51
|
Sensory defects in Necdin deficient mice result from a loss of sensory neurons correlated within an increase of developmental programmed cell death. BMC DEVELOPMENTAL BIOLOGY 2006; 6:56. [PMID: 17116257 PMCID: PMC1687209 DOI: 10.1186/1471-213x-6-56] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Accepted: 11/20/2006] [Indexed: 11/29/2022]
Abstract
Background The human NECDIN gene is involved in a neurodevelopmental disorder, Prader-Willi syndrome (PWS). Previously we reported a mouse Necdin knock-out model with similar defects to PWS patients. Despite the putative roles attributed to Necdin, mainly from in vitro studies, its in vivo function remains unclear. In this study, we investigate sensory-motor behaviour in Necdin deficient mice. We reveal cellular defects and analyse their cause. Results We report sensory differences in Necdin deficient mice compared to wild type animals. These differences led us to investigate sensory neuron development in Necdin deficient mouse embryos. First, we describe the expression pattern of Necdin in developing DRGs and report a reduction of one-third in specified sensory neurons in dorsal roots ganglia and show that this neuronal loss is achieved by E13.5, when DRGs sensory neurons are specified. In parallel, we observed an increase of 41% in neuronal apoptosis during the wave of naturally occurring cell death at E12.5. Since it is assumed that Necdin is a P75NTR interactor, we looked at the P75NTR-expressing cell population in Necdin knock-out embryos. Unexpectedly, Necdin loss of function has no effect on p75NTR expressing neurons suggesting no direct genetic interaction between Necdin and P75NTR in this context. Although we exclude a role of Necdin in axonal outgrowth from spinal sensory neurons in early developmental stages; such a role could occur later in neuronal differentiation. Finally we also exclude an anti-proliferative role of Necdin in developing sensory neurons. Conclusion Overall, our data show clearly that, in early development of the nervous system, Necdin is an anti-apoptotic or survival factor.
Collapse
|
52
|
Galderisi U, Cipollaro M, Giordano A. The retinoblastoma gene is involved in multiple aspects of stem cell biology. Oncogene 2006; 25:5250-5256. [PMID: 16936744 DOI: 10.1038/sj.onc.1209736] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Genetic programs controlling self-renewal and multipotentiality of stem cells have overlapping pathways with cell cycle regulation. Components of cell cycle machinery can play a key role in regulating stem cell self-renewal, proliferation, differentiation and aging. Among the negative regulators of cell cycle progression, the RB family members play a prominent role in controlling several aspects of stem cell biology. Stem cells contribute to tissue homeostasis and must have molecular mechanisms that prevent senescence and hold 'stemness'. RB can induce senescence-associated changes in gene expression and its activity is downregulated in stem cells to preserve self-renewal. Several reports evidenced that RB could play a role in lineage specification of several types of stem cells. RB has a role in myogenesis as well as in cardiogenesis. These effects are not only related to its role in suppressing E2F-responsive genes but also to its ability to modulate the activity of tissue-specific transcription factors. RB is also involved in adipogenesis through a strict control of lineage commitment and differentiation of adipocytes as well in determining the switch between brown and white adipocytes. Also, hematopoietic progenitor cells utilize the RB pathway to modulate cell commitment and differentiation. In this review, we will also discuss the role of the other two RB family members: Rb2/p130 and p107 showing that they have both specific and overlapping functions with RB gene.
Collapse
Affiliation(s)
- U Galderisi
- Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| | | | | |
Collapse
|
53
|
Abstract
The retinoblastoma susceptibility gene was the first tumor suppressor gene identified in humans and the first tumor suppressor gene knocked out by targeted deletion in mice. RB serves as a transducer between the cell cycle machinery and promoter-specific transcription factors, its most documented activity being the repression of the E2F family of transcription factors, which regulate the expression of genes involved in cell proliferation and survival. Recent investigations of RB function suggest that it works as a fundamental regulator to coordinate pathways of cellular growth and differentiation. In this review, we unravel the novel role of an equally important aspect of RB in downregulating the differentiation inhibitor EID-1 during cellular differentiation by teasing apart the signal, which elicit differentiation and limit cell cycle progression, since the molecular mechanisms relating to RB activation of differentiation is much less understood. We review the various roles for RB in differentiation of neurons, muscle, adipose tissue, and the retina. In addition, we provide an update for the current models of the role of RB in cell cycle to entry and exit, extending the view toward chromatin remodeling and expose the dichotomies in the regulation of RB family members. We conclude with a discussion of a novel RB regulatory network, incorporating the dynamic contribution of EID family proteins.
Collapse
Affiliation(s)
- L Khidr
- Department of Biological Chemistry, University of California-Irvine Med Sci 1, Irvine, CA 92697, USA
| | | |
Collapse
|
54
|
Agromayor M, Wloga E, Naglieri B, Abrashkin J, Verma K, Yamasaki L. Visualizing dynamic E2F-mediated repression in vivo. Mol Cell Biol 2006; 26:4448-61. [PMID: 16738312 PMCID: PMC1489115 DOI: 10.1128/mcb.02101-05] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Revised: 11/30/2005] [Accepted: 03/12/2006] [Indexed: 11/20/2022] Open
Abstract
Although many E2F target genes have been identified recently, very little is known about how any single E2F site controls the expression of an E2F target gene in vivo. To test the requirement for a single E2F site in vivo and to learn how E2F-mediated repression is regulated during development and tumorigenesis, we have constructed a novel series of wild-type and mutant Rb promoter-LacZ transgenic reporter lines that allow us to visualize the activity of a crucial E2F target in vivo, the retinoblastoma tumor suppressor gene (Rb). Two mutant Rb promoter-LacZ constructs were used to evaluate the importance of a single E2F site or a nearby activator (Sp1/Ets) site that is found mutated in low-penetrance retinoblastomas. The activity of the wild-type Rb promoter is dynamic, varying spatially and temporally within the developing nervous system. While loss of the activator site silences the Rb promoter, loss of the E2F site stimulates its activity in the neocortex, retina, and trigeminal ganglion. Surprisingly, E2F-mediated repression of Rb does not act globally or in a static manner but, instead, is a highly dynamic process in vivo. Using neocortical extracts, we detected GA-binding protein alpha (GABPalpha, an Ets family member) bound to the activator site and both E2F1 and E2F4 bound to the repressor site of the Rb promoter in vitro. Additionally, we detected binding of both E2F1 and E2F4 to the Rb promoter in vivo using chromatin immunoprecipitation analysis on embryonic day 13.5 brain. Unexpectedly, we detect no evidence for Rb promoter autoregulation in neuroendocrine tumors from Rb+/-; RbP-LacZ mice that undergo loss of heterozygosity at the Rb locus, in contrast to the situation in human retinoblastomas where high RB mRNA levels are found. In summary, this study provides the first demonstration that loss of an E2F site is critical for target gene repression in vivo and underscores the complexity of the Rb and E2F family network in vivo.
Collapse
Affiliation(s)
- Monica Agromayor
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Avenue, 1102 Fairchild Building, Mail Code 2428, New York, NY 10027, USA
| | | | | | | | | | | |
Collapse
|
55
|
Cicero S, Herrup K. Cyclin-dependent kinase 5 is essential for neuronal cell cycle arrest and differentiation. J Neurosci 2006; 25:9658-68. [PMID: 16237170 PMCID: PMC6725732 DOI: 10.1523/jneurosci.1773-05.2005] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a serine/threonine kinase with significant homology to cell cycle-related Cdks but is not believed to be active in a typical cell cycle. In Cdk5-deficient embryos and Cdk5 chimeras, migration and survival of postmitotic neurons is compromised in a cell-autonomous manner. In the present study, we show that loss of Cdk5 leads to both failure of neuronal differentiation and loss of cell cycle control. Using specific cytoskeletal proteins as indices of neuronal differentiation, we find that Cdk5-deficient neurons are significantly arrested or delayed in their developmental program both in vivo and in vitro. For example, immunocytochemistry of embryonic day 16 (E16) cortex reveals that the expression of microtubule-associated protein 2c (Map-2c), a marker of mature neurons, is nearly absent in Cdk5(-/-) cells that have migrated to the cortical plate while these same cells continue to express nestin. Similarly, in vitro, Map-2-positive cells are rare in cultures from E16 Cdk5(-/-) embryos. Cell cycle control is also deficient in Cdk5(-/-) cells. In vivo, neurons engaged in cell cycle activities are found in the cortical plate, and, in vitro, class III beta-tubulin-positive cells continue to label with bromodeoxyuridine even after 5 d of incubation. Transfection of a wild-type Cdk5 construct reveals that cell cycle control can be regained in Cdk5(-/-) cells by overexpression of Cdk5. These data indicate that Cdk5 is necessary for both neuronal differentiation and cell cycle inhibition.
Collapse
Affiliation(s)
- Samantha Cicero
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | | |
Collapse
|
56
|
Kuwako KI, Hosokawa A, Nishimura I, Uetsuki T, Yamada M, Nada S, Okada M, Yoshikawa K. Disruption of the paternal necdin gene diminishes TrkA signaling for sensory neuron survival. J Neurosci 2006; 25:7090-9. [PMID: 16049186 PMCID: PMC6724840 DOI: 10.1523/jneurosci.2083-05.2005] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Necdin is a multifunctional signaling protein that stabilizes terminal differentiation of postmitotic neurons. The human necdin gene in chromosome 15q11-q12 is maternally imprinted, paternally transcribed, and not expressed in Prader-Willi syndrome, a human genomic imprinting-associated neurodevelopmental disorder. Although necdin-deficient mice display several abnormal phenotypes reminiscent of this syndrome, little is known about molecular mechanisms that lead to the neurodevelopmental defects. Here, we demonstrate that paternally expressed necdin is required for physiological development of nerve growth factor (NGF)-dependent sensory neurons. Mouse embryos defective in the paternal necdin allele displayed absent necdin expression in the dorsal root ganglia, in which the tropomyosin-related kinase A (TrkA) receptor tyrosine kinase and the p75 neurotrophin receptor were expressed in a normal manner. Necdin interacted with both TrkA and p75 to facilitate the association between these receptors. NGF-induced phosphorylation of TrkA and mitogen-activated protein kinase was significantly diminished in the necdin-null sensory ganglia. Furthermore, the mice lacking the paternal necdin allele displayed augmented apoptosis in the sensory ganglia in vivo and had a reduced population of substance P-containing neurons. These mutant mice showed significantly high tolerance to thermal pain, which is often seen in individuals with Prader-Willi syndrome. These results suggest that paternally expressed necdin facilitates TrkA signaling to promote the survival of NGF-dependent nociceptive neurons.
Collapse
Affiliation(s)
- Ken-ichiro Kuwako
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Chong ZZ, Li F, Maiese K. Attempted cell cycle induction in post-mitotic neurons occurs in early and late apoptotic programs through Rb, E2F1, and caspase 3. Curr Neurovasc Res 2006; 3:25-39. [PMID: 16472123 PMCID: PMC1986668 DOI: 10.2174/156720206775541741] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Either the absence or dysfunction of a number of critical pathways, such as those that involve the nuclear retinoblastoma protein (Rb) and the transcription factor E2F1, may account for the aberrant induction of the cell cycle in post-mitotic neurons that can be responsible for oxidative stress-induced apoptotic cellular destruction. Yet, it is unclear whether early programs of apoptotic injury that involve membrane phosphatidylserine (PS) exposure and calreticulin expression as well as later phases of apoptotic injury with nuclear DNA injury require the critical modulation of Rb and E2F1. We demonstrate that both the post-translational of phosphorylation of Rb to prevent E2F1 transcription as well as the protein integrity of Rb are closely aligned with the modulation of cell cycle induction in post mitotic neurons during oxidative stress. More importantly, we illustrate that both the initial onset of apoptosis with either membrane PS exposure or calreticulin analysis as well as the more terminal phases of apoptosis that involve nuclear DNA degradation proceed concurrently in the same neuronal cells with cell cycle induction. Progression of attempted cell cycle induction is closely associated with the phosphorylation of Rb, its inability to bind to E2F1, and the degradation of the Rb protein. Inhibition of Rb phosphorylation using cyclin dependent kinase inhibitors maintains the integrity of the E2F1/Rb complex and is neuroprotective during free radical exposure. Furthermore, maintenance of the integrity of the Rb protein is specifically dependent upon caspase 3-like activity, since caspase 3 can cleave Rb during free radical activity and this degradation of Rb can be blocked during the inhibition of caspase 3 activity. Our studies not only highlight the critical role of attempted cell cycle induction during oxidative stress-induced neuronal apoptotic injury, but also bring to light the significant impact of the Rb and E2F1 pathways upon early apoptotic programs that can directly influence both intrinsic cell survival as well as extrinsic inflammatory cell activation.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Faqi Li
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
- Departments of Neurology and Anatomy & Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201
- Center for Molecular and Cellular Toxicology, Wayne State University School of Medicine, Detroit, Michigan 48201
| |
Collapse
|
58
|
Battelli C, Nikopoulos GN, Mitchell JG, Verdi JM. The RNA-binding protein Musashi-1 regulates neural development through the translational repression of p21WAF-1. Mol Cell Neurosci 2006; 31:85-96. [PMID: 16214366 DOI: 10.1016/j.mcn.2005.09.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 08/26/2005] [Accepted: 09/06/2005] [Indexed: 11/30/2022] Open
Abstract
RNA-binding proteins regulate cell fate decisions during nervous system development. The Msi family of RNA-binding proteins is expressed in multipotential neural progenitors, and is required for maintaining cells in a proliferative state. We demonstrate that Msi-1's ability to regulate progenitor maintenance is through the translational inhibition of the cyclin-dependent kinase inhibitor p21WAF-1. Msi-1 ectopic expression increases the proliferation rate and the capacity to regulate p21WAF-1 protein expression, independent of p53. The 3' untranslated region (UTR) of the native p21(WAF-1) mRNA contains a Msi-1 consensus-binding site that permits Msi-1 to directly repress the translation of p21WAF-1 protein. Reduction of Msi-1 through antisense leads to aberrant p21WAF-1 expression, which significantly impairs neural differentiation. A double knockdown for p21WAF-1 and Msi-1 rescues the production of mature MAP+ neurons. Our results further elucidate the symbiotic relationship between cell cycle withdrawal and the onset of differentiation in the developing nervous system, as well as increasing the understanding of the influence that RNA-binding proteins serve in regulating these processes.
Collapse
Affiliation(s)
- Chiara Battelli
- The Center for Regenerative Medicine, Maine Medical Center Research Institute, 81 Research Drive Scarborough, ME 04074, USA
| | | | | | | |
Collapse
|
59
|
Murphree AL, Samuel MA, Harbour JW, Mansfield NC. Retinoblastoma. Retina 2006. [DOI: 10.1016/b978-0-323-02598-0.50028-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
60
|
Kim TA, Jiang S, Seng S, Cha K, Avraham HK, Avraham S. The BTB domain of the nuclear matrix protein NRP/B is required for neurite outgrowth. J Cell Sci 2005; 118:5537-48. [PMID: 16306221 DOI: 10.1242/jcs.02643] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The neuronal nuclear matrix protein, NRP/B, contains a BTB domain and kelch repeats and is expressed in primary neurons but not in primary glial cells. To examine the function of NRP/B in neurons, we analyzed the structure/function of the NRP/B-BTB domain and its role in neurite outgrowth. Based on three-dimensional modeling of NRP/B, we generated an NRP/B-BTB mutant containing three mutations in the conserved amino acids D47A, H60A and R61D that was termed BTB mutant A. BTB mutant A significantly reduced the dimerization of NRP/B compared to wild-type NRP/B. The NRP/B-BTB domain was required for nuclear localization and mediated the association of NRP/B with p110RB through the TR subdomain within the B pocket of p110RB. Overexpression of wild-type NRP/B and NRP/B-BTB domain significantly induced neurite outgrowth in PC12 cells and enhanced the G0-G1 cell population by ∼23% compared to the control cells, whereas NRP/B-BTB mutant A reduced neurite outgrowth by 70-80%, and inhibited NRP/B-p110RB association. Single cell microinjection of NRP/B-specific antibodies also blocked the neurite outgrowth of PC12 cells upon NGF stimulation. Interference of NRP/B expression by small interfering RNA (NRP/B-siRNA) inhibited neurite outgrowth and suppressed the NGF-induced outgrowth of neurites in PC12 cells. Additionally, p110RB phosphorylation at serine residue 795 was significantly reduced in PC12 cells treated with NRP/B siRNA compared to those treated with control GFP-siRNA, indicating that p110RB is a downstream target of NRP/B. Thus, the BTB domain of NRP/B regulates neurite outgrowth through its interaction with the TR subdomain within the B pocket of p110RB, and the conserved amino acids D47A, H60A and R61D within this domain of NRP/B are crucial residues for neurite extension in neuronal cells. These findings support a role for the BTB-domain of NRP/B as an important regulator of neuronal differentiation.
Collapse
Affiliation(s)
- Tae-Aug Kim
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Institutes of Medicine, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
61
|
Ferguson KL, McClellan KA, Vanderluit JL, McIntosh WC, Schuurmans C, Polleux F, Slack RS. A cell-autonomous requirement for the cell cycle regulatory protein, Rb, in neuronal migration. EMBO J 2005; 24:4381-91. [PMID: 16308563 PMCID: PMC1356328 DOI: 10.1038/sj.emboj.7600887] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Accepted: 11/03/2005] [Indexed: 12/28/2022] Open
Abstract
Precise cell cycle regulation is critical for nervous system development. To assess the role of the cell cycle regulator, retinoblastoma (Rb) protein, in forebrain development, we studied mice with telencephalon-specific Rb deletions. We examined the role of Rb in neuronal specification and migration of diverse neuronal populations. Although layer specification occurred at the appropriate time in Rb mutants, migration of early-born cortical neurons was perturbed. Consistent with defects in radial migration, neuronal cell death in Rb mutants specifically affected Cajal-Retzius neurons. In the ventral telencephalon, although calbindin- and Lhx6-expressing cortical neurons were generated at embryonic day 12.5, their tangential migration into the neocortex was dramatically and specifically reduced in the mutant marginal zone. Cell transplantation assays revealed that defects in tangential migration arose owing to a cell-autonomous loss of Rb in migrating interneurons and not because of a defective cortical environment. These results revealed a cell-autonomous role for Rb in regulating the tangential migration of cortical interneurons. Taken together, we reveal a novel requirement for the cell cycle protein, Rb, in the regulation of neuronal migration.
Collapse
Affiliation(s)
- Kerry L Ferguson
- Ottawa Health Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Kelly A McClellan
- Ottawa Health Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | - William C McIntosh
- Ottawa Health Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Carol Schuurmans
- Department of Biochemistry & Molecular Biology, University of Calgary, Health Sciences Centre, Calgary, Alberta, Canada
| | - Franck Polleux
- Department of Pharmacology, Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Ruth S Slack
- Ottawa Health Research Institute, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Health Research Institute, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, Canada K1H 8M5. Tel.: +1 613 562 5800; Fax: +1 613 562 5403; E-mail:
| |
Collapse
|
62
|
Weishaupt JH, Klöcker N, Bähr M. Axotomy-induced early down-regulation of POU-IV class transcription factors Brn-3a and Brn-3b in retinal ganglion cells. J Mol Neurosci 2005; 26:17-25. [PMID: 15968082 DOI: 10.1385/jmn:26:1:017] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Accepted: 11/22/2004] [Indexed: 11/11/2022]
Abstract
It has been proposed that neurons being exposed to proapoptotic stimuli undergo dedifferentiation, a process that can either allow for regeneration and axon regrowth or, if remaining incomplete, can force the cell to activate apoptotic pathways. A pivotal step in the differentiation program from neuronal precursor cells to differentiated, postmitotic neurons is their exit from the cell cycle. The POU domain transcription factors Brn-3b and Brn-3a, which are expressed in retinal ganglion cells (RGCs) directly after the exit of RGC precursors from the cell cycle, can be employed as RGC-specific differentiation markers to study potential dedifferentiation of RGCs after axotomy. Here, we examined mRNAand protein expression of Brn-3a and -3b in rat RGCs following axonal lesion. We observed a rapid down-regulation of Brn-3a and -3b protein expression in axotomized RGCs, clearly preceding apoptosis of RGCs. Using real-time PCR, we show that regulation of Brn-3 expression occurred at the transcriptional level. The small subset of RGCs regenerating into a peripheral nerve graft did not (re-)express Brn-3a or -b. In conclusion, we found further evidence supporting the hypothesis of a dedifferentiation process in severed mature neurons. As Brn-3b expression has been shown to be a prerequisite for developmental survival of most RGCs and Brn-3a activates transcription of anti-apoptotic genes, down-regulation of Brn-3 transcription factors might be causally involved in the secondary death of adult RGCs following axotomy.
Collapse
|
63
|
Wen Y, Yang S, Liu R, Simpkins JW. Cell-cycle regulators are involved in transient cerebral ischemia induced neuronal apoptosis in female rats. FEBS Lett 2005; 579:4591-9. [PMID: 16098510 DOI: 10.1016/j.febslet.2005.07.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2005] [Revised: 06/26/2005] [Accepted: 07/04/2005] [Indexed: 10/25/2022]
Abstract
Recent evidence indicates that cell-cycle regulating proteins are involved in apoptotic process in post-mitotic neurons. In this study, we examined cell-cycle regulators for G1/S cell-cycle progression after a transient focal cerebral ischemia induced by middle cerebral artery (MCA) occlusion. In the cerebral frontoparietal cortex, we observed a marked induction of Cyclin D1 (a coactivator of Cdks), and proliferating cell nuclear antigen (PCNA), together with upregulated Cdk kinase activities. This process is accompanied with multiple phosphorylation of retinoblastoma (Rb) protein at Cdk phosphorylation sites in neurons from the ischemic cortex. We further examined DNA synthesis by the incorporation of BrdU, a nucleotide analog that incorporates into newly synthesized DNA. Within 24-h of reperfusion after 60-min occlusion, substantial BrdU-positive neurons were observed in the ischemic cortex. Inhibition of Cdk4 activity during this ischemia/reperfusion is highly neuroprotective. These results suggest that ischemia/reperfusion cerebral damage induces signalings at the G1/S cell-cycle transition, and may constitute a critical step in the neuronal apoptotic pathway in ischemia/reperfusion induced neuronal damage.
Collapse
Affiliation(s)
- Yi Wen
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX 76107-2699, USA
| | | | | | | |
Collapse
|
64
|
Furuta Y, Behringer RR. Recent innovations in tissue-specific gene modifications in the mouse. ACTA ACUST UNITED AC 2005; 75:43-57. [PMID: 15838923 DOI: 10.1002/bdrc.20036] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Annotating the functions of individual genes in in vivo contexts has become the primary task of mouse genetics in the post-genome era. In addition to conventional approaches using transgenic technologies and gene targeting, the recent development of conditional gene modification techniques has opened novel opportunities for elucidating gene function at the level of the whole mouse to individual tissues or cell types. Tissue-specific gene modifications in the mouse have been made possible using site-specific DNA recombinases and conditional alleles. Recent innovations in this basic technology have facilitated new types of experiments, revealing novel insights into mammalian embryology. In this review, we focus on these recent innovations and new technical issues that impact the success of these conditional gene modification approaches.
Collapse
Affiliation(s)
- Yasuhide Furuta
- Department of Biochemistry and Molecular Biology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | |
Collapse
|
65
|
Bremner R, Chen D, Pacal M, Livne-Bar I, Agochiya M. The RB protein family in retinal development and retinoblastoma: new insights from new mouse models. Dev Neurosci 2005; 26:417-34. [PMID: 15855771 DOI: 10.1159/000082284] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2004] [Accepted: 08/30/2004] [Indexed: 11/19/2022] Open
Abstract
The Rb gene was isolated almost 20 years ago, but fundamental questions regarding its role in retinal development and retinoblastoma remain. What is the normal function of RB protein in retinogenesis? What is the cell-of-origin of retinoblastoma? Why do retinoblastoma tumors have recurrent genetic lesions other than Rb inactivation? Why is retinoblastoma not induced by defects in cell cycle regulators other than Rb? Why is the retina so sensitive to Rb loss? Recently developed conditional Rb knockout models provide new insight into some of these issues. The data suggest that RB protein may not control the rate of progenitor division, but is critical for cell cycle exit when dividing retinal progenitors differentiate into postmitotic transition cells. This finding focuses attention on the ectopically dividing transition cell, rather than the progenitor, as the cell-of-origin. Cell-specific analyses in the RB-deficient retina reveal that ectopically dividing photoreceptors, bipolar and ganglion cells die, but amacrine, horizontal and Muller cells survive and stop dividing when they terminally differentiate. Rare amacrine transition cells escape cell cycle exit and generate tumors. These data suggest that post-Rb mutations are required to overcome growth arrest associated with terminal differentiation, rather than apoptosis as previously suggested. To explain why perturbing cell cycle regulators other than RB does not initiate retinoblastoma, we speculate that mutations in other components of the RB pathway perturb cell cycle arrest, but only RB loss triggers genome instability in retinal transition cells, which may be critical to facilitate post-Rb mutations necessary for transformation. Cell-specific differences in the effect of Rb loss on genome stability may contribute to the tremendous sensitivity of retinal transition cells to tumorigenesis. The new mouse models of retinoblastoma will be invaluable for testing these possibilities.
Collapse
Affiliation(s)
- Rod Bremner
- Toronto Western Research Institute, University Health Network, Rm Mc6-424, 399 Bathurst Street, University of Toronto, Toronto, Ontario M5T 2S8, Canada.
| | | | | | | | | |
Collapse
|
66
|
Stighall M, Manetopoulos C, Axelson H, Landberg G. High ID2 protein expression correlates with a favourable prognosis in patients with primary breast cancer and reduces cellular invasiveness of breast cancer cells. Int J Cancer 2005; 115:403-11. [PMID: 15688367 DOI: 10.1002/ijc.20875] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
ID proteins have been implicated in the regulation of cell proliferation and differentiation in various cell types during normal development as well as in the formation of cancer. Our aim was to delineate the expression of ID2 by immunohistochemistry in primary breast cancer in order to detect potential associations with cell cycle regulatory proteins and/or clinicopathologic parameters. We further overexpressed ID2 in a breast cancer cell line to elaborate potential effects on proliferation and invasiveness. We observed large variations in ID2 expression in primary breast cancer, and the protein was localised to both the nucleus and cytoplasm. Interestingly, a high cytoplasmic ID2 protein level correlated with a favourable prognosis. Overexpressing ID2 in the MDA-MB-468 breast cancer cell line generated a marked cytoplasmic localisation of the protein and reduced the invasive capacity of cells. Modest enhancement of cell proliferation was further detected in ID2-overexpressing cells. In conclusion, ID2 protein expression varies substantially within primary breast tumours and high cytoplasmic levels of ID2 might reflect a less aggressive breast tumour phenotype.
Collapse
Affiliation(s)
- Maria Stighall
- Division of Pathology, Department of Laboratory Medicine, Lund University, Malmö University Hospital, Malmö, Sweden
| | | | | | | |
Collapse
|
67
|
Cinti C, Giordano A. The retinoblastoma gene family: its role in cancer onset and progression. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.4.6.765] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
68
|
Jori FP, Melone MAB, Napolitano MA, Cipollaro M, Cascino A, Giordano A, Galderisi U. RB and RB2/p130 genes demonstrate both specific and overlapping functions during the early steps of in vitro neural differentiation of marrow stromal stem cells. Cell Death Differ 2005; 12:65-77. [PMID: 15459751 DOI: 10.1038/sj.cdd.4401499] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Marrow stromal stem cells (MSCs) are stem-like cells that are currently being tested for their potential use in cell therapy for a number of human diseases. MSCs can differentiate into both mesenchymal and nonmesenchymal lineages. In fact, in addition to bone, cartilage and fat, it has been demonstrated that MSCs are capable of differentiating into neurons and astrocytes. RB and RB2/p130 genes are involved in the differentiation of several systems. For this reason, we evaluated the role of RB and RB2/p130 in the differentiation and apoptosis of MSCs under experimental conditions that allow for MSC differentiation toward the neuron-like phenotype. To this end, we ectopically expressed either RB or RB2/p130 and monitored proliferation, differentiation and apoptosis in rat primary MSC cultures induced to differentiate toward the neuron-like phenotype. Both RB and RB2/P130 decreased cell proliferation rate. In pRb-overexpressing cells, the arrest of cell growth was also observed in the presence of the HDAC-inhibitor TSA, suggesting that its antiproliferative activity does not rely upon the HDAC pathway, while the addition of TSA to pRb2/p130-overexpressing cells relieved growth inhibition. TUNEL reactions and studies on the expression of genes belonging to the Bcl-2 family showed that while RB protected differentiating MSCs from apoptosis, RB2/p130 induced an increase of apoptosis compared to controls. The effects of both RB and RB2/p130 on programmed cell death appeared to be HDAC- independent. Molecular analysis of neural differentiation markers and immunocytochemistry revealed that RB2/p130 contributes mainly to the induction of generic neural properties and RB triggers cholinergic differentiation. Moreover, the differentiation potentials of RB2/p130 and RB appear to rely, at least in part, on the activity of HDACs.
Collapse
Affiliation(s)
- F P Jori
- Department of Neurological Sciences, Second University of Naples, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
69
|
Levine EM, Green ES. Cell-intrinsic regulators of proliferation in vertebrate retinal progenitors. Semin Cell Dev Biol 2004; 15:63-74. [PMID: 15036209 DOI: 10.1016/j.semcdb.2003.09.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The proliferative expansion of retinal progenitor cells (RPCs) is a fundamental mechanism of growth during vertebrate retinal development. Over the past couple of years, significant progress has been made in identifying genes expressed in RPCs that are essential for their proliferation, and the molecular mechanisms are beginning to be resolved. In this review, we highlight recent studies that have identified regulatory components of the RPC cell cycle machinery and implicate a set of homeobox genes as key regulators of proliferative expansion in the retina.
Collapse
Affiliation(s)
- Edward M Levine
- Department of Ophthalmology & Visual Sciences, Eccles Institute of Human Genetics, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA.
| | | |
Collapse
|
70
|
Vanderluit JL, Ferguson KL, Nikoletopoulou V, Parker M, Ruzhynsky V, Alexson T, McNamara SM, Park DS, Rudnicki M, Slack RS. p107 regulates neural precursor cells in the mammalian brain. ACTA ACUST UNITED AC 2004; 166:853-63. [PMID: 15353549 PMCID: PMC2172121 DOI: 10.1083/jcb.200403156] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Here we show a novel function for Retinoblastoma family member, p107 in controlling stem cell expansion in the mammalian brain. Adult p107-null mice had elevated numbers of proliferating progenitor cells in their lateral ventricles. In vitro neurosphere assays revealed striking increases in the number of neurosphere forming cells from p107(-/-) brains that exhibited enhanced capacity for self-renewal. An expanded stem cell population in p107-deficient mice was shown in vivo by (a) increased numbers of slowly cycling cells in the lateral ventricles; and (b) accelerated rates of neural precursor repopulation after progenitor ablation. Notch1 was up-regulated in p107(-/-) neurospheres in vitro and brains in vivo. Chromatin immunoprecipitation and p107 overexpression suggest that p107 may modulate the Notch1 pathway. These results demonstrate a novel function for p107 that is distinct from Rb, which is to negatively regulate the number of neural stem cells in the developing and adult brain.
Collapse
|
71
|
Jori FP, Napolitano MA, Melone MAB, Cipollaro M, Cascino A, Giordano A, Galderisi U. Role of RB and RB2/P130 genes in marrow stromal stem cells plasticity. J Cell Physiol 2004; 200:201-212. [PMID: 15174090 DOI: 10.1002/jcp.20026] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Marrow stromal cells (MSCs) are stem-like cells having a striking somatic plasticity. In fact, besides differentiating into mesenchymal lineages (bone, cartilage, and fat), they are capable of differentiating into neurons and astrocytes in vitro and in vivo. The RB and RB2/P130 genes, belonging to the retinoblastoma gene family, play a key role in neurogenesis, and for this reason, we investigated their role in neural commitment and differentiation of MSCs. In MSCs that were either uncommitted or committed toward neural differentiation, we ectopically expressed RB and RB2/P130 genes and analyzed their role in regulating the cell cycle, apoptosis and differentiation. In uncommitted MSCs, the activity of RB and RB2/P130 appeared limited to negatively regulating cell cycle progression, having no role in apoptosis and differentiation (toward either mesenchymal or neural lineages). On the other hand, in MSCs committed toward the neural phenotype, both RB and RB2/P130 reduced cell proliferation rate and affected the apoptotic process. RB protected differentiating cells from programmed cell death. On the contrary, RB2/P130 increased the percentage of cells in apoptosis. All of these activities were accomplished mainly in an HDAC-independent way. The retinoblastoma genes also influenced differentiation in neural committed MSCs. RB2/P130 contributes mainly to the induction of generic neural properties, while RB triggers cholinergic differentiation. These differentiating activities are HDAC-dependent. Our research shows that there is a critical temporal requirement for the RB genes during neuronal differentiation of MSCs: they are not required for cell commitment but play a role in the maturation process. For the above reasons, RB and RB2/P130 may have a role in neural differentiation but not in neural determination.
Collapse
Affiliation(s)
- Francesco P Jori
- Department of Neurological Sciences, Second University of Naples, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
72
|
Liang XQ, Avraham HK, Jiang S, Avraham S. Genetic alterations of the NRP/B gene are associated with human brain tumors. Oncogene 2004; 23:5890-900. [PMID: 15208678 DOI: 10.1038/sj.onc.1207776] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nearly all brain tumors develop following the progressive accumulation of genetic alterations of oncogenes and tumor suppressor genes (such as p53 and retinoblastoma protein). Furthermore, aberrations in the nuclear matrix often contribute to genomic instabilities and the development of cancer. We have previously shown that nuclear-restricted protein/brain (NRP/B), a member of the BTB/Kelch repeat family, is a nuclear matrix protein normally expressed in neurons but not in astrocytes, and that it is an early and specific marker of neurons during the development of the central nervous system. Here, we show aberrant expression of NRP/B in human brain tissues. NRP/B is expressed in the cytoplasm of human brain tumor cells (glioblastoma, GBM) arising from astrocytes. NRP/B mutations (13 mutations in the Kelch domains, two in the intervening sequence (IVS) domain and two in the BTB domain) were detected in brain tumor cell lines (A-172, CCF-STTG1, SK-N-SH and U87-MG) and in primary human malignant GBM tissues (eight samples). More importantly, we found that NRP/B mutants, but not wild-type (wt) NRP/B, increased the activation of ERK and consequently promoted cell proliferation, attenuated caspase activation and suppressed the cellular apoptosis induced by the stressful stimulus cisplatin (10 microM). These events were observed to occur via a p53-mediated pathway. In addition, while wt NRP/B was associated with actin, mutations in the Kelch domains of NRP/B led to its reduced binding affinity to actin. Thus, alterations and gene mutations within the NRP/B gene may contribute to brain tumorigenesis by promoting cell proliferation, suppressing apoptosis and by affecting nuclear cytoskeleton dynamics.
Collapse
Affiliation(s)
- Xing-Qun Liang
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Institutes of Medicine, 4 Blackfan Circle, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
73
|
Liu Y, Encinas M, Comella JX, Aldea M, Gallego C. Basic helix-loop-helix proteins bind to TrkB and p21(Cip1) promoters linking differentiation and cell cycle arrest in neuroblastoma cells. Mol Cell Biol 2004; 24:2662-72. [PMID: 15024057 PMCID: PMC371129 DOI: 10.1128/mcb.24.7.2662-2672.2004] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Differentiation of precursor into specialized cells involves an increasing restriction in proliferative capacity, culminating in cell cycle exit. In this report we used a human neuroblastoma cell line to study the molecular mechanisms that coordinate cell cycle arrest and neuronal differentiation. Exposure to retinoic acid (RA), a differentiation agent in many cell types, causes an upregulation of neurotrophin receptor TrkB and the cyclin kinase inhibitor p21(Cip1) at a transcriptional level. Full transcriptional activation of these two genes requires canonical E-box sequences found in the TrkB and p21(Cip1) promoters. As reported for other E-box-regulated promoters, ectopic expression of E47 and E12 basic helix-loop-helix (bHLH) proteins enhances RA-dependent expression of TrkB and p21(Cip1), whereas the inhibitory HLH Id2 exerts opposite effects. In addition, ectopic expression of E47 and NeuroD, a neuronal bHLH protein, is able to activate TrkB transcription in the absence of RA. More importantly, we show that E47 and NeuroD proteins bind the TrkB and p21(Cip1) promoter sequences in vivo. Since they establish a direct transcriptional link between a cell cycle inhibitor, p21(Cip1), and a neurotrophic receptor, TrkB, bHLH proteins would play an important role in coordinating key events of cell cycle arrest and neuronal differentiation.
Collapse
Affiliation(s)
- Yuhui Liu
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, 25008 Lleida, Catalunya, Spain, Department of Molecular Biology and Pharmacology, Washington University Medical School, St. Louis, Missouri 63110
| | - Mario Encinas
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, 25008 Lleida, Catalunya, Spain, Department of Molecular Biology and Pharmacology, Washington University Medical School, St. Louis, Missouri 63110
| | - Joan X. Comella
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, 25008 Lleida, Catalunya, Spain, Department of Molecular Biology and Pharmacology, Washington University Medical School, St. Louis, Missouri 63110
| | - Martí Aldea
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, 25008 Lleida, Catalunya, Spain, Department of Molecular Biology and Pharmacology, Washington University Medical School, St. Louis, Missouri 63110
| | - Carme Gallego
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, 25008 Lleida, Catalunya, Spain, Department of Molecular Biology and Pharmacology, Washington University Medical School, St. Louis, Missouri 63110
- Corresponding author. Mailing address: Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Montserrat Roig 2, 25008 Lleida, Catalunya, Spain. Phone: (34)973702411. Fax: (34)973702426. E-mail:
| |
Collapse
|
74
|
Lossi L, Gambino G, Mioletti S, Merighi A. In vivo analysis reveals different apoptotic pathways in pre- and postmigratory cerebellar granule cells of rabbit. ACTA ACUST UNITED AC 2004; 60:437-52. [PMID: 15307148 DOI: 10.1002/neu.20032] [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] [Indexed: 11/06/2022]
Abstract
Naturally occurring neuronal death (NOND) has been described in the postnatal cerebellum of several species, mainly affecting the cerebellar granule cells (CGCs) by an apoptotic mechanism. However, little is known about the cellular pathway(s) of CGC apoptosis in vivo. By immunocytochemistry, in situ detection of fragmented DNA, electron microscopy, and Western blotting, we demonstrate here the existence of two different molecular mechanisms of apoptosis in the rabbit postnatal cerebellum. These two mechanisms affect CGCs at different stages of their maturation and migration. In the external granular layer, premigratory CGCs undergo apoptosis upon phosphorylation of checkpoint kinase 1 (Chk1), and hyperphosphorylation of retinoblastoma protein. In postmigratory CGCs within the internal granular layer, caspase 3 and to a lesser extent 7 and 9 are activated, eventually leading to poly-ADP-ribose polymerase-1 (PARP-1) cleavage and programmed cell death. We conclude that NOND of premigratory CGCs is linked to activation of DNA checkpoint and alteration of normal cell cycle, whereas in postmigratory CGCs apoptosis is, more classically, dependent upon caspase 3 activation.
Collapse
Affiliation(s)
- Laura Lossi
- Department of Veterinary Morphophysiology, University of Turin, Via Leonardo da Vinci 44, 10095 Grugliasco (TO) Italy, UE
| | | | | | | |
Collapse
|
75
|
Chiu R, Rey O, Zheng JQ, Twiss JL, Song J, Pang S, Yokoyama KK. Effects of altered expression and localization of cyclophilin A on differentiation of p19 embryonic carcinoma cells. Cell Mol Neurobiol 2003; 23:929-43. [PMID: 14964780 PMCID: PMC11530197 DOI: 10.1023/b:cemn.0000005321.11544.cc] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
1. The retinoblastoma susceptibility gene product, p105Rb (RB), is an important regulator in the control of cell proliferation, differentiation, and apoptosis. Several cellular factors that complex with RB and exert their cellular regulatory functions have been identified, such as the RB:cyclophilin A (CypA) complex. 2. CypA is a cytoplasmic immunophilin and known for its involvement in T-cell differentiation and proliferation. Although CypA has a pivotal role in the immune response, its function in other signaling pathways is largely unknown. 3. In this study, we used a model of neuronal differentiation to demonstrate that the nuclear translocation of CypA, the appearance of hypophosphorylated RB and the enhancement of RB: CypA complex formation correlates with retinoic acid induced neuronal differentiation. 4. Inhibition of CypA expression results in repression of both the hypophosphorylated RB and the neuron-specific differentiation marker, class III beta tubulin. 5. The evidence of enriched CypA and colocalization of RB with CypA in the nucleus of primary adult sensory neurons substantiated the important event of RB-mediated neuronal differentiation of p19 EC cells.
Collapse
Affiliation(s)
- Robert Chiu
- Dental Research Institute, UCLA School of Dentistry, Los Angeles, California 90095-1668, USA.
| | | | | | | | | | | | | |
Collapse
|
76
|
Abstract
The general mechanisms that control the cell cycle in mammalian cells have been studied in depth and several proteins that are involved in the tight regulation of cell cycle progression have been identified. However, the analysis of which molecules participate in cell cycle exit of specific cell lineages is not exhaustive yet. Moreover, the strict relation between cell cycle exit and induction of differentiation has not been fully understood and seems to depend on the cell type. Several in vivo and in vitro studies have been performed in the last few years to address these issues in cells of the nervous system. In this review, we focus our attention on cyclin-cyclin-dependent kinase complexes, cyclin kinase inhibitors, genes of the retinoblastoma family, p53 and N-Myc, and we aim to summarize the latest evidence indicating their involvement in the control of the cell cycle and induction of differentiation in different cell types of the peripheral and central nervous systems. Studies on nervous system tumors and a possible contributory role in tumorigenesis of polyomavirus T antigen are reported to point out the critical contribution of some cell cycle regulators to normal neural and glial development.
Collapse
Affiliation(s)
- Umberto Galderisi
- Department of Experimental Medicine, School of Medicine, Second University of Naples, Naples, Italy
| | | | | |
Collapse
|
77
|
Marino S, Hoogervoorst D, Brandner S, Berns A. Rb and p107 are required for normal cerebellar development and granule cell survival but not for Purkinje cell persistence. Development 2003; 130:3359-68. [PMID: 12810584 DOI: 10.1242/dev.00553] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The involvement of the retinoblastoma gene product (Rb) and its family members (p107 and p130) in cell cycle exit and terminal differentiation of neural precursor cells has been demonstrated in vitro. To investigate the roles of Rb and p107 in growth, differentiation and apoptosis in the developing and mature cerebellum, we selectively inactivated either Rb alone or in combination with p107 in cerebellar precursor cells or in Purkinje cells. In our mouse models, we show that (1) Rb is required for differentiation, cell cycle exit and survival of granule cell precursors; (2) p107 can not fully compensate for the loss of Rb function in granule cells; (3) Rb and p107 are not required for differentiation and survival of Purkinje cells during embryonic and early postnatal development; (4) Rb function in Purkinje cells is cell autonomous; and (5) loss of Rb deficient CNS precursor cells is mediated by p53-independent apoptosis.
Collapse
Affiliation(s)
- Silvia Marino
- Institute of Clinical Pathology, University Hospital, Schmelzbergstrasse 12, 8091 Zürich, Switzerland.
| | | | | | | |
Collapse
|
78
|
Takahashi C, Bronson RT, Socolovsky M, Contreras B, Lee KY, Jacks T, Noda M, Kucherlapati R, Ewen ME. Rb and N-ras function together to control differentiation in the mouse. Mol Cell Biol 2003; 23:5256-68. [PMID: 12861012 PMCID: PMC165732 DOI: 10.1128/mcb.23.15.5256-5268.2003] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The product of the retinoblastoma tumor suppressor gene (Rb) can control cell proliferation and promote differentiation. Murine embryos nullizygous for Rb die midgestation with defects in cell cycle regulation, control of apoptosis, and terminal differentiation of several tissues, including skeletal muscle, nervous system, and lens. Previous cell culture-based experiments have suggested that the retinoblastoma protein (pRb) and Ras operate in a common pathway to control cellular differentiation. Here we have tested the hypothesis that the proto-oncogene N-ras participates in Rb-dependent regulation of differentiation by generating and characterizing murine embryos deficient in both N-ras and Rb. We show that deletion of N-ras rescues a unique subset of the developmental defects associated with nullizygosity of Rb, resulting in a significant extension of life span. Rb(-/-); N-ras(-/-) skeletal muscle has normal fiber density, myotube length and thickness, in contrast to Rb-deficient embryos. Additionally, Rb(-/-); N-ras(-/-) muscle shows a restoration in the expression of the late muscle-specific gene MCK, and this correlates with a significant potentiation of MyoD transcriptional activity in Rb(-/-); N-ras(-/-), compared to Rb(-/-) myoblasts in culture. The improved differentiation of skeletal muscle in Rb(-/-); N-ras(-/-) embryos occurs despite evidence of deregulated proliferation and apoptosis, as seen in Rb-deficient animals. Our findings suggest that the control of differentiation and proliferation by Rb are genetically separable.
Collapse
Affiliation(s)
- Chiaki Takahashi
- Department of Medical Oncology and Medicine, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Abstract
Cell cycle events play a major role in the loss of neurons in advanced Alzheimer's disease (AD). It is currently unknown, however, whether the same is true for the neuronal losses in early disease stages. To explore this issue we analyzed brain autopsy material from individuals clinically categorized with mild cognitive impairment (MCI), many if not most of whom will progress to AD. Immunocytochemistry for three cell cycle-related proteins, proliferating cell nuclear antigen, cyclin D, and cyclin B, was performed on sections from hippocampus, basal nucleus of Meynert, and entorhinal cortex. The results obtained from MCI cases were compared with material from individuals diagnosed with AD and those without cognitive impairment. In both hippocampus and basal nucleus, there was a significant percentage of cell cycle immunopositive neurons in the MCI cases. These percentages were similar to those found in the AD cases but significantly higher than non-cognitively impaired controls. In entorhinal cortex, the density of cell cycle-positive neurons was greater in MCI than in AD. However, we observed large variations in the percentages of immunopositive neurons from individual to individual. These findings lend support to the hypothesis that both the mechanism of cell loss (a cell cycle-induced death) and the rate of cell loss (a slow atrophy over several months) are identical at all stages of the AD disease process. The implication of the findings for human clinical trials is discussed.
Collapse
|
80
|
Lossi L, Merighi A. In vivo cellular and molecular mechanisms of neuronal apoptosis in the mammalian CNS. Prog Neurobiol 2003; 69:287-312. [PMID: 12787572 DOI: 10.1016/s0301-0082(03)00051-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Apoptosis has been recognized to be an essential process during neural development. It is generally assumed that about half of the neurons produced during neurogenesis die before completion of the central nervous system (CNS) maturation, and this process affects nearly all classes of neurons. In this review, we discuss the experimental data in vivo on naturally occurring neuronal death in normal, transgenic and mutant animals, with special attention to the cerebellum as a study model. The emerging picture is that of a dual wave of apoptotic cell death affecting central neurons at different stages of their life. The first wave consists of an early neuronal death of proliferating precursors and young postmitotic neuroblasts, and appears to be closely linked to cell cycle regulation. The second wave affects postmitotic neurons at later stages, and is much better understood in functional terms, mainly on the basis of the neurotrophic concept in its broader definition. The molecular machinery of late apoptotic death of postmitotic neurons more commonly follows the mitochondrial pathway of intracellular signal transduction, but the death receptor pathway may also be involved.Undoubtedly, analysis of naturally occurring neuronal death (NOND) in vivo will offer a basis for parallel and future studies aiming to elucidate the mechanisms of pathologic neuronal loss occurring as the result of conditions such as neurodegenerative disorders, trauma or ischemia.
Collapse
Affiliation(s)
- L Lossi
- Department of Veterinary Morphophysiology, University of Torino, Via Leonardo da Vinci 44, I-10095 (TO), Grugliasco, Italy.
| | | |
Collapse
|
81
|
Leezer JL, Hackmiller RC, Greene RM, Pisano MM. Expression of the retinoblastoma family of tumor suppressors during murine embryonic orofacial development. Orthod Craniofac Res 2003; 6:32-47. [PMID: 12627794 DOI: 10.1046/j.1439-0280.2003.2c035.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To elucidate the role of the retinoblastoma (Rb) family of tumor suppressors and growth regulators in transforming growth factor beta (TGFbeta)-mediated embryonic palatal growth and morphogenesis. DESIGN The spatio-temporal expression patterns of the RB1, RB2/p130, and p107 tumor suppressor genes, their gene products (pRb, p130 and p107) and phosphoforms were examined in the developing murine secondary palate utilizing reverse transcriptase polymerase chain reaction (RT-PCR) and immunoblot/immunolocalization analyses with phospho-specific antibodies. RESULTS The RB1, RB2/p130, and p107 tumor suppressor genes and their gene products (pRb, p130, and p107) were differentially expressed in embryonic palatal tissue during the critical period of secondary palate development [gestational days (GD) 12-14]. Both hyper- (115 kDa) and hypo-phosphorylated (110 kDa) forms of pRb were expressed, with a notable transient decrease in expression on GD 13. Functional (hypo-phosphorylated) forms of pRB predominated during the critical period of palatogenesis. As opposed to pRb expression, p130 expression was transiently elevated on GD 13 in the embryonic palate, and functional (hypo-phosphorylated) forms were expressed at exceedingly low levels. p107 levels gradually declined over the course of palatogenesis. This diminution in expression, however, was accompanied by a transition to more functional (hypo-phosphorylated) forms of the p107 protein. When compared with Rb expression patterns in the whole embryo/fetus, each of the three Rb proteins exhibited unique and specific temporal patterns of expression in the developing palate. RT-PCR analyses of RB1, RB2/p130, and p107 mRNA expression in embryonic palatal tissue revealed patterns of expression which paralleled steady-state protein levels of pRb, p130, and p107 in the developing tissue. Immunolocalization of the Rb proteins demonstrated ubiquitous expression of pRb, p130, and p107 in embryonic palate mesenchyme and epithelium during GD 12-14 of development with intense nuclear staining of the Rbs in palate epithelial cells on days 12 and 13 of gestation. CONCLUSION Changing patterns of pRb, p130, and p107 protein expression and phosphorylation were evident in the embryonic secondary palate during the course of palatal ontogenesis. As the function of all three Rb proteins is regulated primarily through their phosphorylation, this suggests significant variation in Rb protein functionality during the course of palate development. Additional evidence from our laboratory (manuscript in preparation) indicates that the TGFbetas, key regulators of palatal growth and morphogenesis, are able to differentially modulate phosphorylation of the Rb proteins in this developing tissue, and hence the TGFbetas may regulate the function(s) of the Rb proteins during palatal ontogenesis. Although functionality of the Rb proteins is known to be regulated primarily through post-translational modification (i.e. phopsphorylation), comparison of RB1, RB2/p130, and p107 steady-state mRNA levels with pRb, p130, and p107 steady-state protein levels in the developing palate levels suggests a significant degree of regulation at the transcriptional level. Differential patterns of Rb expression and phosphorylation in the developing palate suggest that pRb, p107, and p130 may each play unique roles in various aspects of growth, morphogenesis and cellular differentiation during palatal ontogenesis. Studies directed at elucidating the precise cellular role(s) of pRb, p107, and p130 during palate development, and at TGFbeta regulation of Rb expression/phosphorylation are ongoing in our laboratory.
Collapse
Affiliation(s)
- J L Leezer
- Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY 40292, USA
| | | | | | | |
Collapse
|
82
|
MacPherson D, Sage J, Crowley D, Trumpp A, Bronson RT, Jacks T. Conditional mutation of Rb causes cell cycle defects without apoptosis in the central nervous system. Mol Cell Biol 2003; 23:1044-53. [PMID: 12529408 PMCID: PMC140709 DOI: 10.1128/mcb.23.3.1044-1053.2003] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Targeted disruption of the retinoblastoma gene in mice leads to embryonic lethality in midgestation accompanied by defective erythropoiesis. Rb(-/-) embryos also exhibit inappropriate cell cycle activity and apoptosis in the central nervous system (CNS), peripheral nervous system (PNS), and ocular lens. Loss of p53 can prevent the apoptosis in the CNS and lens; however, the specific signals leading to p53 activation have not been determined. Here we test the hypothesis that hypoxia caused by defective erythropoiesis in Rb-null embryos contributes to p53-dependent apoptosis. We show evidence of hypoxia in CNS tissue from Rb(-/-) embryos. The Cre-loxP system was then used to generate embryos in which Rb was deleted in the CNS, PNS and lens, in the presence of normal erythropoiesis. In contrast to the massive CNS apoptosis in Rb-null embryos at embryonic day 13.5 (E13.5), conditional mutants did not have elevated apoptosis in this tissue. There was still significant apoptosis in the PNS and lens, however. Rb(-/-) cells in the CNS, PNS, and lens underwent inappropriate S-phase entry in the conditional mutants at E13.5. By E18.5, conditional mutants had increased brain size and weight as well as defects in skeletal muscle development. These data support a model in which hypoxia is a necessary cofactor in the death of CNS neurons in the developing Rb mutant embryo.
Collapse
Affiliation(s)
- D MacPherson
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | |
Collapse
|
83
|
Abstract
Mechanisms coupling cell cycle and cell fate operate at different steps during neural development. Intrinsic factors control the cell proliferation of distinct brain regions and changes of cell fate competence, whereas components of the cell cycle machinery could play a major role in setting the appropriate timing of the generation of different cell types.
Collapse
Affiliation(s)
- Federico Cremisi
- Scuola Normale Superiore/Dipartimento di Fisiologia e Biochimica, Sezione di Biologia Cellulare e dello Sviluppo, Università di Pisa, Via Carducci 13, Ghezzano, 56010, Pisa, Italy.
| | | | | |
Collapse
|
84
|
Abstract
Recent studies have shown that RB can inhibit apoptosis, independently of its ability to block cell proliferation. This poses the question of how cells choose to grow or to die when RB becomes inactivated. RB is phosphorylated following mitogenic stimulation, but it is degraded in response to death stimuli. Most sporadic cancers also inactivate RB by phosphorylation, rather than losing RB entirely--possibly to exploit the survival advantage conferred by RB under stress. Drawing from the different mechanisms of RB inactivation, we propose two models for ways in which cells use RB to make the choice of life versus death.
Collapse
Affiliation(s)
- B Nelson Chau
- Division of Biological Sciences and the Cancer Center, University of California, San Diego, La Jolla, California 92093-0322, USA
| | | |
Collapse
|
85
|
Jordan-Sciutto KL, Dorsey R, Chalovich EM, Hammond RR, Achim CL. Expression patterns of retinoblastoma protein in Parkinson disease. J Neuropathol Exp Neurol 2003; 62:68-74. [PMID: 12528819 DOI: 10.1093/jnen/62.1.68] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cellular mechanisms implicated in Parkinson disease (PD) include oxidative stress, inflammatory response, excess dopamine, DNA damage, and loss of trophic support. These stimuli have been observed to induce changes in cell cycle proteins in several cell types. One of the key regulators of cell cycle progression is the retinoblastoma protein (pRb); therefore, we assessed the staining for pRb and its inactive hyperphosphorylated isoform, ppRb, in autopsy tissue from patients with PD. In PD we found abundant pRb staining in neuronal cytoplasm of the substantia nigra, mid-frontal cortex, and hippocampus by immunohistochemistry. In controls, pRb weakly stained nucleoli of neurons in the substantia nigra and exhibited no detectable staining in mid-frontal cortex and hippocampus. Staining for ppRb resulted in a shift from weak cytoplasmic staining in neurons from control cases to strong nuclear staining in PD cases, especially within the substantia nigra, mid-frontal cortex, and hippocampus. In the substantia nigra, ppRb also co-localized to Lewy bodies, which are a pathologic feature of PD. Lewy bodies are also found in diffuse Lewy body disease (DLBD) that do not consistently exhibit changes in pRb or ppRb. These results indicate that there are changes in pRb and its inactive phospho-isoform in neurons responding to neurodegenerative stimuli associated with PD.
Collapse
|
86
|
Decary S, Decesse JT, Ogryzko V, Reed JC, Naguibneva I, Harel-Bellan A, Cremisi CE. The retinoblastoma protein binds the promoter of the survival gene bcl-2 and regulates its transcription in epithelial cells through transcription factor AP-2. Mol Cell Biol 2002; 22:7877-88. [PMID: 12391156 PMCID: PMC134726 DOI: 10.1128/mcb.22.22.7877-7888.2002] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The retinoblastoma (RB) gene product has been shown to restrict cell proliferation, promote cell differentiation, and inhibit apoptosis. Loss of RB function can induce both p53-dependent apoptosis and p53-independent apoptosis; little is known about the mechanisms of RB-regulated p53-independent apoptosis. Here we show that RB specifically activates transcription of the survival gene bcl-2 in epithelial cells but not in NIH 3T3 mesenchymal cells. This transcriptional activity is mediated by the transcription factor AP-2. By monitoring protein-DNA interactions in living cells using formaldehyde cross-linking and chromatin immunoprecipitation, we show that endogenous RB and AP-2 both bind to the same bcl-2 promoter sequence. In addition, we demonstrate that RB and AP-2 also bind to the E-cadherin gene promoter in vivo, consistent with regulation of this promoter by both AP-2 and RB in epithelial cells. This study provides evidence that RB activates bcl-2 and E-cadherin by binding directly to the respective promoter sequences and not indirectly by repressing an inhibitor. This recruitment is mediated by a transcription factor, in this case AP-2. For the first time, our results suggest a direct molecular mechanism by which RB might inhibit apoptosis independently of p53. The results are discussed in a context where RB and Bcl-2 contribute under nonpathological conditions to the maintenance of cell viability in association with a differentiated phenotype, contributing to the tumor suppressor function of RB and playing important roles in normal development.
Collapse
Affiliation(s)
- Stephanie Decary
- Laboratoire Oncogenese, Differenciation et Transduction du Signal, CNRS UPR 9079, Institut Andre Lwoff, 94801 Villejuif, France. The Burnham Institute, La JollaCalifornia 92037, USA
| | | | | | | | | | | | | |
Collapse
|
87
|
Klein JA, Longo-Guess CM, Rossmann MP, Seburn KL, Hurd RE, Frankel WN, Bronson RT, Ackerman SL. The harlequin mouse mutation downregulates apoptosis-inducing factor. Nature 2002; 419:367-74. [PMID: 12353028 DOI: 10.1038/nature01034] [Citation(s) in RCA: 450] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2002] [Accepted: 07/26/2002] [Indexed: 11/08/2022]
Abstract
Harlequin (Hq) mutant mice have progressive degeneration of terminally differentiated cerebellar and retinal neurons. We have identified the Hq mutation as a proviral insertion in the apoptosis-inducing factor (Aif) gene, causing about an 80% reduction in AIF expression. Mutant cerebellar granule cells are susceptible to exogenous and endogenous peroxide-mediated apoptosis, but can be rescued by AIF expression. Overexpression of AIF in wild-type granule cells further decreases peroxide-mediated cell death, suggesting that AIF serves as a free radical scavenger. In agreement, dying neurons in aged Hq mutant mice show oxidative stress. In addition, neurons damaged by oxidative stress in both the cerebellum and retina of Hq mutant mice re-enter the cell cycle before undergoing apoptosis. Our results provide a genetic model of oxidative stress-mediated neurodegeneration and demonstrate a direct connection between cell cycle re-entry and oxidative stress in the ageing central nervous system.
Collapse
Affiliation(s)
- Jeffrey A Klein
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Delhon GA, González MJ, Murcia PR. Susceptibility of sensory neurons to apoptosis following infection by bovine herpesvirus type 1. J Gen Virol 2002; 83:2257-2267. [PMID: 12185281 DOI: 10.1099/0022-1317-83-9-2257] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Like other members of the alpha subfamily of herpesviruses, bovine herpesvirus type 1 (BHV-1) establishes latent infections in sensory neurons. BHV-1 induces apoptosis in lymphoid cells in vivo and in epithelial cell lines, but the ability of BHV-1 to induce apoptosis in sensory neurons remains unknown. In this report, the susceptibility of rabbit ganglionic neurons to infection by BHV-1 was examined in vitro and in vivo. Following infection of cultured neurons with BHV-1, hallmarks of apoptosis such as chromatin condensation, DNA fragmentation and membrane blebbing were detected. The appearance of these changes was preceded by active viral DNA replication as determined by in situ hybridization. When viral DNA replication was blocked by treatment of cultures with an inhibitor of eukaryotic DNA polymerases, apoptosis but not virus attachment to neurons or bICP0 gene expression was completely prevented. Taken together, these results demonstrate that sensory neurons are not intrinsically resistant to BHV-1-induced apoptosis and that viral DNA replication plays a role in triggering the apoptotic programme. Infection of rabbits with BHV-1 resulted in pathological changes in the trigeminal ganglia (TG) which included mononuclear cell infiltration and neuronophagia. Morphological evidence of apoptosis was not detected in neurons, even in cells with advanced cytophatology. Furthermore, whereas DNA fragmentation was common in infiltrating cells, it was very rare and sporadic in neurons. Therefore, mechanisms in the TG should exist to prevent neuronal apoptosis upon BHV-1 infection.
Collapse
Affiliation(s)
- Gustavo A Delhon
- Department of Virology, School of Veterinary Science, University of Buenos Aires, Chorroarin 280, 1427 Buenos Aires, Argentina1
| | - Marcelo J González
- Department of Virology, School of Veterinary Science, University of Buenos Aires, Chorroarin 280, 1427 Buenos Aires, Argentina1
| | - Pablo R Murcia
- Department of Virology, School of Veterinary Science, University of Buenos Aires, Chorroarin 280, 1427 Buenos Aires, Argentina1
| |
Collapse
|
89
|
Yuan SSF, Yeh YT, Lee EYHP. Pax-2 interacts with RB and reverses its repression on the promoter of Rig-1, a Robo member. Biochem Biophys Res Commun 2002; 296:1019-25. [PMID: 12200151 DOI: 10.1016/s0006-291x(02)02032-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
RB plays dual roles in the regulation of cell proliferation and differentiation. The nervous tissue-specific gene Rig-1, a member of the roundabout (Robo) guidance receptor family, was identified as an RB-regulated gene in the mouse embryo. Herein, we report that a 2.3kb genomic DNA fragment, which contains the first 129 bases of the 5'-untranslated region and 2.2kb of the 5'-flanking region of Rig-1, has a cell type-specific promoter activity. Rig-1 promoter activity is downregulated by RB and upregulated by Pax-2. Furthermore, Rig-1 and Pax-2 mRNAs are coexpressed in the hindbrain and spinal cord of the E11.5 mouse embryo, suggesting that Pax-2 may regulate Rig-1 expression during the embryonic stage. Pax-2 interacts with RB and reverses its transcriptional suppression on the Rig-1 promoter. In summary, the ubiquitous tumor suppressor RB and the neuron-enriched transcription factor Pax-2 may play a role in the regulation of Rig-1 expression during embryogenesis.
Collapse
Affiliation(s)
- Shyng-Shiou F Yuan
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, ROC.
| | | | | |
Collapse
|
90
|
Takazaki R, Nishimura I, Yoshikawa K. Necdin is required for terminal differentiation and survival of primary dorsal root ganglion neurons. Exp Cell Res 2002; 277:220-32. [PMID: 12083804 DOI: 10.1006/excr.2002.5558] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Necdin is expressed predominantly in postmitotic neurons and serves as a growth suppressor that is functionally similar to the retinoblastoma tumor suppressor protein. Using primary cultures of dorsal root ganglion (DRG) of mouse embryos, we investigated the involvement of necdin in the terminal differentiation of neurons. DRG cells were prepared from mouse embryos at 12.5 days of gestation and cultured in the presence of nerve growth factor (NGF). Immunocytochemistry revealed that necdin accumulated in the nucleus of differentiated neurons that showed neurite extension and expressed the neuronal markers microtubule-associated protein 2 and synaptophysin. Suppression of necdin expression in DRG cultures treated with antisense oligonucleotides led to a marked reduction in the number of terminally differentiated neurons. The antisense oligonucleotide-treated cells did not attempt to reenter the cell cycle, but underwent death with characteristics of apoptosis such as caspase-3 activation, nuclear condensation, and chromosomal DNA fragmentation. Furthermore, a caspase-3 inhibitor rescued antisense oligonucleotide-treated cells from apoptosis and significantly increased the population of terminally differentiated neurons. These results suggest that necdin mediates the terminal differentiation and survival of NGF-dependent DRG neurons and that necdin-deficient nascent neurons are destined to caspase-3-dependent apoptosis.
Collapse
Affiliation(s)
- Risa Takazaki
- Division of Regulation of Macromolecular Functions, Institute for Protein Research, Osaka University, Yamadaoka 3-2, Suita, Japan
| | | | | |
Collapse
|
91
|
Ferguson KL, Vanderluit JL, Hébert JM, McIntosh WC, Tibbo E, MacLaurin JG, Park DS, Wallace VA, Vooijs M, McConnell SK, Slack RS. Telencephalon-specific Rb knockouts reveal enhanced neurogenesis, survival and abnormal cortical development. EMBO J 2002; 21:3337-46. [PMID: 12093735 PMCID: PMC126087 DOI: 10.1093/emboj/cdf338] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Correct cell cycle regulation and terminal mitosis are critical for nervous system development. The retinoblastoma (Rb) protein is a key regulator of these processes, as Rb-/- embryos die by E15.5, exhibiting gross hematopoietic and neurological defects. The extensive apoptosis in Rb-/- embryos has been attributed to aberrant S phase entry resulting in conflicting growth control signals in differentiating cells. To assess the role of Rb in cortical development in the absence of other embryonic defects, we examined mice with telencephalon-specific Rb deletions. Animals carrying a floxed Rb allele were interbred with mice in which cre was knocked into the Foxg1 locus. Unlike germline knockouts, mice specifically deleted for Rb in the developing telencephalon survived until birth. In these mutants, Rb-/- progenitor cells divided ectopically, but were able to survive and differentiate. Mutant brains exhibited enhanced cellularity due to increased proliferation of neuroblasts. These studies demonstrate that: (i) cell cycle deregulation during differentiation does not necessitate apoptosis; (ii) Rb-deficient mutants exhibit enhanced neuroblast proliferation; and (iii) terminal mitosis may not be required to initiate differentiation.
Collapse
Affiliation(s)
- Kerry L Ferguson
- Ottawa Health Research Institute, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Oakenfull EA, Riou-Khamlichi C, Murray JAH. Plant D-type cyclins and the control of G1 progression. Philos Trans R Soc Lond B Biol Sci 2002; 357:749-60. [PMID: 12079670 PMCID: PMC1692988 DOI: 10.1098/rstb.2002.1085] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The basic pattern of controls that operate during the G1 phase of the plant cell cycle shows much closer similarity to animals than to the yeasts and other fungi. The activity of D-type cyclin (CycD) kinases is induced in response to stimulatory signals, and these phosphorylate the plant homologue of the retinoblastoma tumour susceptibility (Rb) protein. It is likely that Rb phosphorylation results in the activation of genes under the control of E2F transcription factors, including those required for S phase entry. As the initial triggers of the cascade, attention has focused on the CycDs, and a family of 10 genes is present in Arabidopsis, divided into three major and three minor groups. Analysis to date suggests that these groups are functionally distinct.
Collapse
Affiliation(s)
- E Ann Oakenfull
- Institute of Biotechnology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QT, UK
| | | | | |
Collapse
|
93
|
Smalley KSM, Eisen TG. Differentiation of human melanoma cells through p38 MAP kinase is associated with decreased retinoblastoma protein phosphorylation and cell cycle arrest. Melanoma Res 2002; 12:187-92. [PMID: 12140374 DOI: 10.1097/00008390-200206000-00001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The retinoblastoma protein (pRB), the product of the retinoblastoma gene, is a key regulator of the cell cycle, affecting apoptosis, proliferation and differentiation. Dysregulation of pRB is implicated in the pathogenesis of many cancers, including malignant melanoma. Recently we demonstrated that alpha-melanocyte-stimulating hormone (alpha-MSH)-induced activation of p38 mitogen-activated protein (MAP) kinase leads to differentiation of B16 murine melanoma cells. The current study assesses the ability of alpha-MSH to activate p38 MAP kinase in COLO 853 human melanoma cells and determines whether this is linked to modulation of pRB activity. Treatment of COLO 853 cells with alpha-MSH induced time- and concentration-dependent increases in the phosphorylation of p38 MAP kinase, which corresponded with its ability to induce melanogenesis and inhibit cell growth. SB 203580, a selective inhibitor of p38 MAP kinase, blocked both the alpha-MSH-induced melanogenic response and inhibition of cell growth. Cell cycle analysis using flow cytometry revealed that treatment of COLO 853 cells with alpha-MSH for 72 h led to an increase in the proportion of cells in the G(1) phase and a marked reduction in the amount of phosphorylated pRB. Both of these effects were reversed by pre-treatment of cells with SB 203580. In summary, we have demonstrated for the first time that the alpha-MSH-induced differentiation of COLO 853 human melanoma cells proceeds via a p38 MAP kinase-mediated pathway and is associated with decreased pRB phosphorylation and accumulation of cells in the G(1) phase.
Collapse
Affiliation(s)
- K S M Smalley
- Section of Medicine, Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK.
| | | |
Collapse
|
94
|
Fu WY, Wang JH, Ip NY. Expression of Cdk5 and its activators in NT2 cells during neuronal differentiation. J Neurochem 2002; 81:646-54. [PMID: 12065673 DOI: 10.1046/j.1471-4159.2002.00856.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have recently developed a rapid protocol involving NT2 cell aggregation and treatment with retinoic acid (RA) to produce terminally differentiated CNS neurons. As a first step to explore the functional roles of cell-cycle regulatory proteins in the process of neuronal differentiation, the expression profiles of cyclin-dependent kinases (Cdks) and their regulators were examined in NT2 cells following treatment with RA. One of the Cdks, Cdk5, has been demonstrated to affect the process of neuronal differentiation and suggested to play an important role in development of the nervous system. We found that the expression of Cdk5 was gradually increased, while its activators (p35 and p39) as well as Cdk5 kinase activity were induced in NT2 cells during the process of neuronal differentiation. Moreover, both p35 and p39 were localized along the axons and varicosity-like structures of differentiated NT2 neurons. Taken together, our results demonstrated that NT2 cells provide a good in vitro model system to examine signaling pathways involved in the regulation of Cdk5 activators and to elucidate the functional roles of Cdk5 in neuronal differentiation.
Collapse
Affiliation(s)
- Wing-Yu Fu
- Department of Biochemistry, Molecular Neuroscience Center and Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | | | | |
Collapse
|
95
|
Abstract
In most postmitotic neurons, expression or activation of proteins that stimulate cell cycle progression or DNA replication results in apoptosis. One potential exception to this generalization is neuroblastoma (NB), a tumor derived from the sympathoadrenal lineage. NBs often express high levels of N-myc, a proto-oncogene that can potently activate key components of the cell cycle machinery. Here, we show that in postmitotic sympathetic neurons, N-myc can induce S-phase entry while protecting neurons from death caused by aberrant cell cycle reentry. Specifically, these experiments demonstrate that expression of N-myc at levels similar to those in NBs caused sympathetic neurons to reenter S-phase, as monitored by 5-bromo-2-deoxyuridine incorporation and expression of cell cycle regulatory proteins, and rescued them from apoptosis induced by withdrawal of their obligate survival factor, nerve growth factor. The N-myc-induced cell cycle entry, but not enhanced survival, was inhibited by coexpression of a constitutively hypophosphorylated form of the retinoblastoma tumor suppressor protein, suggesting that these two effects of N-myc are mediated by separate pathways. In contrast, N-myc did not cause S-phase entry in postmitotic cortical neurons. Thus, N-myc both selectively causes sympathetic neurons to reenter the cell cycle and protects them from apoptosis, potentially contributing to their transformation to NBs.
Collapse
|
96
|
Whyatt D, Grosveld F. Cell-nonautonomous function of the retinoblastoma tumour suppressor protein: new interpretations of old phenotypes. EMBO Rep 2002; 3:130-5. [PMID: 11839697 PMCID: PMC1083974 DOI: 10.1093/embo-reports/kvf033] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Loss of the retinoblastoma protein (pRb) induces a cell-nonautonomous defect in both erythroid and neuronal differentiation. It has previously been thought that this reflects a requirement for pRb function in cells that normally support erythropoiesis and neurogenesis, rather than in the erythrocytes or neurons themselves. However, recent studies have challenged this interpretation, and it appears that erythrocytes and neurons themselves have the intrinsic requirement for pRb function. This requirement can be bypassed by signals supplied by wild-type erythroid or neuronal cells. The existence of such a signalling mechanism has implications not only in understanding pRb function but also in the interpretation of other cell-nonautonomous phenotypes.
Collapse
Affiliation(s)
- David Whyatt
- Erasmus University, Department of Cell Biology and Genetics, Medical Genetics Centre, PO Box 1738, 3000 DR Rotterdam, The Netherlands
| | | |
Collapse
|
97
|
Wang F, Corbett D, Osuga H, Osuga S, Ikeda JE, Slack RS, Hogan MJ, Hakim AM, Park DS. Inhibition of cyclin-dependent kinases improves CA1 neuronal survival and behavioral performance after global ischemia in the rat. J Cereb Blood Flow Metab 2002; 22:171-82. [PMID: 11823715 DOI: 10.1097/00004647-200202000-00005] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Increasing evidence suggests that cyclin-dependent kinases participate in neuronal death induced by multiple stresses in vitro. However, their role in cell death paradigms in vivo is not well characterized. Accordingly, the authors examined whether cyclin-dependent kinase inhibition resulted in functionally relevant and sustained neuroprotection in a model of global ischemia. Intracerebroventricular administration of the cyclin-dependent kinase inhibitor flavopiridol, immediately or at 4 hours postreperfusion after a global insult, reduced injury in the CA1 of the hippocampus when examined 7 days after reperfusion. No significant protection was observed when flavopiridol was administered 8 hours after reperfusion. The tumor-suppressor retinoblastoma protein, a substrate of cyclin-dependent kinase, was phosphorylated on a cyclin-dependent kinase consensus site after the global insult; this phosphorylation was inhibited by flavopiridol administration. Importantly, flavopiridol had no effect on core body temperature, suggesting that the mechanism of neuroprotection was through cyclin-dependent kinase inhibition but not through hypothermia. Furthermore, inhibition of cyclin-dependent kinases improved spatial learning behavior as assessed by the Morris water maze 7 to 9 days after reperfusion. However, the histologic protection observed at day 7 was absent 28 days after reperfusion. These results indicate that cyclin-dependent kinase inhibition provides an extended period of morphologic and functional neuroprotection that may allow time for other neuroprotective modalities to be introduced.
Collapse
Affiliation(s)
- Fuhu Wang
- Neuroscience Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Tsai KY, MacPherson D, Rubinson DA, Crowley D, Jacks T. ARF is not required for apoptosis in Rb mutant mouse embryos. Curr Biol 2002; 12:159-63. [PMID: 11818069 DOI: 10.1016/s0960-9822(01)00659-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The retinoblastoma (RB) tumor suppressor gene occupies central roles in cell cycle control and tumor suppression. Homozygous mutant (Rb(-/-)) embryos die at E13.5-E15.5, exhibiting extensive apoptosis and inappropriate S phase entry in the central and peripheral nervous systems, liver, and ocular lens. Mice simultaneously mutant for Rb and other genes can be generated to assess the requirement for these genes in cell cycle control and apoptosis. Using such analysis, E2f-1, E2f-3, p53, and Id2 have been identified as important regulators of cell cycle control and apoptosis in Rb(-/-) embryos. Because unrestrained E2F activity in the absence of Rb function contributes to p53-dependent apoptosis in many systems, we wished to identify genes linking deregulated E2F activity to p53 activation and subsequent apoptosis. As a transcriptional target of E2F-1, a regulator of p53, and an important mediator of apoptosis, ARF was a strong candidate for such a role, especially since it can be upregulated in the absence of Rb. From the analysis of Rb/ARF compound mutants we demonstrate that ARF is not an obligatory link between Rb inactivation and p53-dependent apoptosis.
Collapse
Affiliation(s)
- Kenneth Y Tsai
- MIT Department of Biology and Center for Cancer Research, Cambridge, MA 02139, USA
| | | | | | | | | |
Collapse
|
99
|
Jiang Z, Zacksenhaus E. Activation of retinoblastoma protein in mammary gland leads to ductal growth suppression, precocious differentiation, and adenocarcinoma. J Cell Biol 2002; 156:185-98. [PMID: 11777937 PMCID: PMC2173568 DOI: 10.1083/jcb.200106084] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The retinoblastoma (Rb) tumor suppressor controls cellular proliferation, survival, and differentiation and is functionally inactivated by mutations or hyperphosphorylation in most human cancers. Although activation of endogenous Rb is thought to provide an effective approach to suppress cell proliferation, long-term inhibition of apoptosis by active Rb may have detrimental consequences in vivo. To directly test these paradigms, we targeted phosphorylation-resistant constitutively active Rb alleles, Rb Delta Ks, to the mouse mammary gland. Pubescent transgenic females displayed reduced ductal elongation and cell proliferation at the endbuds. Post-puberty transgenic mice exhibited precocious cellular differentiation and beta-casein expression and extended survival of the mammary epithelium with a moderate but specific effect on the expression of E2F1, IGF1R alpha, and phospho-protein kinase B/AKT. Remarkably, approximately 30% Rb Delta K transgenic females developed focal hyperplastic nodules, and approximately 7% exhibited full-blown mammary adenocarcinomas within 15 mo. Expression of the Rb Delta K transgene in these mammary tumors was reduced greatly. Our results suggest that transient activation of Rb induces cancer by extending cell survival and that the dual effects of Rb on cell proliferation and apoptosis impose an inherent caveat to the use of the Rb pathway for long-term cancer therapy.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adenocarcinoma/therapy
- Alleles
- Animals
- Breast/cytology
- Breast/metabolism
- Breast/pathology
- Cell Differentiation
- Cell Division
- Cell Survival
- Female
- Gene Expression Regulation, Developmental
- Hyperplasia/genetics
- Hyperplasia/metabolism
- Hyperplasia/pathology
- In Situ Hybridization
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mammary Neoplasms, Animal/therapy
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Transgenic
- Milk Proteins/genetics
- Models, Biological
- Mutation
- Parity
- Phosphorylation
- Pregnancy
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Retinoblastoma Protein/genetics
- Retinoblastoma Protein/metabolism
- Transgenes/genetics
Collapse
Affiliation(s)
- Zhe Jiang
- Department of Medicine, University of Toronto, Ontario, Canada M5G 2M1
| | | |
Collapse
|
100
|
Abstract
Cell division is intimately intertwined with plant development, and the mechanisms that link the control of cell proliferation and differentiation with the processes of organogenesis, morphogenesis, and growth are starting to be understood. A recent Juan March meeting explored this interface, and revealed a rich seam of exciting work that is leading toward an integrated view of the role of cell proliferation in the unfolding of developmental programs.
Collapse
Affiliation(s)
- James A H Murray
- Institute of Biotechnology, University of Cambridge, Tennis Court Road, CB2 1QT, Cambridge, United Kingdom.
| |
Collapse
|