1
|
Urbach A, Witte OW. Divide or Commit - Revisiting the Role of Cell Cycle Regulators in Adult Hippocampal Neurogenesis. Front Cell Dev Biol 2019; 7:55. [PMID: 31069222 PMCID: PMC6491688 DOI: 10.3389/fcell.2019.00055] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 03/28/2019] [Indexed: 12/21/2022] Open
Abstract
The adult dentate gyrus continuously generates new neurons that endow the brain with increased plasticity, helping to cope with changing environmental and cognitive demands. The process leading to the birth of new neurons spans several precursor stages and is the result of a coordinated series of fate decisions, which are tightly controlled by extrinsic signals. Many of these signals act through modulation of cell cycle (CC) components, not only to drive proliferation, but also for linage commitment and differentiation. In this review, we provide a comprehensive overview on key CC components and regulators, with emphasis on G1 phase, and analyze their specific functions in precursor cells of the adult hippocampus. We explore their role for balancing quiescence versus self-renewal, which is essential to maintain a lifelong pool of neural stem cells while producing new neurons “on demand.” Finally, we discuss available evidence and controversies on the impact of CC/G1 length on proliferation versus differentiation decisions.
Collapse
Affiliation(s)
- Anja Urbach
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| |
Collapse
|
2
|
Temerozo JR, de Azevedo SSD, Insuela DBR, Vieira RC, Ferreira PLC, Carvalho VF, Bello G, Bou-Habib DC. The Neuropeptides Vasoactive Intestinal Peptide and Pituitary Adenylate Cyclase-Activating Polypeptide Control HIV-1 Infection in Macrophages Through Activation of Protein Kinases A and C. Front Immunol 2018; 9:1336. [PMID: 29951068 PMCID: PMC6008521 DOI: 10.3389/fimmu.2018.01336] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/29/2018] [Indexed: 12/19/2022] Open
Abstract
Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are highly similar neuropeptides present in several tissues, endowed with immunoregulatory functions and other systemic effects. We previously reported that both neuropeptides reduce viral production in HIV-1-infected primary macrophages, with the participation of β-chemokines and IL-10, and now we describe molecular mechanisms engaged in this activity. Macrophages exposed to VIP or PACAP before HIV-1 infection showed resistance to viral replication, comparable to that observed when the cells were treated after infection. Also, multiple treatments with a suboptimal dose of VIP or PACAP after macrophage infection resulted in a decline of virus production similar to the inhibition promoted by a single exposure to the optimal inhibitory concentration. Cellular signaling pathways involving cAMP production and activation of protein kinases A and C were critical components of the VIP and PACAP anti-HIV-1 effects. Analysis of the transcription factors and the transcriptional/cell cycle regulators showed that VIP and PACAP induced cAMP response element-binding protein activation, inhibited NF-kB, and reduced Cyclin D1 levels in HIV-1-infected cells. Remarkably, VIP and PACAP promoted G-to-A mutations in the HIV-1 provirus, matching those derived from the activity of the APOBEC family of viral restriction factors, and reduced viral infectivity. In conclusion, our findings strengthen the antiretroviral potential of VIP and PACAP and point to new therapeutic approaches to control the progression of HIV-1 infection.
Collapse
Affiliation(s)
- Jairo R Temerozo
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Suwellen S D de Azevedo
- Laboratory of AIDS and Molecular Immunology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Daniella B R Insuela
- Laboratory of Inflammation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Rhaíssa C Vieira
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Pedro L C Ferreira
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Vinícius F Carvalho
- National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil.,Laboratory of Inflammation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Gonzalo Bello
- Laboratory of AIDS and Molecular Immunology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Ru Y, Chen XJ, Zhao ZW, Zhang PF, Feng SH, Gao Q, Gao SG, Feng XS. CyclinD1 and p57 kip2 as biomarkers in differentiation, metastasis and prognosis of gastric cardia adenocarcinoma. Oncotarget 2017; 8:73860-73870. [PMID: 29088752 PMCID: PMC5650307 DOI: 10.18632/oncotarget.18008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/19/2017] [Indexed: 12/26/2022] Open
Abstract
Objective This study aims to investigate the expression and significance of p57kip2 and cyclinD1 in gastric cardia adenocarcinoma (GCA). p57kip2 is a negative regulator in the cell cycle. On the contrary, cyclinD1 is a positive regulator of cell cycle progression. Methods Thirty-two cases of GCA tissues and adjacent non-cancerous tissues were collected for this study. Immunohistochemistry and fluorescence qualitative PCR was used to determine the level of p57kip2 and cyclinD1 in GCA and its adjacent non-cancerous tissues. Furthermore, the correlation between the mRNA/protein and GCA clinical pathologic parameters were analyzed, and the relationship of p57kip2 and cyclinD1 in GCA were also evaluated. Results The expression of p57kip2 significantly lower in GCA (P = 0.036), and there was a significant correlation in the different degrees of differentiation (P < 0.05). Furthermore, median survival time was 41 months for patients with high mRNA expression of p57kip2. This was longer compared to patients with low mRNA expression of P57kip2 (37 months, X2 = 4.788, P = 0.029).The expression of cyclinD1 was significantly higher in GCA(P = 0.002), and was significant correlated to clinical stage(P<0.05). Median survival time was 34 months in patients with high mRNA expression of cyclinD1, which was shorter than in patients with low expression of cyclinD1 mRNA (41 months, X2 = 4.071, P = 0.044). The protein expression of p57kip2 was not correlated to the protein expression of cyclinD1 (P = 0.55). Conclusion The expression of p57kip2 and cyclinD1 are likely to suppress or promote the tumorigenesis and progression of GCA.
Collapse
Affiliation(s)
- Yi Ru
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology , Luoyang, Henan 471003, China
| | - Xiao-Jie Chen
- Medical College, Henan University of Science and Technology , Luoyang, Henan 471003, China
| | - Zhi-Wei Zhao
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology , Luoyang, Henan 471003, China
| | - Peng-Fei Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology , Luoyang, Henan 471003, China
| | - Shuai-Hao Feng
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology , Luoyang, Henan 471003, China
| | - Qiang Gao
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology , Luoyang, Henan 471003, China
| | - She-Gan Gao
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology , Luoyang, Henan 471003, China.,Medical College, Henan University of Science and Technology , Luoyang, Henan 471003, China.,Henan Key Laboratory of Cancer Epigenetics, Henan University of Science and Technology , Luoyang, Henan 471003, China.,Cancer Institute, Henan University of Science and Technology , Luoyang, Henan 471003, China
| | - Xiao-Shan Feng
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology , Luoyang, Henan 471003, China.,Henan Key Laboratory of Cancer Epigenetics, Henan University of Science and Technology , Luoyang, Henan 471003, China.,Cancer Institute, Henan University of Science and Technology , Luoyang, Henan 471003, China
| |
Collapse
|
4
|
Chung H, Park S. Ghrelin regulates cell cycle-related gene expression in cultured hippocampal neural stem cells. J Endocrinol 2016; 230:239-50. [PMID: 27325242 DOI: 10.1530/joe-16-0126] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 01/09/2023]
Abstract
We have previously demonstrated that ghrelin stimulates the cellular proliferation of cultured adult rat hippocampal neural stem cells (NSCs). However, little is known about the molecular mechanisms by which ghrelin regulates cell cycle progression. The purpose of this study was to investigate the potential effects of ghrelin on cell cycle regulatory molecules in cultured hippocampal NSCs. Ghrelin treatment increased proliferation assessed by CCK-8 proliferation assay. The expression levels of proliferating cell nuclear antigen and cell division control 2, well-known cell-proliferating markers, were also increased by ghrelin. Fluorescence-activated cell sorting analysis revealed that ghrelin promoted progression of cell cycle from G0/G1 to S phase, whereas this progression was attenuated by the pretreatment with specific inhibitors of MEK/extracellular signal-regulated kinase 1/2, phosphoinositide 3-kinase/Akt, mammalian target of rapamycin, and janus kinase 2/signal transducer and activator of transcription 3. Ghrelin-induced proliferative effect was associated with increased expression of E2F1 transcription factor in the nucleus, as determined by Western blotting and immunofluorescence. We also found that ghrelin caused an increase in protein levels of positive regulators of cell cycle, such as cyclin A and cyclin-dependent kinase (CDK) 2. Moreover, p27(KIP1) and p57(KIP2) protein levels were reduced when cell were exposed to ghrelin, suggesting downregulation of CDK inhibitors may contribute to proliferative effect of ghrelin. Our data suggest that ghrelin targets both cell cycle positive and negative regulators to stimulate proliferation of cultured hippocampal NSCs.
Collapse
Affiliation(s)
- Hyunju Chung
- Department of Core Research LaboratoryClinical Research Institute, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Seungjoon Park
- Department of Pharmacology and Medical Research Center for Bioreaction to ROS and Biomedical Science InstituteSchool of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
5
|
Lee HJ, Dreyfus C, DiCicco-Bloom E. Valproic acid stimulates proliferation of glial precursors during cortical gliogenesis in developing rat. Dev Neurobiol 2015; 76:780-98. [PMID: 26505176 DOI: 10.1002/dneu.22359] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 10/16/2015] [Accepted: 10/23/2015] [Indexed: 11/06/2022]
Abstract
Valproic acid (VPA) is a neurotherapeutic drug prescribed for seizures, bipolar disorder, and migraine, including women of reproductive age. VPA is a well-known teratogen that produces congenital malformations in many organs including the nervous system, as well as later neurodevelopmental disorders, including mental retardation and autism. In developing brain, few studies have examined VPA effects on glial cells, particularly astrocytes. To investigate effects on primary glial precursors, we developed new cell culture and in vivo models using frontal cerebral cortex of postnatal day (P2) rat. In vitro, VPA exposure elicited dose-dependent, biphasic effects on DNA synthesis and proliferation. In vivo VPA (300 mg/kg) exposure from P2 to P4 increased both DNA synthesis and cell proliferation, affecting primarily astrocyte precursors, as >75% of mitotic cells expressed brain lipid-binding protein. Significantly, the consequence of early VPA exposure was increased astrocytes, as both S100-β+ cells and glial fibrillary acidic protein were increased in adolescent brain. Molecularly, VPA served as an HDAC inhibitor in vitro and in vivo as enhanced proliferation was accompanied by increased histone acetylation, whereas it elicited changes in culture in cell-cycle regulators, including cyclin D1 and E, and cyclin-dependent kinase (CDK) inhibitors, p21 and p27. Collectively, these data suggest clinically relevant VPA exposures stimulate glial precursor proliferation, though at higher doses can elicit inhibition through differential regulation of CDK inhibitors. Because changes in glial cell functions are proposed as mechanisms contributing to neuropsychiatric disorders, these observations suggest that VPA teratogenic actions may be mediated through changes in astrocyte generation during development. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 780-798, 2016.
Collapse
Affiliation(s)
- Hee Jae Lee
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, Piscataway, New Jersey.,Department of Pharmacology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Cheryl Dreyfus
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, Piscataway, New Jersey
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, Piscataway, New Jersey.,Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
6
|
Hoffmann A, Daniel G, Schmidt-Edelkraut U, Spengler D. Roles of imprinted genes in neural stem cells. Epigenomics 2015; 6:515-32. [PMID: 25431944 DOI: 10.2217/epi.14.42] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Imprinted genes and neural stem cells (NSC) play an important role in the developing and mature brain. A central theme of imprinted gene function in NSCs is cell survival and G1 arrest to control cell division, cell-cycle exit, migration and differentiation. Moreover, genomic imprinting can be epigenetically switched off at some genes to ensure stem cell quiescence and differentiation. At the genome scale, imprinted genes are organized in dynamic networks formed by interchromosomal interactions and transcriptional coregulation of imprinted and nonimprinted genes. Such multilayered networks may synchronize NSC activity with the demand from the niche resembling their roles in adjusting fetal size.
Collapse
Affiliation(s)
- Anke Hoffmann
- Max Planck Institute of Psychiatry, Translational Research, Kraepelinstrasse 2-10, 80804 Munich, Germany
| | | | | | | |
Collapse
|
7
|
Njaine B, Rocha-Martins M, Vieira-Vieira CH, De-Melo LDB, Linden R, Braas K, May V, Martins RAP, Silveira MS. Pleiotropic functions of pituitary adenylyl cyclase-activating polypeptide on retinal ontogenesis: involvement of KLF4 in the control of progenitor cell proliferation. J Mol Neurosci 2014; 54:430-42. [PMID: 24715357 DOI: 10.1007/s12031-014-0299-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/27/2014] [Indexed: 12/11/2022]
Abstract
We showed previously that the neuropeptide pituitary adenylyl cyclase-activating polypeptide (PACAP) negatively regulates proliferation of postnatal rat retinal progenitor cells through the downregulation of cyclin D1 in a cAMP/protein kinase A dependent manner. In the present study, we describe by microarray analysis several putative PACAP targets regulated by different transcription factor families. One of these families is the Sp/Klf family of transcriptional factors capable of regulating cyclin D1, and among members, we demonstrate by immunocytochemistry that KLF4 is expressed throughout rat retinal development by retinal progenitor cells and in most differentiated cell types. Using retinal explants preparations, PACAP treatment can transiently increase Klf4 mRNA levels; from electrophoretic mobility shift assays, PACAP is also able to increase the nuclear KLF4 content. From these results, we suggest that KLF4 may be involved in the anti-proliferative effects of PACAP as one mechanism regulating progenitor cell transition from proliferation to differentiation throughout retinal development.
Collapse
Affiliation(s)
- Brian Njaine
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Edifício do Centro de Ciencias da Saude, Bloco G, Cidade Universitaria, Ilha do Fundão, 21941-902, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Rossman IT, Lin L, Morgan KM, Digiovine M, Van Buskirk EK, Kamdar S, Millonig JH, Dicicco-Bloom E. Engrailed2 modulates cerebellar granule neuron precursor proliferation, differentiation and insulin-like growth factor 1 signaling during postnatal development. Mol Autism 2014; 5:9. [PMID: 24507165 PMCID: PMC3932947 DOI: 10.1186/2040-2392-5-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 01/14/2014] [Indexed: 01/16/2023] Open
Abstract
Background The homeobox transcription factor Engrailed2 (En2) has been studied extensively in neurodevelopment, particularly in the midbrain/hindbrain region and cerebellum, where it exhibits dynamic patterns of expression and regulates cell patterning and morphogenesis. Because of its roles in regulating cerebellar development and evidence of cerebellar pathology in autism spectrum disorder (ASD), we previously examined an ENGRAILED2 association and found evidence to support EN2 as a susceptibility gene, a finding replicated by several other investigators. However, its functions at the cell biological level remain undefined. In the mouse, En2 gene is expressed in granule neuron precursors (GNPs) just as they exit the cell cycle and begin to differentiate, raising the possibility that En2 may modulate these developmental processes. Methods To define En2 functions, we examined proliferation, differentiation and signaling pathway activation in En2 knockout (KO) and wild-type (WT) GNPs in response to a variety of extracellular growth factors and following En2 cDNA overexpression in cell culture. In vivo analyses of cerebellar GNP proliferation as well as responses to insulin-like growth factor-1 (IGF1) treatment were also conducted. Results Proliferation markers were increased in KO GNPs in vivo and in 24-h cultures, suggesting En2 normally serves to promote cell cycle exit. Significantly, IGF1 stimulated greater DNA synthesis in KO than WT cells in culture, a finding associated with markedly increased phospho-S6 kinase activation. Similarly, there was three-fold greater DNA synthesis in the KO cerebellum in response to IGF1 in vivo. On the other hand, KO GNPs exhibited reduced neurite outgrowth and differentiation. Conversely, En2 overexpression increased cell cycle exit and promoted neuronal differentiation. Conclusions In aggregate, our observations suggest that the ASD-associated gene En2 promotes GNP cell cycle exit and differentiation, and modulates IGF1 activity during postnatal cerebellar development. Thus, genetic/epigenetic alterations of EN2 expression may impact proliferation, differentiation and IGF1 signaling as possible mechanisms that may contribute to ASD pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Emanuel Dicicco-Bloom
- Department of Neuroscience & Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, 675 Hoes, Lane, Piscataway, NJ 08854, USA.
| |
Collapse
|
9
|
Huang HY, Liu DD, Chang HF, Chen WF, Hsu HR, Kuo JS, Wang MJ. Histone deacetylase inhibition mediates urocortin-induced antiproliferation and neuronal differentiation in neural stem cells. Stem Cells 2013; 30:2760-73. [PMID: 22961741 DOI: 10.1002/stem.1226] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 08/14/2012] [Indexed: 01/15/2023]
Abstract
During cortical development, cell proliferation and cell cycle exit are carefully regulated to ensure that the appropriate numbers of cells are produced. Urocortin (UCN) is a member of the corticotrophin releasing hormone (CRH) family of neuropeptides that regulates stress responses. UCN is widely distributed in adult rat brain. However, the expression and function of UCN in embryonic brain is, as yet, unclear. Here, we show that UCN is endogenously expressed in proliferative zones of the developing cerebral cortex and its receptors are exhibited in neural stem cells (NSCs), thus implicating the neuropeptide in cell cycle regulation. Treatment of cultured NSCs or organotypic slice cultures with UCN markedly reduced cell proliferation. Furthermore, blocking of endogenous UCN/CRHRs system either by treatment with CRHRs antagonists or by neutralization of secreted UCN with anti-UCN antibody increased NSCs proliferation. Cell cycle kinetics analysis demonstrated that UCN lengthened the total cell cycle duration via increasing the G1 phase and accelerated cell cycle exit. UCN directly inhibited the histone deacetylase (HDAC) activity and induced a robust increase in histone H3 acetylation levels. Using pharmacological and RNA interference approaches, we further demonstrated that antiproliferative action of UCN appeared to be mediated through a HDAC inhibition-induced p21 upregulation. Moreover, UCN treatment in vitro and in vivo led to an increase in neuronal differentiation of NSCs. These findings suggest that UCN might contribute to regulate NSCs proliferation and differentiation during cortical neurogenesis.
Collapse
Affiliation(s)
- Hsin-Yi Huang
- Department of Medical Research, Neuro-Medical Scientific Center, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | | | | | | | | | | | | |
Collapse
|
10
|
Manecka DL, Mahmood SF, Grumolato L, Lihrmann I, Anouar Y. Pituitary adenylate cyclase-activating polypeptide (PACAP) promotes both survival and neuritogenesis in PC12 cells through activation of nuclear factor κB (NF-κB) pathway: involvement of extracellular signal-regulated kinase (ERK), calcium, and c-REL. J Biol Chem 2013; 288:14936-48. [PMID: 23564451 DOI: 10.1074/jbc.m112.434597] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The pituitary adenylate cyclase-activating polypeptide (PACAP) is a trophic factor that promotes neuronal survival and neurite outgrowth. However, the signaling pathways and the transcriptional mechanisms involved are not completely elucidated. Our previous studies aimed at characterizing the transcriptome of PACAP-differentiated PC12 cells revealed an increase in the expression of nuclear factor κB2 (NF-κB2) gene coding for p100/p52 subunit of NF-κB transcription factor. Here, we examined the role of the NF-κB pathway in neuronal differentiation promoted by PACAP. We first showed that PACAP-driven survival and neuritic extension in PC12 cells are inhibited following NF-κB pathway blockade. PACAP stimulated both c-Rel and p52 NF-κB subunit gene expression and nuclear translocation, whereas c-Rel down-regulation inhibited cell survival and neuritogenesis elicited by the neuropeptide. PACAP-induced c-Rel nuclear translocation was inhibited by ERK1/2 and Ca(2+) blockers. Furthermore, the neuropeptide stimulated NF-κB p100 subunit processing into p52, indicative of activation of the NF-κB alternative pathway. Taken together, our data show that PACAP promotes both survival and neuritogenesis in PC12 cells by activating NF-κB pathway, most likely via classical and alternative signaling cascades involving ERK1/2 kinases, Ca(2+), and c-Rel/p52 dimers.
Collapse
Affiliation(s)
- Destiny-Love Manecka
- INSERM, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France
| | | | | | | | | |
Collapse
|
11
|
Tury A, Mairet-Coello G, DiCicco-Bloom E. The multiple roles of the cyclin-dependent kinase inhibitory protein p57(KIP2) in cerebral cortical neurogenesis. Dev Neurobiol 2012; 72:821-42. [PMID: 22076965 DOI: 10.1002/dneu.20999] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The members of the CIP/KIP family of cyclin-dependent kinase (CDK) inhibitory proteins (CKIs), including p57(KIP2), p27(KIP1), and p21(CIP1), block the progression of the cell cycle by binding and inhibiting cyclin/CDK complexes of the G1 phase. In addition to this well-characterized function, p57(KIP2) and p27(KIP1) have been shown to participate in an increasing number of other important cellular processes including cell fate and differentiation, cell motility and migration, and cell death/survival, both in peripheral and central nervous systems. Increasing evidence over the past few years has characterized the functions of the newest CIP/KIP member p57(KIP2) in orchestrating cell proliferation, differentiation, and migration during neurogenesis. Here, we focus our discussion on the multiple roles played by p57(KIP2) during cortical development, making comparisons to p27(KIP1) as well as the INK4 family of CKIs.
Collapse
Affiliation(s)
- Anna Tury
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | | | | |
Collapse
|
12
|
Falluel-Morel A, Lin L, Sokolowski K, McCandlish E, Buckley B, DiCicco-Bloom E. N-acetyl cysteine treatment reduces mercury-induced neurotoxicity in the developing rat hippocampus. J Neurosci Res 2012; 90:743-50. [PMID: 22420031 DOI: 10.1002/jnr.22819] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mercury is an environmental toxicant that can disrupt brain development. However, although progress has been made in defining its neurotoxic effects, we know far less about available therapies that can effectively protect the brain in exposed individuals. We previously developed an animal model in which we defined the sequence of events underlying neurotoxicity: Methylmercury (MeHg) injection in postnatal rat acutely induced inhibition of mitosis and stimulated apoptosis in the hippocampus, which later resulted in intermediate-term deficits in structure size and cell number. N-acetyl cysteine (NAC) is the N-acetyl derivative of L-cysteine used clinically for treatment of drug intoxication. Here, based on its known efficacy in promoting MeHg urinary excretion, we evaluated NAC for protective effects in the developing brain. In immature neurons and precursors, MeHg (3 μM) induced a >50% decrease in DNA synthesis at 24 hr, an effect that was completely blocked by NAC coincubation. In vivo, injection of MeHg (5 μg/g bw) into 7-day-old rats induced a 22% decrease in DNA synthesis in whole hippocampus and a fourfold increase in activated caspase-3-immunoreactive cells at 24 hr and reduced total cell numbers by 13% at 3 weeks. Treatment of MeHg-exposed rats with repeated injections of NAC abolished MeHg toxicity. NAC prevented the reduction in DNA synthesis and the marked increase in caspase-3 immunoreactivity. Moreover, the intermediate-term decrease in hippocampal cell number provoked by MeHg was fully blocked by NAC. Altogether these results suggest that MeHg toxicity in the perinatal brain can be ameliorated by using NAC, opening potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Anthony Falluel-Morel
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | | | | | |
Collapse
|
13
|
Ma Y, Liu W, Wang Y, Chao X, Qu Y, Wang K, Fei Z. VEGF protects rat cortical neurons from mechanical trauma injury induced apoptosis via the MEK/ERK pathway. Brain Res Bull 2011; 86:441-6. [DOI: 10.1016/j.brainresbull.2011.07.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 07/11/2011] [Accepted: 07/12/2011] [Indexed: 02/07/2023]
|
14
|
Hirose M, Niewiadomski P, Tse G, Chi GC, Dong H, Lee A, Carpenter EM, Waschek JA. Pituitary adenylyl cyclase-activating peptide counteracts hedgehog-dependent motor neuron production in mouse embryonic stem cell cultures. J Neurosci Res 2011; 89:1363-74. [PMID: 21674568 DOI: 10.1002/jnr.22675] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 03/31/2011] [Indexed: 12/14/2022]
Abstract
Pituitary adenylyl cyclase-activating peptide (PACAP; ADCYAP1) is a neuropeptide that regulates a wide array of functions within the brain and periphery. We and others have previously demonstrated that PACAP and its high-affinity receptor PAC1 are expressed in the embryonic mouse neural tube, suggesting that PACAP plays a role in early brain development. Moreover, we previously showed that PACAP antagonizes the mitotic action of Sonic hedgehog (Shh) in postnatal cerebellar granule precursors. In the present study, we demonstrate that PACAP completely blocked Shh-dependent motor neuron generation from embryonic stem cell cultures and reduced mRNA levels of the Shh target gene Gli-1 and several ventral spinal cord patterning genes. In vivo examination of motor neuron and other patterning markers in embryonic day 12.5 spinal cords of wild-type and PACAP-deficient mice by immunofluorescence, on the other hand, revealed no obvious alterations in expressions of Islet1/2, MNR2, Lim1/2, Nkx2.2, or Shh, although the Pax6-positive area was slightly expanded in PACAP-deficient spinal cord. Caspase-3 staining revealed low, and similar, numbers of cells undergoing apoptosis in embryonic wild-type vs. PACAP-deficient spinal cords, whereas a slight but significant increase in number of mitotic cells was observed in PACAP-deficient mice. Thus, although PACAP has a strong capacity to counteract Shh signaling and motor neuron production in vitro, corresponding patterning defects associated with PACAP loss may be obscured by compensatory mechanisms.
Collapse
Affiliation(s)
- Megumi Hirose
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-7332, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
The hallmarks of CDKN1C (p57, KIP2) in cancer. Biochim Biophys Acta Rev Cancer 2011; 1816:50-6. [PMID: 21447370 DOI: 10.1016/j.bbcan.2011.03.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 03/18/2011] [Accepted: 03/22/2011] [Indexed: 12/18/2022]
Abstract
Cyclin-dependent kinase inhibitor 1C CDKN1C (p57(KIP2)) regulates several hallmarks of cancer, including apoptosis, cell invasion and metastasis, tumor differentiation and angiogenesis. p57(KIP2) is generally not mutated in cancer, but its expression is downregulated through epigenetic changes such as DNA methylation and repressive histone marks at the promoter. This opens up possibilities for therapeutic intervention through reactivation of p57(KIP2) gene expression. Furthermore, p57(KIP2) has been tested as a prognostic factor for many types of cancer, even differentiating between early and late stage cancer. In this review, the multifunctional tumor suppressor capabilities of p57(KIP2), the mechanisms of p57(KIP2) transcriptional repression in cancer, and the therapeutic potential of reactivation of p57(KIP2) protein expression will be discussed.
Collapse
|
16
|
Tury A, Mairet-Coello G, DiCicco-Bloom E. The cyclin-dependent kinase inhibitor p57Kip2 regulates cell cycle exit, differentiation, and migration of embryonic cerebral cortical precursors. ACTA ACUST UNITED AC 2011; 21:1840-56. [PMID: 21245411 DOI: 10.1093/cercor/bhq254] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mounting evidence indicates cyclin-dependent kinase (CDK) inhibitors (CKIs) of the Cip/Kip family, including p57(Kip2) and p27(Kip1), control not only cell cycle exit but also corticogenesis. Nevertheless, distinct activities of p57(Kip2) remain poorly defined. Using in vivo and culture approaches, we show p57(Kip2) overexpression at E14.5-15.5 elicits precursor cell cycle exit, promotes transition from proliferation to neuronal differentiation, and enhances process outgrowth, while opposite effects occur in p57(Kip2)-deficient precursors. Studies at later ages indicate p57(Kip2) overexpression also induces precocious glial differentiation, suggesting stage-dependent effects. In embryonic cortex, p57(Kip2) overexpression advances cell radial migration and alters postnatal laminar positioning. While both CKIs induce differentiation, p57(Kip2) was twice as effective as p27(Kip1) in inducing neuronal differentiation and was not permissive to astrogliogenic effects of ciliary neurotrophic factor, suggesting that the CKIs differentially modulate cell fate decisions. At molecular levels, although highly conserved N-terminal regions of both CKIs elicit cycle withdrawal and differentiation, the C-terminal region of p57(Kip2) alone inhibits in vivo migration. Furthermore, p57(Kip2) effects on neurogenesis and gliogenesis require the N-terminal cyclin/CDK binding/inhibitory domains, while previous p27(Kip1) studies report cell cycle-independent functions. These observations suggest p57(Kip2) coordinates multiple stages of corticogenesis and exhibits distinct and common activities compared with related family member p27(Kip1).
Collapse
Affiliation(s)
- Anna Tury
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | | | | |
Collapse
|
17
|
Callihan P, Mumaw J, Machacek DW, Stice SL, Hooks SB. Regulation of stem cell pluripotency and differentiation by G protein coupled receptors. Pharmacol Ther 2010; 129:290-306. [PMID: 21073897 DOI: 10.1016/j.pharmthera.2010.10.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 10/08/2010] [Indexed: 01/25/2023]
Abstract
Stem cell-based therapeutics have the potential to effectively treat many terminal and debilitating human diseases, but the mechanisms by which their growth and differentiation are regulated are incompletely defined. Recent data from multiple systems suggest major roles for G protein coupled receptor (GPCR) pathways in regulating stem cell function in vivo and in vitro. The goal of this review is to illustrate common ground between the growing field of stem cell therapeutics and the long-established field of G protein coupled receptor signaling. Herein, we briefly introduce basic stem cell biology and discuss how several conserved pathways regulate pluripotency and differentiation in mouse and human stem cells. We further discuss general mechanisms by which GPCR signaling may impact these pluripotency and differentiation pathways, and summarize specific examples of receptors from each of the major GPCR subfamilies that have been shown to regulate stem cell function. Finally, we discuss possible therapeutic implications of GPCR regulation of stem cell function.
Collapse
Affiliation(s)
- Phillip Callihan
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, United States
| | | | | | | | | |
Collapse
|
18
|
Ye W, Blain SW. S phase entry causes homocysteine-induced death while ataxia telangiectasia and Rad3 related protein functions anti-apoptotically to protect neurons. Brain 2010; 133:2295-312. [PMID: 20639548 DOI: 10.1093/brain/awq139] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A major phenotype seen in neurodegenerative disorders is the selective loss of neurons due to apoptotic death and evidence suggests that inappropriate re-activation of cell cycle proteins in post-mitotic neurons may be responsible. To investigate whether reactivation of the G1 cell cycle proteins and S phase entry was linked with apoptosis, we examined homocysteine-induced neuronal cell death in a rat cortical neuron tissue culture system. Hyperhomocysteinaemia is a physiological risk factor for a variety of neurodegenerative diseases, including Alzheimer's disease. We found that in response to homocysteine treatment, cyclin D1, and cyclin-dependent kinases 4 and 2 translocated to the nucleus, and p27 levels decreased. Both cyclin-dependent kinases 4 and 2 regained catalytic activity, the G1 gatekeeper retinoblastoma protein was phosphorylated and DNA synthesis was detected, suggesting transit into S phase. Double-labelling immunofluorescence showed a 95% co-localization of anti-bromodeoxyuridine labelling with apoptotic markers, demonstrating that those cells that entered S phase eventually died. Neurons could be protected from homocysteine-induced death by methods that inhibited G1 phase progression, including down-regulation of cyclin D1 expression, inhibition of cyclin-dependent kinases 4 or 2 activity by small molecule inhibitors, or use of the c-Abl kinase inhibitor, Gleevec, which blocked cyclin D and cyclin-dependent kinase 4 nuclear translocation. However, blocking cell cycle progression post G1, using DNA replication inhibitors, did not prevent apoptosis, suggesting that death was not preventable post the G1-S phase checkpoint. While homocysteine treatment caused DNA damage and activated the DNA damage response, its mechanism of action was distinct from that of more traditional DNA damaging agents, such as camptothecin, as it was p53-independent. Likewise, inhibition of the DNA damage sensors, ataxia-telangiectasia mutant and ataxia telangiectasia and Rad3 related proteins, did not rescue apoptosis and in fact exacerbated death, suggesting that the DNA damage response might normally function neuroprotectively to block S phase-dependent apoptosis induction. As cell cycle events appear to be maintained in vivo in affected neurons for weeks to years before apoptosis is observed, activation of the DNA damage response might be able to hold cell cycle-induced death in check.
Collapse
Affiliation(s)
- Weizhen Ye
- Department of Paediatrics, State University of New York, Downstate Medical Centre, Brooklyn, NY 11203, USA
| | | |
Collapse
|
19
|
Njaine B, Martins RAP, Santiago MF, Linden R, Silveira MS. Pituitary adenylyl cyclase-activating polypeptide controls the proliferation of retinal progenitor cells through downregulation of cyclin D1. Eur J Neurosci 2010; 32:311-21. [PMID: 20646049 DOI: 10.1111/j.1460-9568.2010.07286.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
During retinal development, cell proliferation and exit from the cell cycle must be precisely regulated to ensure the generation of the appropriate numbers and proportions of the various retinal cell types. Previously, we showed that pituitary adenylyl cyclase-activating polypeptide (PACAP) exerts a neuroprotective effect in the developing retina of rats, through the cAMP-cAMP-dependent protein kinase (protein kinase A) (PKA) pathway. Here, we show that PACAP also regulates the proliferation of retinal progenitor cells. PACAP, PACAP-specific receptor (PAC1), and the receptors activated by both PACAP and vasoactive intestinal peptide (VIP), VPAC1 and VPAC2, are expressed during embryonic and postnatal development of the rat retina. Treatment of retinal explants with PACAP38 reduced the incorporation of [(3)H]thymidine as well as the number of 5-bromo-2'-deoxyuridine-positive and cyclin D1-positive cells. Pharmacological experiments indicated that PACAP triggers this antiproliferative effect through the activation of both PAC1 and VPACs, and the cAMP-PKA pathway. In addition, PACAP receptor activation decreased both cyclin D1 mRNA and protein content. Altogether, the data support the hypothesis that PACAP is a cell-extrinsic regulator with multiple roles during retinal development, including the regulation of proliferation in a subpopulation of retinal progenitor cells.
Collapse
Affiliation(s)
- Brian Njaine
- Instituto de Biofisica Carlos Chagas Filho-UFRJ, Edifício do Centro de Ciencias da Saude, Ilha do Fundão, Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
20
|
Dejda A, Chan P, Seaborn T, Coquet L, Jouenne T, Fournier A, Vaudry H, Vaudry D. Involvement of stathmin 1 in the neurotrophic effects of PACAP in PC12 cells. J Neurochem 2010; 114:1498-510. [PMID: 20569302 DOI: 10.1111/j.1471-4159.2010.06873.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rat pheochromocytoma PC12 cells have been widely used to investigate the neurotrophic activities of pituitary adenylate cyclase-activating polypeptide (PACAP). In particular, PACAP has been shown to promote differentiation and to inhibit apoptosis of PC12 cells. In order to identify the mechanisms mediating these effects, we sought for proteins that are phosphorylated upon PACAP treatment. High-performance liquid chromatography and 2D gel electrophoresis analysis, coupled with mass spectrometry, revealed that stathmin 1 is strongly phosphorylated within only 5 min of exposure to PACAP. Western blot experiments confirmed that PACAP induced a robust phosphorylation of stathmin 1 in a time-dependent manner. On the other hand, PACAP decreased stathmin 1 gene expression. Investigations of the signaling mechanisms known to be activated by PACAP revealed that phosphorylation of stathmin 1 was mainly mediated through the protein kinase A and mitogen-activated protein kinase pathways. Blockage of stathmin 1 expression with small interfering RNA did not affect PC12 cell differentiation induced by PACAP but reduced the ability of the peptide to inhibit caspase 3 activity and significantly decreased its neuroprotective action. Taken together, these data demonstrate that stathmin 1 is involved in the neurotrophic effect of PACAP in PC12 cells.
Collapse
Affiliation(s)
- Agnieszka Dejda
- INSERM U982, Institut Fédératif de Recherches Multidisciplinaires sur les Peptides (IFRMP 23), Université de Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | |
Collapse
|
21
|
May V, Lutz E, MacKenzie C, Schutz KC, Dozark K, Braas KM. Pituitary adenylate cyclase-activating polypeptide (PACAP)/PAC1HOP1 receptor activation coordinates multiple neurotrophic signaling pathways: Akt activation through phosphatidylinositol 3-kinase gamma and vesicle endocytosis for neuronal survival. J Biol Chem 2010; 285:9749-9761. [PMID: 20093365 PMCID: PMC2843224 DOI: 10.1074/jbc.m109.043117] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
MAPK and Akt pathways are predominant mediators of trophic signaling for many neuronal systems. Among the vasoactive intestinal peptide/secretin/glucagon family of related peptides, pituitary adenylate cyclase-activating polypeptide (PACAP) binding to specific PAC(1) receptor isoforms can engage multiple signaling pathways and promote neuroprotection through mechanisms that are not well understood. Using a primary sympathetic neuronal system, the current studies demonstrate that PACAP activation of PAC(1)HOP1 receptors engages both MAPK and Akt neurotrophic pathways in an integrated program to facilitate neuronal survival after growth factor withdrawal. PACAP not only stimulated prosurvival ERK1/2 and ERK5 activation but also abrogated SAPK/JNK and p38 MAPK signaling in parallel. In contrast to the potent and rapid effects of PACAP in ERK1/2 phosphorylation, PACAP stimulated Akt phosphorylation in a late phase of PAC(1)HOP1 receptor signaling. From inhibitor and immunoprecipitation analyses, the PACAP/PAC(1)HOP1 receptor-mediated Akt responses did not represent transactivation mechanisms but appeared to depend on G alpha(q)/phosphatidylinositol 3-kinase gamma activity and vesicular internalization pathways. Phosphatidylinositol 3-kinase gamma-selective inhibitors blocked PACAP-stimulated Akt phosphorylation in primary neuronal cultures and in PAC(1)HOP1-overexpressing cell lines; RNA interference-mediated knockdown of the receptor effectors attenuated PACAP-mediated Akt activation. Similarly, perturbation of endocytic pathways also blocked Akt phosphorylation. Between ERK and Akt pathways, PACAP-stimulated Akt signaling was the primary cascade that attenuated cultured neuron apoptosis after growth factor withdrawal. The partitioning of PACAP-mediated Akt signaling in endosomes may be a key mechanism contributing to the high spatial and temporal specificity in signal transduction necessary for survival pathways.
Collapse
Affiliation(s)
- Victor May
- Departments of Anatomy and Neurobiology and of Pharmacology, The University of Vermont College of Medicine, Burlington, Vermont 05405.
| | - Eve Lutz
- University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical Sciences, Royal College, 204 George Street, Glasgow G1 1XW, Scotland, United Kingdom
| | - Christopher MacKenzie
- University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical Sciences, Royal College, 204 George Street, Glasgow G1 1XW, Scotland, United Kingdom
| | - Kristin C Schutz
- Departments of Anatomy and Neurobiology and of Pharmacology, The University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Kate Dozark
- Departments of Anatomy and Neurobiology and of Pharmacology, The University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Karen M Braas
- Departments of Anatomy and Neurobiology and of Pharmacology, The University of Vermont College of Medicine, Burlington, Vermont 05405
| |
Collapse
|
22
|
GABAergic amacrine cells and visual function are reduced in PAC1 transgenic mice. Neuropharmacology 2010; 58:215-25. [DOI: 10.1016/j.neuropharm.2009.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 06/25/2009] [Accepted: 07/02/2009] [Indexed: 01/22/2023]
|
23
|
Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BKC, Hashimoto H, Galas L, Vaudry H. Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol Rev 2009; 61:283-357. [PMID: 19805477 DOI: 10.1124/pr.109.001370] [Citation(s) in RCA: 858] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38-amino acid C-terminally alpha-amidated peptide that was first isolated 20 years ago from an ovine hypothalamic extract on the basis of its ability to stimulate cAMP formation in anterior pituitary cells (Miyata et al., 1989. PACAP belongs to the vasoactive intestinal polypeptide (VIP)-secretin-growth hormone-releasing hormone-glucagon superfamily. The sequence of PACAP has been remarkably well conserved during evolution from protochordates to mammals, suggesting that PACAP is involved in the regulation of important biological functions. PACAP is widely distributed in the brain and peripheral organs, notably in the endocrine pancreas, gonads, respiratory and urogenital tracts. Characterization of the PACAP precursor has revealed the existence of a PACAP-related peptide, the activity of which remains unknown. Two types of PACAP binding sites have been characterized: type I binding sites exhibit a high affinity for PACAP and a much lower affinity for VIP, whereas type II binding sites have similar affinity for PACAP and VIP. Molecular cloning of PACAP receptors has shown the existence of three distinct receptor subtypes: the PACAP-specific PAC1-R, which is coupled to several transduction systems, and the PACAP/VIP-indifferent VPAC1-R and VPAC2-R, which are primarily coupled to adenylyl cyclase. PAC1-Rs are particularly abundant in the brain, the pituitary and the adrenal gland, whereas VPAC receptors are expressed mainly in lung, liver, and testis. The development of transgenic animal models and specific PACAP receptor ligands has strongly contributed to deciphering the various actions of PACAP. Consistent with the wide distribution of PACAP and its receptors, the peptide has now been shown to exert a large array of pharmacological effects and biological functions. The present report reviews the current knowledge concerning the pleiotropic actions of PACAP and discusses its possible use for future therapeutic applications.
Collapse
Affiliation(s)
- David Vaudry
- Institut National de la Santé et de la Recherche Médicale U413, European Institute for Peptide Research (Institut Fédératif de Recherches Multidisciplinaires sur les Peptides 23), Mont-Saint-Aignan, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Nielsen JA, Lau P, Maric D, Barker JL, Hudson LD. Integrating microRNA and mRNA expression profiles of neuronal progenitors to identify regulatory networks underlying the onset of cortical neurogenesis. BMC Neurosci 2009; 10:98. [PMID: 19689821 PMCID: PMC2736963 DOI: 10.1186/1471-2202-10-98] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 08/19/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cortical development is a complex process that includes sequential generation of neuronal progenitors, which proliferate and migrate to form the stratified layers of the developing cortex. To identify the individual microRNAs (miRNAs) and mRNAs that may regulate the genetic network guiding the earliest phase of cortical development, the expression profiles of rat neuronal progenitors obtained at embryonic day 11 (E11), E12 and E13 were analyzed. RESULTS Neuronal progenitors were purified from telencephalic dissociates by a positive-selection strategy featuring surface labeling with tetanus-toxin and cholera-toxin followed by fluorescence-activated cell sorting. Microarray analyses revealed the fractions of miRNAs and mRNAs that were up-regulated or down-regulated in these neuronal progenitors at the beginning of cortical development. Nearly half of the dynamically expressed miRNAs were negatively correlated with the expression of their predicted target mRNAs. CONCLUSION These data support a regulatory role for miRNAs during the transition from neuronal progenitors into the earliest differentiating cortical neurons. In addition, by supplying a robust data set in which miRNA and mRNA profiles originate from the same purified cell type, this empirical study may facilitate the development of new algorithms to integrate various "-omics" data sets.
Collapse
Affiliation(s)
- Joseph A Nielsen
- Section of Developmental Genetics, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | |
Collapse
|
25
|
p57Kip2 is a repressor of Mash1 activity and neuronal differentiation in neural stem cells. Cell Death Differ 2009; 16:1256-65. [PMID: 19590511 DOI: 10.1038/cdd.2009.72] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Mammalian central nervous system (CNS) development is a highly organized process involving the precise and coordinated timing of cell-cycle exit, differentiation, survival, and migration. These events require proper expression of pro-neuronal genes but also repression of alternative cell fates and restriction of cell-type-specific gene expression. Here, we show that the cyclin-dependent kinase (CDK) inhibitor p57Kip2 interacted with pro-neuronal basic helix-loop-helix (bHLH) factors such as Mash1, NeuroD, and Nex/Math2. Increased levels of p57Kip2 inhibited Mash1 transcriptional activity independently of CDK interactions and acted as a direct repressor in transcriptional assays. Proliferating telencephalic neural progenitors co-expressed basal levels of Mash1 and p57Kip2, and endogenous p57Kip2 accumulated transiently in the nuclei of neural stem cells (NSCs) during early stages of astrocyte differentiation mediated by ciliary neurotrophic factor (CNTF), independent of cell-cycle exit and at times when Mash1 expression was still prominent. In accordance with these observations, gain- and loss-of-function studies showed that p57Kip2 repressed neuronal differentiation after mitogen withdrawal, but exerted little or no effect on CNTF-mediated astroglial differentiation of NSCs. Our data suggest a novel role for p57Kip2 as a context-dependent repressor of neurogenic transcription factors and telencephalic neuronal differentiation.
Collapse
|
26
|
Ye W, Mairet-Coello G, Pasoreck E, Dicicco-Bloom E. Patterns of p57Kip2 expression in embryonic rat brain suggest roles in progenitor cell cycle exit and neuronal differentiation. Dev Neurobiol 2009; 69:1-21. [PMID: 18814313 DOI: 10.1002/dneu.20680] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In developing central nervous system, a variety of mechanisms couple cell cycle exit to differentiation during neurogenesis. The cyclin-dependent kinase (CDK) inhibitor p57Kip2 controls the transition from proliferation to differentiation in many tissues, but roles in developing brain remain uncertain. To characterize possible functions, we defined p57Kip2 protein expression in embryonic (E) day 12.5 to 20.5 rat brains using immunohistochemistry combined with markers of proliferation and differentiation. The p57Kip2 was localized primarily in cell nuclei and positive cells formed two distinct patterns including wide dispersion and laminar aggregation that were brain region-specific. From E12.5 to E16.5, p57Kip2 expression was detected mainly in ventricular zone (VZ) and/or mantle zone of hippocampus, septum, basal ganglia, thalamus, hypothalamus, midbrain, and spinal cord. After E18.5, p57Kip2 was detected in select regions undergoing differentiation. The p57Kip2 expression was also compared with regional transcription factors, including Ngn2, Nkx2.1, and Pax6. Time course studies performed in diencephalon showed that p57Kip2 immunoreactivity colocalized with BrdU at 8 hr in nuclei exhibiting the wide dispersion pattern, whereas colocalization in the laminar pattern occurred only later. Moreover, p57Kip2 frequently colocalized with neuronal marker, beta-III tubulin. Finally, we characterized relationships of p57Kip2 to CDK inhibitor p27Kip1: in proliferative regions, p57Kip2 expression preceded p27Kip1 as cells underwent differentiation, though the proteins colocalized in substantial numbers of cells, suggesting potentially related yet distinct functions of Cip/Kip family members during neurogenesis. Our observations that p57Kip2 exhibits nuclear expression as precursors exit the cell cycle and begin expressing neuronal characteristics suggests that the CDK inhibitor contributes to regulating the transition from proliferation to differentiation during brain development.
Collapse
Affiliation(s)
- Weizhen Ye
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | | | | | |
Collapse
|
27
|
Fila T, Trazzi S, Crochemore C, Bartesaghi R, Ciani E. Lot1 is a key element of the pituitary adenylate cyclase-activating polypeptide (PACAP)/cyclic AMP pathway that negatively regulates neuronal precursor proliferation. J Biol Chem 2009; 284:15325-38. [PMID: 19346254 DOI: 10.1074/jbc.m109.002329] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The tumor suppressor gene Lot1 is highly expressed during brain development. During cerebellar development, Lot1 is expressed by proliferating granule cells with a time course matching the expression of the pituitary adenylate cyclase-activating polypeptide (PACAP) receptor, a neuropeptide receptor that plays an important role in the regulation of granule cell proliferation/survival. Although it has become clear that Lot1 is a negative regulator of cell division in tumor cells, its role in neuronal proliferation is not understood. We previously demonstrated that in cerebellar granule cells Lot1 expression is regulated by the PACAP/cAMP system. The aim of this study was to investigate the role played by Lot1 in neuron proliferation/survival and to identify the molecular mechanisms underlying its actions. Using a Lot1-inducible expression system, we found that in PC12 cells Lot1 negatively regulates proliferation and favors differentiation by up-regulating the expression of the PACAP receptor. In cerebellar granule cells in culture, an increase in Lot1 expression was paralleled by inhibition of proliferation and up-regulation of the PACAP receptor, which in turn positively regulated Lot1 expression. Silencing of Lot1 leads to an increase in granule cell proliferation and a reduction in survival. Confirming the in vitro results, in vivo experiments showed that PACAP induced an increase in Lot1 expression that was paralleled by inhibition of cerebellar granule cell proliferation. These data show that Lot1 is a key element of the PACAP/cAMP pathway that negatively regulates neuronal precursor proliferation. The existence of a PACAP receptor/Lot1-positive feedback loop may powerfully regulate neural proliferation during critical phases of cerebellar development.
Collapse
Affiliation(s)
- Tatiana Fila
- Department of Human and General Physiology, University of Bologna, Piazza di Porta San Donato 2, Bologna, Italy
| | | | | | | | | |
Collapse
|
28
|
Vallejo M. PACAP signaling to DREAM: a cAMP-dependent pathway that regulates cortical astrogliogenesis. Mol Neurobiol 2009; 39:90-100. [PMID: 19238593 DOI: 10.1007/s12035-009-8055-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 01/19/2009] [Indexed: 12/23/2022]
Abstract
Astrocytes constitute a very abundant cell type in the mammalian central nervous system and play critical roles in brain function. During development, astrocytes are generated from neural progenitor cells only after these cells have generated neurons. This so called gliogenic switch is tightly regulated by intrinsic factors that inhibit the generation of astrocytes during the neurogenic period. Once neural progenitors acquire gliogenic competence, they differentiate into astrocytes in response to specific extracellular signals. Some of these signals are delivered by neurotrophic cytokines via activation of the gp130-JAK-signal transducer and activator of transcription system, whereas others depend on the activity of pituitary adenylate cyclase-activating polypeptide (PACAP) on specific PAC1 receptors that stimulate the production of cAMP. This results in the activation of the small GTPases Rap1 and Ras, and in the cAMP-dependent entry of extracellular calcium into the cell. Calcium, in turn, stimulates the transcription factor downstream regulatory element antagonist modulator (DREAM), which is bound to specific sites of the promoter of the glial fibrillary acidic protein gene, stimulating its expression during astrocyte differentiation. Lack of DREAM in vivo results in alterations in the number of neurons and astrocytes generated during development. Thus, the PACAP-cAMP-Ca(2+)-DREAM signaling cascade constitutes an important pathway to activate glial-specific gene expression during astrocyte differentiation.
Collapse
Affiliation(s)
- Mario Vallejo
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, 28029, Madrid, Spain.
| |
Collapse
|
29
|
Sanchez A, Rao HV, Grammas P. PACAP38 protects rat cortical neurons against the neurotoxicity evoked by sodium nitroprusside and thrombin. ACTA ACUST UNITED AC 2008; 152:33-40. [PMID: 18682263 DOI: 10.1016/j.regpep.2008.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 07/02/2008] [Accepted: 07/08/2008] [Indexed: 10/21/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) 38 is a multifunctional anti-inflammatory and anti-apoptotic neuropeptide widely distributed in the nervous system. The objective of this study is to determine whether PACAP38 is neuroprotective against sodium nitroprusside (SNP) and thrombin, two mechanistically distinct neurotoxic agents. Treatment of primary cortical neuronal cultures with 1 mM SNP for 4 h causes neuronal cell death that is significantly reduced by 100 nM PACAP38. PACAP38 down-regulates SNP-induced cell cycle protein (cyclin E) expression and up-regulates p57(KIP2), a cyclin-dependent kinase inhibitor as well as the anti-apoptotic protein Bcl-2. Similarly, neuronal death induced by 100 nM thrombin or the thrombin receptor activating peptide (TRAP 6) is reduced by PACAP38 treatment. Thrombin-stimulated cell cycle protein (cdk4) expression is decreased by PACAP38 while PACAP38 inhibits thrombin-mediated reduction of p57(KIP2). However, the decrease in Bcl-2 evoked by thrombin is not affected by PACAP38. Finally, both SNP and thrombin (or TRAP) increase caspase 3 activity, an effect that is decreased by PACAP38. These data show that PACAP38 supports neuronal survival in vitro suppressing cell cycle progression and enhancing anti-apoptotic proteins. Our results support the possibility that PACAP could be a useful therapeutic agent for reducing neuronal cell death in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alma Sanchez
- Department of Neuropsychiatry, Texas Tech University Health Sciences Center, Lubbock TX 79430, USA
| | | | | |
Collapse
|
30
|
Scharf E, May V, Braas KM, Shutz KC, Mao-Draayer Y. Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) regulate murine neural progenitor cell survival, proliferation, and differentiation. J Mol Neurosci 2008; 36:79-88. [PMID: 18629655 DOI: 10.1007/s12031-008-9097-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Accepted: 05/01/2008] [Indexed: 01/17/2023]
Abstract
Neural stem/progenitor cells (NPC) have gained wide interest over the last decade from their therapeutic potential, either through transplantation or endogenous replacement, after central nervous system (CNS) disease and damage. Whereas several growth factors and cytokines have been shown to promote NPC survival, proliferation, or differentiation, the identification of other regulators will provide much needed options for NPC self-renewal or lineage development. Although previous studies have shown that pituitary adenylate cyclase-activating polypeptide (PACAP)/vasoactive intestinal peptide (VIP) can regulate stem/progenitor cells, the responses appeared variable. To examine the direct roles of these peptides in NPCs, postnatal mouse NPC cultures were withdrawn from epidermal growth factor (EGF) and fibroblastic growth factor (FGF) and maintained under serum-free conditions in the presence or absence of PACAP27, PACAP38, or VIP. The NPCs expressed the PAC1(short)null receptor isoform, and the activation of these receptors decreased progenitor cell apoptosis more than 80% from TUNEL assays and facilitated proliferation more than fivefold from bromodeoxyuridine (BrdU) analyses. To evaluate cellular differentiation, replicate control and peptide-treated cultures were examined for cell fate marker protein and transcript expression. In contrast with previous work, PACAP peptides downregulated NPC differentiation, which appeared consistent with the proliferation status of the treated cells. Accordingly, these results demonstrate that PACAP signaling is trophic and can maintain NPCs in a multipotent state. With these attributes, PACAP may be able to promote endogenous NPC self-renewal in the adult CNS, which may be important for endogenous self-repair in disease and ageing processes.
Collapse
Affiliation(s)
- Eugene Scharf
- Department of Neurology, College of Medicine, University of Vermont, 1 South Prospect Street, UHC-Neurology, Burlington, VT 05401, USA
| | | | | | | | | |
Collapse
|
31
|
PACAP decides neuronal laminar fate via PKA signaling in the developing cerebral cortex. Biochem Biophys Res Commun 2008; 369:1144-9. [DOI: 10.1016/j.bbrc.2008.03.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Accepted: 03/04/2008] [Indexed: 11/20/2022]
|
32
|
Granule cell survival is deficient in PAC1-/- mutant cerebellum. J Mol Neurosci 2008; 36:38-44. [PMID: 18409023 DOI: 10.1007/s12031-008-9066-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Accepted: 03/10/2008] [Indexed: 12/23/2022]
Abstract
PACAP exerts neuroprotective effects during development, especially in the cerebellum where PAC1 receptor and ligand are both expressed. However, while previous studies using PACAP injections in postnatal animals defined trophic effects of exogenous peptide, the role of endogenous PACAP remains unexplored. Here, we used PAC1(-/-) mice to investigate the role of PACAP receptor signaling in postnatal day 7 cerebellum. There was no difference in DNA synthesis in the cerebellar EGL of PAC1(-/-) compared to wild type animals, assessed using thymidine incorporation and BrdU immunohistochemistry. In contrast, we found that a significant proportion of newly generated neurons were eliminated before they successfully differentiated in the granule cell layer. In aggregate, these results suggest that endogenous PACAP plays an important role in cell survival during cerebellar development, through the activation of the PAC1 receptor.
Collapse
|
33
|
Ghzili H, Grumolato L, Thouënnon E, Tanguy Y, Turquier V, Vaudry H, Anouar Y. Role of PACAP in the physiology and pathology of the sympathoadrenal system. Front Neuroendocrinol 2008; 29:128-41. [PMID: 18048093 DOI: 10.1016/j.yfrne.2007.10.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 08/24/2007] [Accepted: 10/01/2007] [Indexed: 01/09/2023]
Abstract
Sympathetic neurons and chromaffin cells derive from common sympathoadrenal precursors which arise from the neural crest. Cells from this lineage migrate to their final destination and differentiate by acquiring a catecholaminergic phenotype in response to different environmental factors. It has been shown that the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) and its PAC1 receptor are expressed at early stages of sympathetic development, and participate to the control of neuroblast proliferation and differentiation. PACAP also acts as a neurotransmitter to stimulate catecholamine and neuropeptide biosynthesis and release from sympathetic neurons and chromaffin cells, during development and in adulthood. In addition, PACAP and its receptors have been described in neuroblastoma and pheochromocytoma, and the neuropeptide regulates the differentiation and activity of sympathoadrenal-derived tumoral cell lines, suggestive of an important role in the pathophysiology of the sympathoadrenal lineage. Transcriptome studies uncovered genes and pathways of known and unknown roles that underlie the effects of PACAP in the sympathoadrenal system.
Collapse
Affiliation(s)
- Hafida Ghzili
- INSERM, U413, Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research (IFRMP23), University of Rouen, 76821 Mont-Saint-Aignan, France
| | | | | | | | | | | | | |
Collapse
|
34
|
Gandhi R, Luk KC, Rymar VV, Sadikot AF. Group I mGluR5 metabotropic glutamate receptors regulate proliferation of neuronal progenitors in specific forebrain developmental domains. J Neurochem 2007; 104:155-72. [PMID: 17944877 DOI: 10.1111/j.1471-4159.2007.04955.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Major classical neurotransmitters including GABA and glutamate play novel morphogenic roles during development of the mammalian CNS. During forebrain neurogenesis, glutamate regulates neuroblast proliferation in different germinal domains using receptor subtype-specific mechanisms. For example, ionotropic N-methyl-D-aspartate (NMDA) or alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) glutamate receptors mediate distinct proliferative effects in ventral or dorsal forebrain germinal domains, and regulate the correct number of neurons that populate the striatum or cerebral cortex. Recent work suggests metabotropic receptors may also mediate glutamate's proliferative effects. Group I mGluR5 receptor subtypes are highly expressed in forebrain germinal zones. Using in vitro and in vivo methods, we demonstrate mGluR5 receptor activation plays an important role in neuroblast proliferation in the ventral telencephalon, and helps determine the complement of striatum projection neurons. mGluR5 receptor-mediated effects on striatal neuronal progenitors are restricted mainly to early cycling populations in the ventricular zone, with little effect on secondary proliferative populations in the subventricular zone. In contrast to proliferative effects in the ventral telencephalon, mGluR5 receptors do not modulate proliferation of dorsal telencephalon-derived cortical neuroblasts. Heterogeneous domain-specific proliferative effects of glutamate-mediated by specific receptor subtypes provide an important developmental mechanism allowing generation of the correct complement of neuronal subtypes that populate the mammalian forebrain.
Collapse
Affiliation(s)
- Rina Gandhi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
35
|
Candal E, Alunni A, Thermes V, Jamen F, Joly JS, Bourrat F. Ol-insm1b, a SNAG family transcription factor involved in cell cycle arrest during medaka development. Dev Biol 2007; 309:1-17. [PMID: 17559827 DOI: 10.1016/j.ydbio.2007.04.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2007] [Revised: 04/14/2007] [Accepted: 04/26/2007] [Indexed: 11/28/2022]
Abstract
Through whole-mount in situ hybridisation screen on medaka (Oryzias latipes) brain, Ol-insm1b, a member of the Insm1/Mlt1 subfamily of SNAG-domain containing genes, has been isolated. It is strongly expressed during neurogenesis and pancreas organogenesis, with a pattern that suggests a role in cell cycle exit. Here, we describe Ol-insm1b expression pattern throughout development and in adult brain, and we report on its functional characterisation. Our data point to a previously unravelled role for Ol-insm1b as a down-regulator of cell proliferation during development, as it slows down the cycle without triggering apoptosis. Clonal analysis demonstrates that this effect is cell-autonomous, and, through molecular dissection studies, we demonstrate that it is likely to be non-transcriptional, albeit mediated by zinc-finger domains. Additionally, we report that Ol-insm1b mRNA, when injected in one cell of two-cell stage embryos, exhibits a surprising behaviour: it does not spread uniformly amongst daughter cells but remains cytoplasmically localised in the progeny of the injected blastomere. Our experiments suggest that Insm1 is a negative regulator of cell proliferation, possibly through mechanisms that do not involve modulation of transcription.
Collapse
Affiliation(s)
- Eva Candal
- INRA MSNC Group, DEPSN, Institut Fessard, CNRS, 1 Avenue de la Terrasse, 91198 GIF-SUR-YVETTE, France.
| | | | | | | | | | | |
Collapse
|
36
|
Falluel-Morel A, Chafai M, Vaudry D, Basille M, Cazillis M, Aubert N, Louiset E, de Jouffrey S, Le Bigot JF, Fournier A, Gressens P, Rostène W, Vaudry H, Gonzalez BJ. The neuropeptide pituitary adenylate cyclase-activating polypeptide exerts anti-apoptotic and differentiating effects during neurogenesis: focus on cerebellar granule neurones and embryonic stem cells. J Neuroendocrinol 2007; 19:321-7. [PMID: 17425606 DOI: 10.1111/j.1365-2826.2007.01537.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) was originally isolated from ovine hypothalamus on the basis of its hypophysiotrophic activity. It has subsequently been shown that PACAP and its receptors are widely distributed in the central nervous system of adult mammals, indicating that PACAP may act as a neurotransmitter and/or neuromodulator. It has also been found that PACAP and its receptors are expressed in germinative neuroepithelia, suggesting that PACAP could be involved in neurogenesis. There is now compelling evidence that PACAP exerts neurotrophic activities in the developing cerebellum and in embryonic stem (ES) cells. In particular, the presence of PACAP receptors has been demonstrated in the granule layer of the immature cerebellar cortex, and PACAP has been shown to promote survival, inhibit migration and activate neurite outgrowth of granule cell precursors. In cerebellar neuroblasts, PACAP is a potent inhibitor of the mitochondrial apoptotic pathway through activation of the MAPkinase extracellular regulated kinase. ES cells and embryoid bodies (EB) also express PACAP receptors and PACAP facilitates neuronal orientation and induces the appearance of an electrophysiological activity. Taken together, the anti-apoptotic and pro-differentiating effects of PACAP characterised in cerebellar neuroblasts as well as ES and EB cells indicate that PACAP acts not only as a neurohormone and a neurotransmitter, but also as a growth factor.
Collapse
Affiliation(s)
- A Falluel-Morel
- INSERM U413, Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research (IFRMP 23), University of Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Delcourt N, Thouvenot E, Chanrion B, Galéotti N, Jouin P, Bockaert J, Marin P. PACAP type I receptor transactivation is essential for IGF-1 receptor signalling and antiapoptotic activity in neurons. EMBO J 2007; 26:1542-51. [PMID: 17332755 PMCID: PMC1829375 DOI: 10.1038/sj.emboj.7601608] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Accepted: 01/24/2007] [Indexed: 11/10/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) and pituitary adenylyl cyclase activating polypeptide (PACAP) are both potent neurotrophic and antiapoptotic factors, which exert their effects via phosphorylation cascades initiated by tyrosine kinase and G-protein-coupled receptors, respectively. Here, we have adapted a recently described phosphoproteomic approach to neuronal cultures to characterize the phosphoproteomes generated by these neurotrophic factors. Unexpectedly, IGF-1 and PACAP increased the phosphorylation state of a common set of proteins in neurons. Using PACAP type 1 receptor (PAC1R) null mice, we showed that IGF-1 transactivated PAC1Rs constitutively associated with IGF-1 receptors. This effect was mediated by Src family kinases, which induced PAC1R phosphorylation on tyrosine residues. PAC1R transactivation was responsible for a large fraction of the IGF-1-associated phosphoproteome and played a critical role in the antiapoptotic activity of IGF-1. Hence, in contrast to the general opinion that the trophic activity of IGF-1 is solely mediated by tyrosine kinase receptor-associated signalling, we show that it involves a more complex signalling network dependent on the PAC1 Gs-protein-coupled receptor in neurons.
Collapse
Affiliation(s)
- Nicolas Delcourt
- CNRS UMR 5203, Montpellier, France
- INSERM, U661, Montpellier, France
- University Montpellier I, Montpellier, France
- University Montpellier II, Montpellier, France
- Département de Neurobiologie, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Eric Thouvenot
- CNRS UMR 5203, Montpellier, France
- INSERM, U661, Montpellier, France
- University Montpellier I, Montpellier, France
- University Montpellier II, Montpellier, France
- Département de Neurobiologie, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Benjamin Chanrion
- CNRS UMR 5203, Montpellier, France
- INSERM, U661, Montpellier, France
- University Montpellier I, Montpellier, France
- University Montpellier II, Montpellier, France
- Département de Neurobiologie, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Nathalie Galéotti
- CNRS UMR 5203, Montpellier, France
- INSERM, U661, Montpellier, France
- University Montpellier I, Montpellier, France
- University Montpellier II, Montpellier, France
- Département de Neurobiologie, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Patrick Jouin
- CNRS UMR 5203, Montpellier, France
- INSERM, U661, Montpellier, France
- University Montpellier I, Montpellier, France
- University Montpellier II, Montpellier, France
- Département de Neurobiologie, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Joël Bockaert
- CNRS UMR 5203, Montpellier, France
- INSERM, U661, Montpellier, France
- University Montpellier I, Montpellier, France
- University Montpellier II, Montpellier, France
- Département de Neurobiologie, Institut de Génomique Fonctionnelle, Montpellier, France
- Département de Neurobiologie, Institut de Génomique Fonctionnelle, 141 Rue de la Cardonille, 34094 Montpellier Cedex 5, France. Tel.: +33 467 14 29 30; Fax: +33 467 14 29 10; E-mail:
| | - Philippe Marin
- CNRS UMR 5203, Montpellier, France
- INSERM, U661, Montpellier, France
- University Montpellier I, Montpellier, France
- University Montpellier II, Montpellier, France
- Département de Neurobiologie, Institut de Génomique Fonctionnelle, Montpellier, France
| |
Collapse
|
38
|
Burke K, Cheng Y, Li B, Petrov A, Joshi P, Berman R, Reuhl KR, DiCicco-Bloom E. Methylmercury elicits rapid inhibition of cell proliferation in the developing brain and decreases cell cycle regulator, cyclin E. Neurotoxicology 2006; 27:970-81. [PMID: 17056119 PMCID: PMC2013736 DOI: 10.1016/j.neuro.2006.09.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 08/21/2006] [Accepted: 09/08/2006] [Indexed: 01/12/2023]
Abstract
The developing brain is highly sensitive to methylmercury (MeHg). Still, the initial changes in cell proliferation that may contribute to long-term MeHg effects are largely undefined. Our previous studies with growth factors indicate that acute alterations of the G1/S-phase transition can permanently affect cell numbers and organ size. Therefore, we determined whether an environmental toxicant could also impact brain development with rapid (6-7h) effects on DNA synthesis and cell cycle machinery in neuronal precursors. In vivo studies in newborn rat hippocampus and cerebellum, two regions of postnatal neurogenesis, were followed by in vitro analysis of two precursor models, cortical and cerebellar cells, focusing on the proteins that regulate the G1/S transition. In postnatal day 7 (P7) pups, a single subcutaneous injection of MeHg (3microg/g) acutely (7h) decreased DNA synthesis in the hippocampus by 40% and produced long-term (2 weeks) reductions in total cell number, estimated by DNA quantification. Surprisingly, cerebellar granule cells were resistant to MeHg effects in vivo at comparable tissue concentrations, suggesting region-specific differences in precursor populations. In vitro, MeHg altered proliferation and cell viability, with DNA synthesis selectively inhibited at an early timepoint (6h) corresponding to our in vivo observations. Considering that G1/S regulators are targets of exogenous signals, we used a well-defined cortical cell model to examine MeHg effects on relevant cyclin-dependent kinases (CDK) and CDK inhibitors. At 6h, MeHg decreased by 75% levels of cyclin E, a cell cycle regulator with roles in proliferation and apoptosis, without altering p57, p27, or CDK2 nor levels of activated caspase 3. In aggregate, our observations identify the G1/S transition as an early target of MeHg toxicity and raise the possibility that cyclin E degradation contributes to both decreased proliferation and eventual cell death.
Collapse
Affiliation(s)
- Kelly Burke
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Yinghong Cheng
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Baogang Li
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Alex Petrov
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Pushkar Joshi
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Robert Berman
- Department of Neurological Surgery, University of California at Davis
| | | | - Emanuel DiCicco-Bloom
- Department of Neuroscience & Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey
- Department of Pediatrics; Member of the Cancer Institute of New Jersey
| |
Collapse
|
39
|
Georgopoulou N, Hurel C, Politis PK, Gaitanou M, Matsas R, Thomaidou D. BM88 is a dual function molecule inducing cell cycle exit and neuronal differentiation of neuroblastoma cells via cyclin D1 down-regulation and retinoblastoma protein hypophosphorylation. J Biol Chem 2006; 281:33606-33620. [PMID: 16893893 DOI: 10.1074/jbc.m602689200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Control of cell cycle progression/exit and differentiation of neuronal precursors is of paramount importance during brain development. BM88 is a neuronal protein associated with terminal neuron-generating divisions in vivo and is implicated in mechanisms underlying neuronal differentiation. Here we have used mouse neuroblastoma Neuro 2a cells as an in vitro model of neuronal differentiation to dissect the functional properties of BM88 by implementing gain- and loss-of-function approaches. We demonstrate that stably transfected cells overexpressing BM88 acquire a neuronal phenotype in the absence of external stimuli, as judged by enhanced expression of neuronal markers and neurite outgrowth-inducing signaling molecules. In addition, cell cycle measurements involving cell growth assays, BrdUrd incorporation, and fluorescence-activated cell sorting analysis revealed that the BM88-transfected cells have a prolonged G(1) phase, most probably corresponding to cell cycle exit at the G(0) restriction point, as compared with controls. BM88 overexpression also results in increased levels of the cell cycle regulatory protein p53, and accumulation of the hypophosphorylated form of the retinoblastoma protein leading to cell cycle arrest, with concomitant decreased levels and, in many cells, cytoplasmic localization of cyclin D1. Conversely, BM88 gene silencing using RNA interference experiments resulted in acceleration of cell proliferation accompanied by impairment of retinoic acid-induced neuronal differentiation of Neuro 2a cells. Taken together, our results suggest that BM88 plays an essential role in regulating cell cycle exit and differentiation of Neuro 2a cells toward a neuronal phenotype and further support its involvement in the proliferation/differentiation transition of neural stem/progenitor cells during embryonic development.
Collapse
Affiliation(s)
- Niki Georgopoulou
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 115 21 Athens, Greece
| | | | | | | | | | | |
Collapse
|
40
|
Ye W, Mairet-Coello G, DiCicco-Bloom E. DNAse I pre-treatment markedly enhances detection of nuclear cyclin-dependent kinase inhibitor p57Kip2 and BrdU double immunostaining in embryonic rat brain. Histochem Cell Biol 2006; 127:195-203. [PMID: 17024454 DOI: 10.1007/s00418-006-0238-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2006] [Indexed: 11/27/2022]
Abstract
As a member of the CIP/KIP family of cyclin-dependent kinase inhibitors (CKIs), p57Kip2 binds tightly to G1 cyclin/cyclin-dependent kinase complexes to block cell cycle progression. CKIs play critical roles in regulating the transition from proliferation to differentiation in many tissues, including the nervous system. Conversely, CKI dys-regulation contributes to neoplasia and cancer progression. While the combined detection of CKI immunoreactivity and S phase entry using bromodeoxyuridine (BrdU) incorporation may be particularly informative, successful immunostaining may be limited due to "masked" antigen epitopes and acid-induced signal degradation. We now report an improved double immunofluorescent method for detecting p57Kip2 and BrdU in paraformaldehyde-fixed frozen sections of embryonic rat brain. We substituted deoxyribonuclease I (DNAse I) for HCl pre-treatment to expose antigenic sites in frozen sections, and employed a biotinylated tyramide-based system to enhance p57Kip2 visualization. We identified a time- and dose-dependent relationship between DNAse I treatment and double labeling of p57Kip2 and BrdU, increasing both the numbers and intensities of immunopositive nuclei. With excess DNAse I treatment, however, there was signal degradation for both BrdU and total DNA, as reflected by DAPI staining. The use of DNAse I pre-treatment significantly increases the reliability and sensitivity of immunodetection of CKI nuclear factors, and should be useful for both developmental neurobiology studies as well as cancer diagnostic applications.
Collapse
Affiliation(s)
- Weizhen Ye
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, 675 Hoes Lane, RWJ SPH 362, Piscataway, NJ, 08854, USA,
| | | | | |
Collapse
|
41
|
The effects of PACAP on neural cell proliferation. ACTA ACUST UNITED AC 2006; 137:50-7. [PMID: 17011642 DOI: 10.1016/j.regpep.2006.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 03/20/2006] [Accepted: 03/30/2006] [Indexed: 01/25/2023]
Abstract
PACAP and its receptors are expressed in growth zones of the brain. By stimulating PAC(1)-receptors PACAP can enhance, as well as reduce, the proliferation rate in a cell-type dependent manner. PACAP can enhance the proliferation rate by activating phospholipase C and protein kinase C, although other signal transduction pathways may also be responsible. PACAP can suppress proliferation by inhibiting protein complexes of the cyclins D and E with the cyclin-dependent kinases 4/6 and 2, respectively, which are necessary for entry into the cell cycle. PACAP seems to exert these inhibitory effects by acting via the Sonic hedgehog glycoprotein and the small GTPase RhoA. Also, the activation of a cyclin-dependent kinase inhibitor has been suggested. The signal transduction pathways mediating the effects of PACAP on proliferation are discussed.
Collapse
|
42
|
Martins RAP, Linden R, Dyer MA. Glutamate regulates retinal progenitors cells proliferation during development. Eur J Neurosci 2006; 24:969-80. [PMID: 16925590 DOI: 10.1111/j.1460-9568.2006.04966.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The precise coordination of cell cycle exit and cell fate specification is essential for generating the correct proportion of retinal cell types during development. The decision to exit the cell cycle is regulated by intrinsic and extrinsic cues. There is growing evidence that neurotransmitters can regulate cell proliferation and cell fate specification during the early stages of CNS development prior to the formation of synaptic connections. We found that the excitatory neurotransmitter glutamate regulates retinal progenitor cell proliferation during embryonic development of the mouse. AMPA/kainate and N-methyl-d-aspartate receptors are expressed in embryonic retinal progenitor cells. Addition of exogenous glutamate leads to a dose-dependent decrease in cell proliferation without inducing cell death or activating the p53 pathway. Activation of AMPA/kainate receptors induced retinal progenitor cells to prematurely exit the cell cycle. Using a replication-incompetent retrovirus to follow the clonal expansion of individual retinal progenitor cells, it was observed that blockade of AMPA/kainate receptors increased the proportion of large clones, showing that modulation of endogenous glutamatergic activity can have long-term consequences on retinal cell proliferation. Real time reverse transcriptase-polymerase chain reaction and immunoblot analyses demonstrated that glutamate does not alter the levels of the mRNA and proteins that regulate the G1/S-phase transition. Instead, the activity of the Cdk2 kinase is reduced in the presence of glutamate. These data indicate that glutamate regulates retinal progenitor cell proliferation by post-translational modulation of cyclin/Cdk2 kinase activity.
Collapse
Affiliation(s)
- Rodrigo A P Martins
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | |
Collapse
|
43
|
Kim HJ, Hida H, Jung CG, Miura Y, Nishino H. Treatment with deferoxamine increases neurons from neural stem/progenitor cells. Brain Res 2006; 1092:1-15. [PMID: 16697980 DOI: 10.1016/j.brainres.2006.02.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2004] [Revised: 11/24/2005] [Accepted: 02/11/2006] [Indexed: 01/05/2023]
Abstract
Neural transplantation is a promising approach for treating neurodegenerative disease. Neural stem/progenitor cells (NPCs) are self-renewing and multipotent and thus are good candidates for donor cells when they have been clearly defined to differentiate into neurons. As neuronal differentiation follows cell cycle exit, we investigated whether neuron production from NPCs is increased by treatment with cell cycle blockers. NPCs from E12.5 rat ventral mesencephalon were cultured as neurospheres in DMEM/F12 medium containing N2 supplements and bFGF. Treatment of NPCs with deferoxamine, a G1/S phase blocker, increased the number of beta-tubulin III-positive cells after differentiation, concomitant with increases of MAP2 mRNA and protein, and a decrease of GFAP protein. Further, an increase in beta-tubulin III/BrdU double-positive cells and a decrease in GFAP/BrdU double-positive cells were confirmed. In real-time PCR, the expressions of p21(cip1), p27(kip1) and p57(kip2) mRNAs remained unaltered for 8 h after treatment with deferoxamine but were significantly elevated after 1 day. Deferoxamine specifically enhanced the elevation of p27(kip1) mRNA at 1-2 days and the accumulation of p27(kip1) protein at 3 days, along with the activation of neuroD promoter and the elevation of neuroD mRNA. Transfection of p27(kip1) into NPCs induced activation of neuroD promoter and increase of number of beta-tubulin III-positive cells. These data suggest that pretreatment with deferoxamine increases the number of neurons from NPCs related to prolonged p27(kip1) elevation and activation of the neuroD signaling pathway. In this way, regulation of the cell cycle should be a useful first step in engineering NPCs for neural transplantation.
Collapse
Affiliation(s)
- Hye-Jung Kim
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya 467-8601, Japan
| | | | | | | | | |
Collapse
|
44
|
Benayed R, Gharani N, Rossman I, Mancuso V, Lazar G, Kamdar S, Bruse SE, Tischfield S, Smith BJ, Zimmerman RA, Dicicco-Bloom E, Brzustowicz LM, Millonig JH. Support for the homeobox transcription factor gene ENGRAILED 2 as an autism spectrum disorder susceptibility locus. Am J Hum Genet 2005; 77:851-868. [PMID: 16252243 PMCID: PMC1271392 DOI: 10.1086/497705] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2005] [Accepted: 08/26/2005] [Indexed: 11/03/2022] Open
Abstract
Our previous research involving 167 nuclear families from the Autism Genetic Resource Exchange (AGRE) demonstrated that two intronic SNPs, rs1861972 and rs1861973, in the homeodomain transcription factor gene ENGRAILED 2 (EN2) are significantly associated with autism spectrum disorder (ASD). In this study, significant replication of association for rs1861972 and rs1861973 is reported for two additional data sets: an independent set of 222 AGRE families (rs1861972-rs1861973 haplotype, P=.0016) and a separate sample of 129 National Institutes of Mental Health families (rs1861972-rs1861973 haplotype, P=.0431). Association analysis of the haplotype in the combined sample of both AGRE data sets (389 families) produced a P value of .0000033, whereas combining all three data sets (518 families) produced a P value of .00000035. Population-attributable risk calculations for the associated haplotype, performed using the entire sample of 518 families, determined that the risk allele contributes to as many as 40% of ASD cases in the general population. Linkage disequilibrium (LD) mapping with the use of polymorphisms distributed throughout the gene has shown that only intronic SNPs are in strong LD with rs1861972 and rs1861973. Resequencing and association analysis of all intronic SNPs have identified alleles associated with ASD, which makes them candidates for future functional analysis. Finally, to begin defining the function of EN2 during development, mouse En2 was ectopically expressed in cortical precursors. Fewer En2-transfected cells than controls displayed a differentiated phenotype. Together, these data provide further genetic evidence that EN2 might act as an ASD susceptibility locus, and they suggest that a risk allele that perturbs the spatial/temporal expression of EN2 could significantly alter normal brain development.
Collapse
Affiliation(s)
- Rym Benayed
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ, 08854-5638, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Candal E, Nguyen V, Joly JS, Bourrat F. Expression domains suggest cell-cycle independent roles of growth-arrest molecules in the adult brain of the medaka, Oryzias latipes. Brain Res Bull 2005; 66:426-30. [PMID: 16144625 DOI: 10.1016/j.brainresbull.2005.05.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Teleost fish are unique for their enormous potential to produce new neurons in the adult brain. Nevertheless, the regulation of this adult neurogenesis remains to be characterized. Does it resort to the same molecular mechanisms as those at play in embryonic development? Here, we analyse the expression of the neurogenic gene Ol-DeltaA in the brain of medaka (Oryzias latipes) embryos and adults. To determine the relationships between neurogenic and growth-arrest genes in the adult brain, we compare the expression domains of Ol-DeltaA with those of Ol-KIP and Ol-Gadd45gamma, two well-characterized genes involved in cell-cycle arrest and growth inhibition. While it is widely assumed that genes controlling cell-cycle exit show restricted expression domains next to proliferating cells (in the sites of prospective cell differentiation), we observe highly particular expression domains of Ol-KIP and Ol-Gadd45gamma not associated to proliferating areas of the adult brain, suggesting locally different and cell-cycle independent roles of these molecules in the adult brain.
Collapse
Affiliation(s)
- Eva Candal
- JE INRA U1126, INRA/CNRS Group, UPR 2197 DEPSN, CNRS, Institut de Neurosciences A. Fessard, Gif-sur-Yvette, France.
| | | | | | | |
Collapse
|
46
|
Davidovics Z, DiCicco-Bloom E. Moderate lead exposure elicits neurotrophic effects in cerebral cortical precursor cells in culture. J Neurosci Res 2005; 80:817-25. [PMID: 15884012 DOI: 10.1002/jnr.20539] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Lead (Pb) persists as an environmental toxicant despite aggressive environmental and occupational regulation. Neurotoxicological effects of acute Pb poisoning range from subtle cognitive deficits, to clumsiness and ataxia, to coma and seizures. In adult neurotoxicity, reductions of blood Pb levels are often associated with reversal of clinical signs. In children, however, the effects are more likely to endure, with even low levels of chronic Pb exposure correlating with decreasing IQ. These persistent effects likely result from neurodevelopmental insults, such as altered cell survival or maturation, although the mechanisms have not been fully defined. In the present study we define the effects of moderate-level Pb exposure on mammalian neurogenesis using a well-characterized cortical precursor model. Gestational day 14.5 rat cerebral cortical precursor cells were cultured in defined media and cell number, precursor proliferation, apoptosis, and neuritic process outgrowth were assessed following exposure to a range of Pb acetate concentrations. Surprisingly, whereas a concentration of 30 microg/ml Pb acetate was acutely toxic to neurons, concentrations between 1 and 10 microg/ml Pb acetate (approximately 3 microM and 30 microM Pb, respectively) increased cell number: 10 times as many cells exposed to 10 microg/ml Pb were present on day 4 as compared to control. The increase in cell number was not a result of increased proliferation, however, as DNA synthesis did not increase. Rather, Pb exposure maintained the survival of cortical precursors, as the progressive apoptosis occurring under control conditions was markedly reduced by the metal. Additionally, neuritic process initiation and outgrowth increased in a concentration-dependent manner, with processes four times as abundant on day 1 and twice as long on day 2. These results suggest that brief exposure to lead during neurogenesis directly affects cell survival and process development, potentially altering cortical arrangement. Consequently, alterations in neural circuitry may underlie some of the neurological effects of Pb exposure during brain development.
Collapse
Affiliation(s)
- Zev Davidovics
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, 08854, USA
| | | |
Collapse
|
47
|
Riaz SS, Bradford HF. Factors involved in the determination of the neurotransmitter phenotype of developing neurons of the CNS: Applications in cell replacement treatment for Parkinson's disease. Prog Neurobiol 2005; 76:257-78. [PMID: 16256257 DOI: 10.1016/j.pneurobio.2005.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2003] [Revised: 06/07/2005] [Accepted: 08/04/2005] [Indexed: 02/08/2023]
Abstract
The developmental stages involved in the conversion of stem cells to fully functional neurons of specific neurotransmitter phenotype are complex and not fully understood. Over the past decade many studies have been published that demonstrate that in vitro manipulation of the epigenetic environment of the stem cells allows experimental control of final neuronal phenotypic choice. This review presents the evidence for the involvement of a number of endogenous neurobiochemicals, which have been reported to potently influence DAergic (and other neurotransmitter) phenotype expression in vitro. They act at different stages on the pathway to neurotransmitter phenotype determination, and in different ways. Many are better known for their involvement in other aspects of development, and in other biochemical roles. Their proper place, and precise roles, in neurotransmitter phenotype determination in vivo will no doubt be determined in the future. Meanwhile, considerable medical benefits are offered from producing large, long-term, viable cryostores of self-regenerating multipotential neural precursor cells (i.e., brain stem cells), which can be used for cell replacement therapies in the treatment of degenerative brain diseases, such as Parkinson's disease.
Collapse
Affiliation(s)
- S S Riaz
- Department of Biological Sciences, Imperial College of Science, Technology and Medicine, Biochemistry Building, South Kensington Campus, Imperial College Road, SW7 2AZ London, UK
| | | |
Collapse
|
48
|
Meyer DK, Fischer C, Becker U, Göttsching I, Boutillier S, Baermann C, Schmidt G, Klugbauer N, Leemhuis J. Pituitary Adenylyl Cyclase-activating Polypeptide 38 Reduces Astroglial Proliferation by Inhibiting the GTPase RhoA. J Biol Chem 2005; 280:25258-66. [PMID: 15870074 DOI: 10.1074/jbc.m501630200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pituitary adenylyl cyclase-activating polypeptide 38 (PACAP38) plays an important role in the proliferation and differentiation of neural cells. In the present study, we have investigated how PACAP38 inhibits the proliferation of cultured neocortical astroglial cells. When applied to synchronized cells during the G(1) phase of the cell cycle, PACAP38 diminished the subsequent nuclear uptake of bromodeoxyuridine. When applied for 2 days, it reduced the cell number. PACAP38 did not exert its antiproliferative effect by activating protein kinase A. It also did not reduce the activity of mitogen-activated protein kinases essential for G(1) phase progression. Instead, PACAP38 acted on a member of the Rho family of small GTPases. It reduced the activity of RhoA as was shown with a Rhotekin pull-down assay. The decrease in endogenous RhoA activity induced by treatment of the cells with C3 exotoxin or by expression of dominant negative RhoA also reduced the nuclear uptake of bromodeoxyuridine. In contrast, expression of constitutively active RhoA prevented the effect of PACAP38. Our data show a novel signal transduction pathway by which the neuropeptide influences cell proliferation.
Collapse
Affiliation(s)
- Dieter K Meyer
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Zentrum für Neurowissenschaften, Albert-Ludwigs-Universität, D-79104 Freiburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Borba JC, Henze IP, Silveira MS, Kubrusly RCC, Gardino PF, de Mello MCF, Hokoç JN, de Mello FG. Pituitary adenylate cyclase-activating polypeptide (PACAP) can act as determinant of the tyrosine hydroxylase phenotype of dopaminergic cells during retina development. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 156:193-201. [PMID: 16099306 DOI: 10.1016/j.devbrainres.2005.02.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2004] [Revised: 02/21/2005] [Accepted: 02/27/2005] [Indexed: 11/16/2022]
Abstract
In the chick retina, dopaminergic cells are generated between embryonic days 3 and 7 (E3/E7). However, the expression of tyrosine hydroxylase (TH), the first enzyme in the catecholamine synthetic pathway, is only detected after E11/E12. During the interval comprising E7 to E12, signals conveyed by cAMP are important to determine the TH phenotype. The present study shows that pituitary adenylyl cyclase-activating polypeptide (PACAP), via cAMP, is a major endogenous component in defining the TH phenotype of retina dopaminergic cells during development. PACAP type 1 receptor and its mRNA were detected in retinas since E6. PACAP was also immunodetected in cells localized in the inner nuclear layer of retinas since E8. This peptide promoted greater than 10-fold increase in cAMP accumulation of retinas obtained from embryos since E8, an effect that was blocked by PACAP6-38 (PAC1 receptor antagonist). In cultured retina cells from E8 and E9, maintained for 6 days in vitro with 10 nM PACAP (for 5 days), the number of dopaminergic cells expressing tyrosine hydroxylase increased 2.4-fold. The cAMP analog, 8-Br-cAMP and 3-isobutyl-1-methylxanthine (IBMX, a phosphodiesterase inhibitor) also increased the number of tyrosine hydroxylase-positive cells by 4- to 6-fold. IBMX plus PACAP treatment resulted in 17-fold increase in the number of cells positive for tyrosine hydroxylase. Under this condition the amount of tyrosine hydroxylase expression, as detected by western blot analysis, was also increased. The protein kinase-A inhibitor, rp-cAMPS, significantly reduced the effect of PACAP. Our data show that this peptide is an important factor influencing the definition of the tyrosine hydroxylase phenotype of retina dopaminergic cells within a narrow window of development.
Collapse
MESH Headings
- 1-Methyl-3-isobutylxanthine/pharmacology
- 8-Bromo Cyclic Adenosine Monophosphate/pharmacology
- Age Factors
- Animals
- Animals, Newborn
- Blotting, Western/methods
- Cell Count/methods
- Cell Culture Techniques
- Chick Embryo
- Colforsin/pharmacology
- Cyclic AMP/metabolism
- Dopamine/metabolism
- Dose-Response Relationship, Drug
- Drug Interactions
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/physiology
- Immunohistochemistry/methods
- Microscopy, Confocal/methods
- Nerve Growth Factors/antagonists & inhibitors
- Nerve Growth Factors/physiology
- Neurons/drug effects
- Neurons/enzymology
- Neurons/metabolism
- Neuropeptides/antagonists & inhibitors
- Neuropeptides/physiology
- Neurotransmitter Agents/antagonists & inhibitors
- Neurotransmitter Agents/physiology
- Phenotype
- Phosphodiesterase Inhibitors/pharmacology
- Pituitary Adenylate Cyclase-Activating Polypeptide
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I
- Retina/cytology
- Retina/embryology
- Retina/enzymology
- Retina/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Time Factors
- Tyrosine 3-Monooxygenase/genetics
- Tyrosine 3-Monooxygenase/metabolism
Collapse
Affiliation(s)
- Juliana Carrazzoni Borba
- Laboratórios de Neuroquímica and Neurobiologia da Retina, Instituto de Biofísica Carlos Chagas Filho-UFRJ, Centro de Ciências da Saúde-Bloco G, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, RJ 21949-900, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Li B, DiCicco-Bloom E. Basic fibroblast growth factor exhibits dual and rapid regulation of cyclin D1 and p27 to stimulate proliferation of rat cerebral cortical precursors. Dev Neurosci 2005; 26:197-207. [PMID: 15711060 DOI: 10.1159/000082137] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2004] [Accepted: 07/12/2004] [Indexed: 02/05/2023] Open
Abstract
While extracellular signals play a major role in brain neurogenesis, little is known about the cell cycle machinery underlying mitogen stimulation of precursor proliferation. Current models suggest that the D cyclins function as primary sensors of extracellular mitogens. Here we define the mechanisms by which basic fibroblast growth factor (bFGF) stimulates cortical precursors, with particular attention to the responses of cell cycle promitogenic and antimitogenic regulators. bFGF produced a 4-fold increase in DNA synthesis and a 3-fold rise in bromodeoxyuridine labeling, suggesting that the factor promotes the G1/S transition. There was also a 3-fold increase in cyclin-dependent kinase 2 (CDK2) kinase activity, which is critical for S phase entry. CDK2 activation was apparently cyclin E dependent, since only its protein and mRNA levels were elevated at 24 h, whereas CDK2, p27KIP1 and p57KIP2 levels were unaltered. Late G1 phase CDK2/cyclin E activity depends on early G1 D cyclin function. Indeed, cyclin D1, but not cyclin D3, was upregulated selectively at 8 h after bFGF treatment, a time when cyclin E was unchanged. The sequential activation of cyclin D1 and cyclin E supports the idea that cyclin E gene transcription is regulated by cyclin-D/CDK4/6-mediated pRb phosphorylation and subsequent E2F transcription factor release. However, in addition to increased D1 cyclin, we unexpectedly detected a 75% reduction in p27KIP1 protein at 8 h, suggesting that both pro- and antimitogenic regulators are targets of extracellular mitogens during brain development.
Collapse
Affiliation(s)
- Baogang Li
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | | |
Collapse
|