noradrenergic sympathetic neurons we now show that dendrite length, complexity, and neuron cell body size are reduced in adult mice deficient of Bmpr1a. The combined deletion of Bmpr1a and Bmpr1b causes no further decrease in dendritic features. Sympathetic neurons devoid of Bmpr1a/1b display normal Smad1/5/8 phosphorylation, which suggests that Smad-independent signaling paths are involved in dendritic growth control downstream of BMPR1A/B. Indeed, in the Smad4 conditional knock-out dendrite and cell body size are not affected and dendrite complexity and number are increased. Together, these results demonstrate an in vivo function for BMPs in the generation of mature sympathetic neuron dendrites. BMPR1 signaling controls dendrite complexity postnatally during the major dendritic growth period of sympathetic neurons.
Collapse
|
30
|
Chen X, Yao Y, Guan J, Chen X, Zhang F. Up-regulation of FoxN4 expression in adult spinal cord after injury. J Mol Neurosci 2013; 52:403-9. [PMID: 24217796 PMCID: PMC3924027 DOI: 10.1007/s12031-013-0166-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/28/2013] [Indexed: 11/26/2022]
Abstract
FoxN4 (forkhead box N4), which is a transcription factor involved in developing spinal cord and spinal neurogenesis, implied important roles in the central nervous system (CNS). However, its expression and function in the adult CNS lesion are still unclear. In this study, we established a spinal cord injury (SCI) model in adult rats and investigated the expression of FoxN4 in the spinal cord. Western blot analysis revealed that FoxN4 was present in normal spinal cord. It gradually increased, peaked at day 3 after SCI, and then decreased during the following days. Immunohistochemistry further confirmed that FoxN4 was expressed at low levels in gray and white matters in normal condition and increased after SCI. Double immunofluorescence staining showed that FoxN4 is located on neurons and astrocytes, and FoxN4 expression was increased progressively in reactive astrocytes within the vicinity of the lesion, predominately in the white matter. In addition, almost all FoxN4-positive cells also expressed nestin or PCNA. Our data suggested that FoxN4 might play important roles in CNS pathophysiology after SCI.
Collapse
Affiliation(s)
- Xiangdong Chen
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Yu Yao
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Junjie Guan
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Xiaoqing Chen
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Feng Zhang
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| |
Collapse
|
31
|
Gámez B, Rodriguez-Carballo E, Ventura F. BMP signaling in telencephalic neural cell specification and maturation. Front Cell Neurosci 2013; 7:87. [PMID: 23761735 PMCID: PMC3671186 DOI: 10.3389/fncel.2013.00087] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 05/21/2013] [Indexed: 12/13/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) make up a family of morphogens that are critical for patterning, development, and function of the central and peripheral nervous system. Their effects on neural cells are pleiotropic and highly dynamic depending on the stage of development and the local niche. Neural cells display a broad expression profile of BMP ligands, receptors, and transducer molecules. Moreover, interactions of BMP signaling with other incoming morphogens and signaling pathways are crucial for most of these processes. The key role of BMP signaling suggests that it includes many regulatory mechanisms that restrict BMP activity both temporally and spatially. BMPs affect neural cell fate specification in a dynamic fashion. Initially they inhibit proliferation of neural precursors and promote the first steps in neuronal differentiation. Later on, BMP signaling effects switch from neuronal induction to promotion of astroglial identity and inhibition of neuronal or oligodendroglial lineage commitment. Furthermore, in postmitotic cells, BMPs regulate cell survival and death, to modulate neuronal subtype specification, promote dendritic and axonal growth and induce synapse formation and stabilization. In this review, we examine the canonical and non-canonical mechanisms of BMP signal transduction. Moreover, we focus on the specific role of BMPs in the nervous system including their ability to regulate neural stem cell proliferation, self-renewal, lineage specification, and neuronal function.
Collapse
Affiliation(s)
- Beatriz Gámez
- Departament de Ciències Fisiològiques II, Institut d'Investigació Biomèdica de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat Spain
| | | | | |
Collapse
|
32
|
Persano L, Rampazzo E, Basso G, Viola G. Glioblastoma cancer stem cells: Role of the microenvironment and therapeutic targeting. Biochem Pharmacol 2013; 85:612-622. [DOI: 10.1016/j.bcp.2012.10.001] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 10/01/2012] [Accepted: 10/01/2012] [Indexed: 12/22/2022]
|
33
|
Hepatocyte growth factor activator inhibitor-1 is induced by bone morphogenetic proteins and regulates proliferation and cell fate of neural progenitor cells. PLoS One 2013; 8:e56117. [PMID: 23409135 PMCID: PMC3567048 DOI: 10.1371/journal.pone.0056117] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 01/04/2013] [Indexed: 12/20/2022] Open
Abstract
Background Neural progenitor cells (NPCs) in the developing neuroepithelium are regulated by intrinsic and extrinsic factors. There is evidence that NPCs form a self-supporting niche for cell maintenance and proliferation. However, molecular interactions and cell-cell contacts and the microenvironment within the neuroepithelium are largely unknown. We hypothesized that cellular proteases especially those associated with the cell surface of NPCs play a role in regulation of progenitor cells in the brain. Methodology/Principal Findings In this work, we show that NPCs, isolated from striatal anlage of developing rat brain, express hepatocyte growth factor activator inhibitor-1 and -2 (HAI-1 and HAI-2) that are cell surface-linked serine protease inhibitors. In addition, radial glia cells derived from mouse embryonic stem cells also express HAI-1 and HAI-2. To study the functional significance of HAI-1 and HAI-2 in progenitor cells, we modulated their levels using expression plasmids or silencing RNA (siRNA) transfected into the NPCs. Data showed that overexpression of HAI-1 or HAI-2 decreased cell proliferation of cultured NPCs, whilst their siRNAs had opposite effects. HAI-1 also influenced NPC differentiation by increasing the number of glial fibrillary acidic protein (GFAP) expressing cells in the culture. Expression of HAI-1 in vivo decreased cell proliferation in developing neuroepithelium in E15 old animals and promoted astrocyte cell differentiation in neonatal animals. Studying the regulation of HAI-1, we observed that Bone morphogenetic protein-2 (BMP-2) and BMP-4 increased HAI-1 levels in the NPCs. Experiments using HAI-1-siRNA showed that these BMPs act on the NPCs partly in a HAI-1-dependent manner. Conclusions This study shows that the cell-surface serine protease inhibitors, HAI-1 and HAI-2 influence proliferation and cell fate of NPCs and their expression levels are linked to BMP signaling. Modulation of the levels and actions of HAI-1 in NPCs may be of a potential value in stem cell therapies in various brain diseases.
Collapse
|
34
|
Musser MA, Michelle Southard-Smith E. Balancing on the crest - Evidence for disruption of the enteric ganglia via inappropriate lineage segregation and consequences for gastrointestinal function. Dev Biol 2013; 382:356-64. [PMID: 23376538 DOI: 10.1016/j.ydbio.2013.01.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/21/2013] [Accepted: 01/22/2013] [Indexed: 01/28/2023]
Abstract
Normal enteric nervous system (ENS) development relies on numerous factors, including appropriate migration, proliferation, differentiation, and maturation of neural crest (NC) derivatives. Incomplete rostral to caudal migration of enteric neural crest-derived progenitors (ENPs) down the gut is at least partially responsible for the absence of enteric ganglia that is a hallmark feature of Hirschsprung disease (HSCR). The thought that ganglia proximal to aganglionosis are normal has guided surgical procedures for HSCR patients. However, chronic gastrointestinal dysfunction suffered by a subset of patients after surgery as well as studies in HSCR mouse models suggest that aberrant NC segregation and differentiation may be occurring in ganglionated regions of the intestine. Studies in mouse models that possess enteric ganglia throughout the length of the intestine (non-HSCR) have also found that certain genetic alterations affect neural crest lineage balance and interestingly many of these mutants also have functional gastrointestinal (GI) defects. It is possible that many GI disorders can be explained in part by imbalances in NC-derived lineages. Here we review studies evaluating ENS defects in HSCR and non-HSCR mouse models, concluding with clinical implications while highlighting areas requiring further study.
Collapse
Affiliation(s)
- Melissa A Musser
- Division of Genetic Medicine, Department of Medicine and the PhD Program in Human Genetics, Center for Human Genetic Research, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | |
Collapse
|
35
|
Liu H, Jia D, Li A, Chau J, He D, Ruan X, Liu F, Li J, He L, Li B. p53 regulates neural stem cell proliferation and differentiation via BMP-Smad1 signaling and Id1. Stem Cells Dev 2013. [PMID: 23199293 DOI: 10.1089/scd.2012.0370] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neural stem cells (NSCs) play essential roles in nervous system development and postnatal neuroregeneration and their deregulation underlies the development of neurodegenerative disorders. Yet how NSC proliferation and differentiation are controlled is not fully understood. Here we present evidence that tumor suppressor p53 regulates NSC proliferation and differentiation via the bone morphogenetic proteins (BMP)-Smad1 pathway and its target gene inhibitor of DNA binding 1 (Id1). p53 deficiency led to increased neurogenesis in vivo, and biased neuronal differentiation and augmented NSC proliferation of ex vivo NSCs. This is accompanied by elevated Smad1 expression/activation in the brain and NSC, which contributes to accelerated neuronal differentiation of p53(-/-) NSCs. p53 deficiency also leads to upregulation of Id1, whose expression is repressed by p53 in BMP-Smad1-dependent and -independent manners. Elevated Id1 expression contributes to augmented proliferation and, unexpectedly, accelerated neuronal differentiation of p53(-/-) NSCs as well. This study reveals a molecular mechanism by which tumor suppressor p53 controls NSC proliferation and differentiation and establishes a connection between p53 and Id1.
Collapse
Affiliation(s)
- Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Rodríguez-Martínez G, Molina-Hernández A, Velasco I. Activin A promotes neuronal differentiation of cerebrocortical neural progenitor cells. PLoS One 2012; 7:e43797. [PMID: 22928036 PMCID: PMC3425505 DOI: 10.1371/journal.pone.0043797] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 07/26/2012] [Indexed: 02/06/2023] Open
Abstract
Background Activin A is a protein that participates principally in reproductive functions. In the adult brain, Activin is neuroprotective, but its role in brain development is still elusive. Methodology/Principal Findings We studied if Activin A influences proliferation, differentiation or survival in rat cerebrocortical neural progenitor cells (NPC). After stimulation of NPC with Activin A, phosphorylation and nuclear translocation of Smad 2/3 were induced. In proliferating NPC, Activin produced a significant decrease in cell area and also a discrete increase in the number of neurons in the presence of the mitogen Fibroblast Growth Factor 2. The percentages of cells incorporating BrdU, or positive for the undifferentiated NPC markers Nestin and Sox2, were unchanged after incubation with Activin. In differentiating conditions, continuous treatment with Activin A significantly increased the number of neurons without affecting astroglial differentiation or causing apoptotic death. In cells cultured by extended periods, Activin treatment produced further increases in the proportion of neurons, excluding premature cell cycle exit. In clonal assays, Activin significantly increased neuronal numbers per colony, supporting an instructive role. Activin-induced neurogenesis was dependent on activation of its receptors, since incubation with the type I receptor inhibitor SB431542 or the ligand-trap Follistatin prevented neuronal differentiation. Interestingly, SB431542 or Follistatin by themselves abolished neurogenesis and increased astrogliogenesis, to a similar extent to that induced by Bone Morphogenetic Protein (BMP)4. Co-incubation of these Activin inhibitors with the BMP antagonist Dorsomorphin restored neuronal and astrocytic differentiation to control levels. Conclusions Our results show an instructive neuronal effect of Activin A in cortical NPC in vitro, pointing out to a relevant role of this cytokine in the specification of NPC towards a neuronal phenotype.
Collapse
Affiliation(s)
| | - Anayansi Molina-Hernández
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México, México, México
| | - Iván Velasco
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México, México, México
- * E-mail:
| |
Collapse
|
37
|
Sakaki-Yumoto M, Katsuno Y, Derynck R. TGF-β family signaling in stem cells. Biochim Biophys Acta Gen Subj 2012; 1830:2280-96. [PMID: 22959078 DOI: 10.1016/j.bbagen.2012.08.008] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 07/11/2012] [Accepted: 08/07/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND The diversity of cell types and tissue types that originate throughout development derives from the differentiation potential of embryonic stem cells and somatic stem cells. While the former are pluripotent, and thus can give rise to a full differentiation spectrum, the latter have limited differentiation potential but drive tissue remodeling. Additionally cancer tissues also have a small population of self-renewing cells with stem cell properties. These cancer stem cells may arise through dedifferentiation from non-stem cells in cancer tissues, illustrating their plasticity, and may greatly contribute to the resistance of cancers to chemotherapies. SCOPE OF REVIEW The capacity of the different types of stem cells for self-renewal, the establishment and maintenance of their differentiation potential, and the selection of differentiation programs are greatly defined by the interplay of signaling molecules provided by both the stem cells themselves, and their microenvironment, the niche. Here we discuss common and divergent roles of TGF-β family signaling in the regulation of embryonic, reprogrammed pluripotent, somatic, and cancer stem cells. MAJOR CONCLUSIONS Increasing evidence highlights the similarities between responses of normal and cancer stem cells to signaling molecules, provided or activated by their microenvironment. While TGF-β family signaling regulates stemness of normal and cancer stem cells, its effects are diverse and depend on the cell types and physiological state of the cells. GENERAL SIGNIFICANCE Further mechanistic studies will provide a better understanding of the roles of TGF-β family signaling in the regulation of stem cells. These basic studies may lead to the development of a new therapeutic or prognostic strategies for the treatment of cancers. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Masayo Sakaki-Yumoto
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, CA 94143-0669, USA
| | | | | |
Collapse
|
38
|
Expression of Bone Morphogenetic Protein-4 in the Cortical Lesions of Focal Cortical Dysplasia IIb and the Tuberous Sclerosis Complex. J Mol Neurosci 2012; 50:7-13. [DOI: 10.1007/s12031-012-9841-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 06/11/2012] [Indexed: 10/28/2022]
|
39
|
Simone S, Cosola C, Loverre A, Cariello M, Sallustio F, Rascio F, Gesualdo L, Schena FP, Grandaliano G, Pertosa G. BMP-2 induces a profibrotic phenotype in adult renal progenitor cells through Nox4 activation. Am J Physiol Renal Physiol 2012; 303:F23-34. [PMID: 22496405 DOI: 10.1152/ajprenal.00328.2011] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adult renal progenitor cells (ARPCs) isolated from the human kidney may contribute to repair featuring acute kidney injury (AKI). Bone morphogenetic proteins (BMPs) regulate differentiation, modeling, and regeneration processes in several tissues. The aim of this study was to evaluate the biological actions of BMP-2 in ARPCs in vitro and in vivo. BMP-2 was expressed in ARPCs of normal adult human kidneys, and it was upregulated in vivo after delayed graft function (DGF) of renal transplantation, a condition of AKI. ARPCs expressed BMP receptors, suggesting their potential responsiveness to BMP-2. Incubation of ARPCs with this growth factor enhanced reactive oxygen species (ROS) production, NADPH oxidase activity, and Nox4 protein expression. In vivo, Nox4 was localized in BMP-2-expressing CD133+ cells at the tubular level after DGF. BMP-2 incubation induced α-smooth muscle actin (SMA), collagen I, and fibronectin protein expression in ARPCs. Moreover, α-SMA colocalized with CD133 in vivo after DGF. The oxidative stimulus (H2O2) induced α-SMA expression in ARPCs, while the antioxidant N-acetyl-cysteine inhibited BMP-2-induced α-SMA expression. Nox4 silencing abolished BMP-2-induced NADPH oxidase activation and myofibroblastic induction. We showed that 1) ARPCs express BMP-2, 2) this expression is increased in a model of AKI; 3) BMP-2 may induce the commitment of ARPCs toward a myofibroblastic phenotype in vitro and in vivo; and 4) this profibrotic effect is mediated by Nox4 activation. Our findings suggest a novel mechanism linking AKI with progressive renal damage.
Collapse
Affiliation(s)
- Simona Simone
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, Bari, Italy; and
| | - Carmela Cosola
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, Bari, Italy; and
| | - Antonia Loverre
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, Bari, Italy; and
| | - Marica Cariello
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, Bari, Italy; and
| | - Fabio Sallustio
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, Bari, Italy; and
| | - Federica Rascio
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, Bari, Italy; and
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, Bari, Italy; and
| | - Francesco Paolo Schena
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, Bari, Italy; and
| | | | - Giovanni Pertosa
- Department of Emergency and Organ Transplantation, University Aldo Moro of Bari, Bari, Italy; and
| |
Collapse
|
40
|
Liu S, Yin F, Fan W, Wang S, Guo XR, Zhang JN, Tian ZM, Fan M. Over-expression of BMPR-IB reduces the malignancy of glioblastoma cells by upregulation of p21 and p27Kip1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:52. [PMID: 22650359 PMCID: PMC3408360 DOI: 10.1186/1756-9966-31-52] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 05/31/2012] [Indexed: 11/23/2022]
Abstract
Background In our previous study, we detected decreased expression of phospho-Smad1/5/8 and its upstream signaling molecule, bone morphogenetic protein receptor IB subunit (BMPR-IB), in certain glioblastoma tissues, unlike normal brain tissues. In order to clarify the functional roles and mechanism of BMPR-IB in the development of glioblastoma, we studied the effects of BMPR-IB overexpression on glioblastoma cell lines in vitro and in vivo. Methods We selected glioblastoma cell lines U251, U87, SF763, which have different expression of BMPR-IB to be the research subjects. Colony formation analysis and FACS were used to detect the effects of BMPR-IB on the growth and proliferation of glioblastoma cells in vivo. Immunofluresence was used to detect the differentiation changes after BMPR-IB overexpression or knocking-down. Then we used subcutaneous and intracranial tumor models to study the effect of BMPR-IB on the growth and differentiation of glioblastoma cells in vivo. The genetic alterations involved in this process were examined by real-time PCR and western blot analysis.ed. Results and conclusion Forced BMPR-IB expression in malignant human glioma cells, which exhibit lower expression of BMPR-IB, induced the phosphorylation and nuclear localization of smad1/5/8 and arrested the cell cycle in G1. Additionally, BMPR-IB overexpression could suppress anchorage-independent growth and promote differentiation of theses glioblastoma cells. Furthermore, overexpression of BMPR-IB inhibited the growth of subcutaneous and intracranial tumor xenografts and prolonged the survival of mice injected intracranially with BMPR-IB-overexpressing glioblastoma cells. Conversely, inhibition of BMPR-IB caused SF763 malignant glioma cells, a line known to exhibit high BMPR-IB expression that does not form tumors when used for xenografts, to show increased growth and regain tumorigenicity in a nude mouse model system, ultimately shortening the survival of these mice. We also observed significant accumulation of p21 and p27kip1 proteins in response to BMPR-IB overexpression. Our study suggests that overexpression of BMPR-IB may arrest and induce the differentiation of glioblastoma cells due to upregulation of p21 and p27kip1 in vitro and that in vivo and decreased expression of BMPR-IB in human glioblastoma cells contributes to glioma tumorigenicity. BMPR-IB could represent a new potential therapeutic target for malignant human gliomas.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Neurosurgery, Navy General Hospital, 100048 Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lei ZN, Liu F, Zhang LM, Huang YL, Sun FY. Bcl-2 increases stroke-induced striatal neurogenesis in adult brains by inhibiting BMP-4 function via activation of β-catenin signaling. Neurochem Int 2012; 61:34-42. [PMID: 22521772 DOI: 10.1016/j.neuint.2012.04.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 03/15/2012] [Accepted: 04/05/2012] [Indexed: 01/13/2023]
Abstract
Our previous experiments suggest that treatment with Bcl-2 increases proliferation and differentiation of neuronal progenitors induced by ischemic injury and ameliorates neurological functional deficits after stroke. However, in addition to its traditional anti-apoptotic effect, little is known about the concrete molecular modulation mechanism. In this study, Bcl-2-expressing plasmids were injected into the lateral ventricle of rat brains immediately following a 30-min occlusion of the middle cerebral artery to determine the role of Bcl-2 in adult neurogenesis. Bcl-2 overexpression reduced ischemic infarct and astrogenesis, and enhanced ischemia-induced striatal neurogenesis. We further found that Bcl-2 increased β-catenin, a key mediator of canonical Wnt/β-catenin signaling pathway, and reduced bone morphogenetic proteins-4 (BMP-4) expression in the ipsilateral striatum following ischemia. Treatment of stroke with β-catenin siRNA (i.c.v.) showed that β-catenin siRNA antagonized Bcl-2 neuroprotection against ischemic brain injury. More interestingly, β-catenin siRNA simultaneously abolished Bcl-2-mediated reduction of BMP-4 expression and enhancement of neurogenesis in the ipsilateral striatum. This effect is independent of Noggin, the known BMP antagonist. These findings highlight a new regulatory mechanism that Bcl-2 elevates ischemia-induced striatal neurogenesis by down-regulating expression of BMP-4 via activation of the Wnt/β-catenin signaling pathway in adult rat brains.
Collapse
Affiliation(s)
- Zhi-Nian Lei
- Department of Neurobiology and Institute for Biomedical Science, State Key Laboratory of Medical Neurobiology, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | | | | | | | | |
Collapse
|
42
|
Mi HW, Lee MC, Chiang YC, Chow LP, Lin CP. Single-Molecule Imaging of Bmp4 Dimerization on Human Periodontal Ligament Cells. J Dent Res 2011; 90:1318-24. [DOI: 10.1177/0022034511418340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We expressed bone morphogenetic protein 4 (BMP4) fused with enhanced green fluorescent protein (BMP4-EGFP) in the secretory pathways of producer cells. Fluorescent EGFP was acquired only after we interrupted the transport of BMP4-EGFP by culturing cells at a lower temperature (20°C), and the dynamics of BMP4-EGFP could be monitored by single-molecule microscopy. Western blotting analysis confirmed that exposure to low temperature helped the integrated formation of BMP4-EGFP fusion proteins. In this study, for the first time, we could image the fluorescently labeled BMP4 molecules localized on the plasma membrane of living hPDL cells. The one-step photobleaching with EGFP and the “blinking” behavior of quantum dots suggest that the fluorescent spots represent the events of single BMP4 molecules. Single-molecule tracking showed that the BMP receptors (BMPR) dimerize after BMP4 stimulation, or that a complex of one BMP4 molecule and a pre-formed BMPR dimer develops first, followed by the binding of the second BMP4 molecule. Furthermore, BMP4-EGFP enhanced the osteogenic differentiation of hPDL cells via signal transduction involving BMP receptors. This single-molecule imaging technique might be a valuable tool for the future development of BMP4 gene therapy and regenerative medicine mediated by hPDLs. Abbreviations: BMP4, bone morphogenetic protein 4; BMPR, BMP receptor; EGFP, enhanced green fluorescent protein; hPDL cells, human periodontal ligament cells; QDs, quantum dots; TIRFM, total internal reflection fluorescence microscopy; 293 cells, human embryonic kidney cells; oDM, osteogenic differentiation medium; HcoI, type I collagen; ALP, alkaline phosphatase; BSP, bone sialoprotein; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.
Collapse
Affiliation(s)
- H.-W. Mi
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University and National Taiwan University Hospital, No.1, Changde St., Jhongjheng District, Taipei 100, Taiwan, ROC
| | - M.-C. Lee
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Y.-C. Chiang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University and National Taiwan University Hospital, No.1, Changde St., Jhongjheng District, Taipei 100, Taiwan, ROC
| | - L.-P. Chow
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - C.-P. Lin
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University and National Taiwan University Hospital, No.1, Changde St., Jhongjheng District, Taipei 100, Taiwan, ROC
| |
Collapse
|
43
|
Virtanen S, Alarmo EL, Sandström S, Ampuja M, Kallioniemi A. Bone morphogenetic protein -4 and -5 in pancreatic cancer--novel bidirectional players. Exp Cell Res 2011; 317:2136-46. [PMID: 21704030 DOI: 10.1016/j.yexcr.2011.06.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 06/08/2011] [Accepted: 06/09/2011] [Indexed: 11/19/2022]
Abstract
Bone morphogenetic proteins (BMPs) are multifunctional signaling molecules that have gained increasing interest in cancer research. To obtain a systematic view on BMP signaling in pancreatic cancer we first determined the mRNA expression levels of seven BMP ligands (BMP2-BMP8) and six BMP specific receptors in pancreatic cancer cell lines and normal pancreatic tissue. BMP receptor expression was seen in all cancer and normal samples. Low expression levels of BMP5 and BMP8 were detected in cancer cells compared to the normal samples, whereas BMP4 expression was elevated in 25% of the cases. The impact of BMP4 and BMP5 signaling on cell phenotype was then evaluated in five pancreatic cancer cell lines. Both ligands suppressed the growth of three cell lines (up to 79% decrease in BMP4-treated PANC-1 cells), mainly due to cell cycle changes. BMP4 and BMP5 concurrently increased cell migration and invasion (maximally a 10.8-fold increase in invaded BMP4-treated PANC-1 cells). The phenotypic changes were typically associated with the activation of the canonical SMAD pathway, although such activation was not observed in the PANC-1 cells. Taken together, BMP4 and BMP5 simultaneously inhibit the growth and promote migration and invasion of the same pancreatic cells and thus exhibit a biphasic role with both detrimental and beneficial functions in pancreatic cancer progression.
Collapse
Affiliation(s)
- Siru Virtanen
- Laboratory of Cancer Genetics, Institute of Medical Technology, University of Tampere and Tampere University Hospital, Finland.
| | | | | | | | | |
Collapse
|
44
|
Abstract
OLs (oligodendrocytes) are the myelinating cells of the CNS (central nervous system), wrapping axons in conductive sheathes to ensure effective transmission of neural signals. The regulation of OL development, from precursor to mature myelinating cell, is controlled by a variety of inhibitory and inductive signalling factors. The dorsal spinal cord contains signals that inhibit OL development, possibly to prevent premature and ectopic precursor differentiation. The Wnt and BMP (bone morphogenic protein) signalling pathways have been identified as dorsal spinal cord signals with overlapping temporal activity, and both have similar inhibitory effects on OL differentiation. Both these pathways feature prominently in many developmental processes and demyelinating events after injury, and they are known to interact in complex inductive, inhibitive and synergistic manners in many developing systems. The interaction between BMP and Wnt signalling in OL development, however, has not been extensively explored. In the present study, we examine the relationship between the canonical Wnt and BMP pathways. We use pharmacological and genetic paradigms to show that both Wnt3a and BMP4 will inhibit OL differentiation in vitro. We also show that when the canonical BMP signalling pathway is blocked, neither Wnt3a nor BMP4 have inhibitory effects on OL differentiation. In contrast, abrogating the Wnt signalling pathway does not alter the actions of BMP4 treatment. Our results indicate that the BMP signalling pathway is necessary for the canonical Wnt signalling pathway to exert its effects on OL development, but not vice versa, suggesting that Wnt signals upstream of BMP.
Collapse
|
45
|
Wang Q, Huang C, Xue M, Zhang X. Expression of endogenous BMP-2 in periosteal progenitor cells is essential for bone healing. Bone 2011; 48:524-32. [PMID: 21056707 PMCID: PMC3039040 DOI: 10.1016/j.bone.2010.10.178] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 10/27/2010] [Accepted: 10/27/2010] [Indexed: 11/19/2022]
Abstract
Bone morphogenic protein 2 (BMP-2) plays a key role in skeletal development, repair and regeneration. To gain a better understanding of the role of BMP-2 in periosteum-mediated bone repair, we deleted BMP-2 postnatally at the initiation stage of healing utilizing a Tamoxifen-inducible CreER mouse model. To mark the mutant cells, we further generated a BMP-2(f/f); CreER; RosaR mouse model that enabled the activation of a LacZ reporter gene upon treatment of Tamoxifen. We demonstrated that deletion of BMP-2 at the onset of healing abolished periosteum-mediated bone/cartilage callus formation. In a chimeric periosteal callus with cells derived from both wild type and the mutant, over 90% of the mutant mesenchymal progenitors remained undifferentiated. Within differentiated bone and cartilage tissues, only a few cells could be identified as mutants. Using a bone graft transplantation approach, we further showed that transplantation of a mutant bone graft into a wild type host failed to rescue the deficient differentiation of the mutant cells at day 10 post-grafting. These data strongly suggest that the endogenous expression of BMP-2 plays a critical role in osteogenic and chondrogenic differentiation of periosteal progenitors during repair. To determine whether BMP-2 deficient cells remained responsive to exogenous BMP-2, we isolated periosteal mesenchymal progenitors from BMP-2 deficient bone autografts. The isolated cells demonstrated a 90% reduction of endogenous BMP-2 expression, accompanied by significant decrease in cellular proliferation and a near blockade of osteogenic differentiation. The addition of exogenous BMP-2 partially rescued impaired proliferation and further enhanced osteogenic differentiation in a dose dependent manner. Taken together, our data show that the initiation of the cortical bone repair in vivo is controlled by endogenous BMP-2. Future studies are necessary to determine the mechanisms by which the BMP-2 pathway is activated in periosteal progenitor cells at the onset of cortical bone repair.
Collapse
Affiliation(s)
- Qun Wang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY
| | - Chunlan Huang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY
| | - Ming Xue
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
46
|
Mi HW, Lee MC, Fu E, Chow LP, Lin CP. Highly efficient multipotent differentiation of human periodontal ligament fibroblasts induced by combined BMP4 and hTERT gene transfer. Gene Ther 2011; 18:452-61. [DOI: 10.1038/gt.2010.158] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
Li J, Zhang B, Han H, Cao Z, Lian Z, Li N. Metabolic properties of chicken embryonic stem cells. SCIENCE CHINA-LIFE SCIENCES 2010; 53:1073-84. [PMID: 21104367 DOI: 10.1007/s11427-010-4055-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 04/19/2010] [Indexed: 11/26/2022]
Abstract
Cellular energy metabolism correlates with cell fate, but the metabolic properties of chicken embryonic stem (chES) cells are poorly understood. Using a previously established chES cell model and electron microscopy (EM), we found that undifferentiated chES cells stored glycogen. Additionally, undifferentiated chES cells expressed lower levels of glucose transporter 1 (GLUT1) and phosphofructokinase (PFK) mRNAs but higher levels of hexokinase 1 (HK1) and glycogen synthase (GYS) mRNAs compared with control primary chicken embryonic fibroblast (CEF) cells, suggesting that chES cells direct glucose flux towards the glycogenic pathway. Moreover, we demonstrated that undifferentiated chES cells block gluconeogenic outflow and impede the accumulation of glucose-6-phosphate (G6P) from this pathway, as evidenced by the barely detectable levels of pyruvate carboxylase (PCX) and mitochondrial phosphoenolpyruvate carboxykinase (PCK2) mRNAs. Additionally, cell death occurred in undifferentiated chES cells as shown by Hoechst 33342 and propidium iodide (PI) double staining, but it could be rescued by exogenous G6P. However, we found that differentiated chES cells decreased the glycogen reserve through the use of PAS staining. Moreover, differentiated chES cells expressed higher levels of GLUT1, HK1 and PFK mRNAs, while the level of GYS mRNA remained similar in control CEF cells. These data indicate that undifferentiated chES cells continue to synthesize glycogen from glucose at the expense of G6P, while differentiated chES cells have a decreased glycogen reserve, which suggests that the amount of glycogen is indicative of the chES cell state.
Collapse
Affiliation(s)
- Jia Li
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100194, China
| | | | | | | | | | | |
Collapse
|
48
|
Vicente López MA, Vázquez García MN, Entrena A, Olmedillas Lopez S, García-Arranz M, García-Olmo D, Zapata A. Low doses of bone morphogenetic protein 4 increase the survival of human adipose-derived stem cells maintaining their stemness and multipotency. Stem Cells Dev 2010; 20:1011-9. [PMID: 20846028 DOI: 10.1089/scd.2010.0355] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as important tools for cell therapy; therefore, identification of factors capable of governing their ex vivo expansion become essential. In this study we demonstrate that human adipose-derived stem cells (ASCs) express all components of the bone morphogenetic protein (BMP)/BMP receptor signaling pathway and respond to BMP4 inducing upregulated expression of its specific target genes Id1-Id4. Moreover, ASCs grown in a medium reduced in serum produce endogenous BMP4 that could affect autocrinely ASC growth. On the contrary, dorsomorphin, an inhibitor of BMP signaling pathway, decreases cell numbers yielded from ASC cultures in correlation with increased apoptosis and decreased cycling cells. Therefore, BMP4 emerges as a possible factor for ex vivo expanding human ASCs. Our results demonstrate that, as other morphogens, BMP4 effects on human MSCs are dose dependent. High doses significantly increased apoptosis and drastically reduced cell proliferation, whereas low doses of BMP4 (0.01-0.1 ng/mL) significantly increase culture cell content, reduce the number of apoptotic cells, and increase that of cycling cells. Further, treatment of human ASCs with low doses of BMP4 does not modify expression of Nanog and Oct4, two transcription factors involved in self-renewal and pluripotency of stem cells or avoid their osteogenic or osteoblastic differentiation capacities when cultured in adequate inducing media, as shown by the induction of specific gene expression (CEBP, PPARγ, and RUNX2). Our results therefore support BMP4 as a promising factor for expanding human adipose tissue-derived MSCs maintaining their properties of stemness and multipotency.
Collapse
Affiliation(s)
- María A Vicente López
- Department of Cell Biology, School of Medicine, Complutense University, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
49
|
Mira H, Andreu Z, Suh H, Lie DC, Jessberger S, Consiglio A, San Emeterio J, Hortigüela R, Marqués-Torrejón MA, Nakashima K, Colak D, Götz M, Fariñas I, Gage FH. Signaling through BMPR-IA regulates quiescence and long-term activity of neural stem cells in the adult hippocampus. Cell Stem Cell 2010; 7:78-89. [PMID: 20621052 DOI: 10.1016/j.stem.2010.04.016] [Citation(s) in RCA: 366] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 11/13/2009] [Accepted: 04/13/2010] [Indexed: 12/27/2022]
Abstract
Neural stem cells (NSCs) in the adult hippocampus divide infrequently, and the molecules that modulate their quiescence are largely unknown. Here, we show that bone morphogenetic protein (BMP) signaling is active in hippocampal NSCs, downstream of BMPR-IA. BMPs reversibly diminish proliferation of cultured NSCs while maintaining their undifferentiated state. In vivo, acute blockade of BMP signaling in the hippocampus by intracerebral infusion of Noggin first recruits quiescent NSCs into the cycle and increases neurogenesis; subsequently, it leads to decreased stem cell division and depletion of precursors and newborn neurons. Consistently, selective ablation of Bmpr1a in hippocampal NSCs, or inactivation of BMP canonical signaling in conditional Smad4 knockout mice, transiently enhances proliferation but later leads to a reduced number of precursors, thereby limiting neuronal birth. BMPs are therefore required to balance NSC quiescence/proliferation and to prevent loss of the stem cell activity that supports continuous neurogenesis in the mature hippocampus.
Collapse
Affiliation(s)
- Helena Mira
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Id gene regulation and function in the prosensory domains of the chicken inner ear: a link between Bmp signaling and Atoh1. J Neurosci 2010; 30:11426-34. [PMID: 20739564 DOI: 10.1523/jneurosci.2570-10.2010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Bone morphogenetic proteins (Bmps) regulate the expression of the proneural gene Atoh1 and the generation of hair cells in the developing inner ear. The present work explored the role of Inhibitor of Differentiation genes (Id1-3) in this process. The results show that Id genes are expressed in the prosensory domains of the otic vesicle, along with Bmp4 and Bmp7. Those domains exhibit high levels of the phosphorylated form of Bmp-responding R-Smads (P-Smad1,5,8), and of Bmp-dependent Smad transcriptional activity as shown by the BRE-tk-EGFP reporter. Increased Bmp signaling induces the expression of Id1-3 along with the inhibition of Atoh1. Conversely, the Bmp antagonist Noggin or the Bmp-receptor inhibitor Dorsomorphin elicit opposite effects, indicating that Bmp signaling is necessary for Id expression and Atoh1 regulation in the otocyst. The forced expression of Id3 is sufficient to reduce Atoh1 expression and to prevent the expression of hair cell differentiation markers. Together, these results suggest that Ids are part of the machinery that mediates the regulation of hair cell differentiation exerted by Bmps. In agreement with that, during hair cell differentiation Bmp4 expression, P-Smad1,5,8 levels and Id expression are downregulated from hair cells. However, Ids are also downregulated from the supporting cells which contrarily to hair cells exhibit high levels of Bmp4 expression, P-Smad1,5,8, and BRE-tk-EGFP activity, suggesting that in these cells Ids escape from Bmp/Smad signaling. The differential regulation of Ids in time and space may underlie the multiple functions of Bmp signaling during sensory organ development.
Collapse
|