U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Madame Curie Bioscience Database [Internet]. Austin (TX): Landes Bioscience; 2000-2013.

Cover of Madame Curie Bioscience Database

Madame Curie Bioscience Database [Internet].

Show details

The Role of Mammalian Coronins in Development and Disease

, , and .

Author Information and Affiliations
Read this chapter in the Madame Curie Bioscience Database here.

Coronins have maintained a high degree of conservation over the roughly 800 million years of eukaryotic evolution.1,2 From its origins as a single gene in simpler eukaryotes, the mammalian Coronin gene family has expanded to include at least six members (see Chapter 4). Increasing evidence indicates that Coronins play critical roles as regulators of actin dependent processes such as cell motility and vesicle trafficking3,4 (see Chapters 6-9). Considering the importance of these processes, it is not surprising that recent findings have implicated the involvement of Coronins in multiple diseases. This review primarily focuses on Coronin 1C (HGNC symbol: CORO1C, also known as Coronin 3) which is a transcriptionally dynamic gene that is up-regulated in multiple types of clinically aggressive cancer. In addition to reviewing the molecular signals and events that lead to Coronin 1C transcription, we summarize the results of several studies describing the possible functional roles of Coronin 1C in development as well as disease progression. Here, the main focus is on brain development and on the progression of melanoma and glioma. Finally, we will also review the role of other mammalian Coronin genes in clinically relevant processes such as neural regeneration and pathogenic bacterial infections (see Chapter 10).

Coronin 1C: A Transcriptionally Dynamic Coronin Gene

One member of the mammalian Coronin gene family that is frequently observed to be up- or down-regulated in DNA microarray experiments is Coronin 1C. These data strongly suggest that this gene is transcriptionally dynamic, a finding supported by the fact that Coronin 1C was discovered in the early ‘90s as a gene that is induced by serum stimulation in fibroblasts.5 In this early work, Coronin 1C was identified as mig-3 for mitogen induced gene 3 and was not pursued. In 2004, Coronin 1C was independently identified as a serum-induced gene as part of a systematic search for genes that reflected a ‘wound signature’ in fibroblasts.6 The authors reasoned that fibroblast wound healing and tumor metastasis likely share a common set of up-regulated genes that may have prognostic value for clinical outcomes. Indeed, the expression of the 512 genes identified as part of the ‘core serum response’ does have strong predictive value for patient survival across a variety of tumors. More recently, Coronin 1C was identified as a delayed primary response gene in PDGF-stimulated glioblastoma cells.7 Together, these studies demonstrate that Coronin 1C is up-regulated at the transcriptional level by extracellular stimuli such as serum and growth factors. However, since these stimuli activate a large number of signaling cascades, these studies do not reveal much about the specific molecular events that increase Coronin 1C transcription.

In addition to these studies, other experiments have identified Coronin 1C as a target of specific pathways utilizing the transcription factors c-Myc/Max and Slug/Snail/E47. The activity of c-Myc and its partner Max has been frequently associated with human malignancies by contributing to cellular transformation and evasion of apoptosis. Coronin 1C was identified in a genome wide screen for promoters that bound to c-Myc in Burkitt's lymphoma cells.8 While this finding is intriguing, it is worth noting that this screen yielded a large number of target genes (>900) and may reflect global transcriptional regulation by c-Myc. In a more specific screen, Coronin 1C was identified as one of twenty target genes that were up-regulated upon expression of the transcription factors Snail, Slug and E47 in epithelial cells.9 Each of these transcription factors has been associated with epithelial-to-mesenchymal transition (EMT) in epithelial cells and they are consistently over-expressed in carcinoma cell lines with invasive and metastatic properties. This result suggests that Coronin 1C may be a strong candidate as both a biomarker for invasive cancer and functionally important for disease progression.

Consistent with the transcriptional dynamism of Coronin 1C, many other studies have identified this gene as either up- or down-regulated under various conditions (Fig. 1 and Table 1) and here we will highlight several of these studies that relate to cancer progression. Endometrial carcinoma (EC) is an extremely common gynecological malignancy that comprises two different types of tumor with a more benign type I EC and the more clinically aggressive nonendometriod endometrial carcinoma (NEEC). Coronin 1C is up-regulated 2.3 fold in NEEC relative to type I EC.10 In another study, Coronin 1C was identified as part of a 99-gene hypoxia expression signature in head and neck squamous cell carcinomas (HNSCC).11 Since hypoxia is a strong negative prognostic factor in many tumors including HNSCC, this also points to the involvement of Coronin 1C in disease progression in this malignancy.12 Finally, Coronin 1C was identified as one of 79 genes up-regulated in androgen-insensitive prostate cancer cell lines relative to androgen-sensitive cell lines.13 Since androgen insensitivity is a hallmark of aggressive prostate cancer, this further reinforces the link between Coronin 1C expression and invasion and metastasis.14

Figure 1. Inputs into the Coronin 1C promoter.

Figure 1

Inputs into the Coronin 1C promoter. Schematic diagram of the known signaling inputs that affect Coronin 1C transcription.

Table 1. Conditions under which Coronin 1C is transcriptionally regulated.

Table 1

Conditions under which Coronin 1C is transcriptionally regulated.

Coronin 1C as a Marker of Melanoma Progression

Advanced melanoma is one of the most feared human cancers.15,16 Although curable through surgery when diagnosed at early stage, melanoma is characterized by its therapeutic resistance and aggressive clinical behavior. A proclivity for early metastasis is a key clinical feature of melanoma and in perhaps no other malignancy is the ability to metastasize more closely correlated with clinical outcome.17,18 On the molecular level, one of the signature genetic events of melanoma is activation of the Ras/Raf pathway, with the majority (90%) of human melanomas harboring activating mutations in either N-RAS or B-RAF.19-21 Activation of either of these molecules induces persistent activation of the Erk mitogen-activated protein kinase (MAPK) cascade,22 suggesting that aberrant activation of the Erk pathway is a critical step in melanoma development. A major consequence of Erk activation is regulation of gene expression. However, the gene targets of this pathway important for oncogenesis in melanoma have only recently begun to be elucidated.23 These gene expression experiments and those from other groups have shown that cytoskeletal regulatory genes are frequently up-regulated during metastasis,24,25 highlighting the importance of cell migration in the etiology of metastatic cancer. Understanding how actin dynamics become mis-regulated during metastasis may allow the identification of new diagnostic markers that predict metastatic potential and provide candidate targets for clinical intervention.

Recent work has examined Coronin 1C protein levels in both normal human skin and human melanoma.26 Immunohistochemical staining reveals that, in normal skin, Coronin 1C levels are relatively low (Fig. 2A). This starkly contrasts with severe dysplastic nevi (abnormal moles more likely to develop into melanomas) and metastases, where the staining of tumor cells is much stronger (Fig. 2A). In order to determine if Erk is responsible for Coronin 1C up-regulation, three B-RAF mutant melanoma cell lines with high phospho-Erk levels (WM2664, SKMel24 and SKMel28) were examined by quantitative real-time PCR.23 All three contain high levels of Coronin 1C mRNA which are significantly reduced upon treatment with the Erk inhibitor U0126 (Fig. 2B). These data indicate that Coronin 1C is a potential marker for melanoma progression and that its expression is at least partly dependent on activation of the Erk pathway.

Figure 2. Coronin 1C in melanoma.

Figure 2

Coronin 1C in melanoma. A) Immunohistochemical staining of the indicated tissue samples for Coronin 1C. 5 μm sections from melanoma tumors were stained with a new Coronin 1C specific mAb (Roadcap and Bear, unpublished reagent) for 30min at 37°C (more...)

Coronin 1C Expression Is Regulated during Murine Brain Development

The dynamics of Coronin 1C (Coronin 3) expression have also been studied in both the development and diseased state of the brain. Studies of Coronin 1C in brain tumor cells revealed a similar story to that presented above for melanoma and are presented in the next section of this chapter. Coronin 1C expression was also high in all regions of the developing brain and this section discusses the developmental and cell specific expression patterns that are observed during further brain maturation.27

The neocortex consists of different laminae that are formed sequentially during the first postnatal days with the inner laminae differentiating first.28 Coronin 1C expression follows this inside-out gradient and deeper cortical layers are the first to show a decrease in the expression of Coronin 1C.

By the later stages of brain development, Coronin 1C is detected only in a small peripheral cortical layer. Finally, only weak basal levels of Coronin 1C remain detectable after terminal differentiation of the cortical neurons (Fig. 3A, P1 and P20 are shown). Similarly, the high levels of Coronin 1C in immature neurons of the cerebellum are decreased after formation of the granule cell layers (Fig. 3B). Some types of neurons, including hippocampal pyramidal CA1-CA3 and Purkinje cells, retain strong expression of Coronin 1C until early murine adulthood (Fig. 3C, left image, analyzed up to 80 days). These neurons harbor a high degree of synaptic plasticity, which enables them to undergo long-term potentiation and depression processes like learning and memory.29-32 Inversely to the patterns of the grey matter, areas of the white matter increase the expression of Coronin 1C (Fig. 3C, right image, only P30 is shown) during the progress of myelination.33,34 It has been suggested that these localization patterns place Coronin 1C in a position to play a role in diverse processes such as neuronal migration, differentiation, formation of neurites and the formation of lamellar protrusions and myelin sheaths of oligodendroglial cells.27

Figure 3. Coronin 1C expression levels during maturation of the grey and white matter of murine brain.

Figure 3

Coronin 1C expression levels during maturation of the grey and white matter of murine brain. Localization of Coronin 1C (Coronin 3) in sections of postnatal neocortex (A) and cerebellar sections (B, one folium is shown) of stages P1 and P20 analyzed by (more...)

These tightly controlled expression patterns suggest that imbalances in the expression of Coronin 1C during brain development might lead to defects in brain morphology. This hypothesis is supported by the observation that another member of the Coronin protein family, Coronin 1A, exhibits reduced expression levels in Down syndrome brain.35 It is noteworthy that other WD40-repeat containing proteins—Coronin proteins represent the cytoskeleton-related subfamily of WD40-repeat proteins (see Chapter 2)—also have been linked to human diseases with malformations of the nervous system, namely the lissencephaly,36,37 Cockayne syndrome,38 late-onset sensorineural deafness39 and the alacrima-achalasia-adrenal insufficiency (triple-A-syndrome).40,41

Coronin 1C Is Associated with the Malignant Phenotype of Human Diffuse Gliomas

In parallel to the work on Coronin 1C (Coronin 3) in melanoma progression, Coronin 1C's role in coordinating migration and invasion of tumor cells in the human brain has been investigated. Previously, Coronin 1C was reported to exert a role in regulation of F-actin turnover during cell migration and neurite outgrowth.4,42 To address a possible role of Coronin 1C in brain tumor cells, its expression in normal human brain tissue and various entities of human brain tumors was determined.43

In the normal adult human brain tissue, neurons in the cortex have little Coronin 1C, while the white matter do not show any significant expression. In brain tumors, the expression levels of Coronin 1C depend on the tumor type. Benign meningiomas (Fig. 4A, left image), pilocytic astrocytomas, high-grade anaplastic astrocytomas, oligodendrogliomas, oligoastrocytomas and glioblastomas all strongly express Coronin 1C. Low grade diffuse astrocytomas (Fig. 4A, right image), oligodendrogliomas and oligoastrocytomas show only weak staining of Coronin 1C in the tumor cells, while no expression of Coronin 1C is detectable in tumors of neuronal origin like the desmoplastic medulloblastoma and neuroblastoma. In mixed glial tumors, the astroglial portion of the tumor often expresses Coronin 1C at higher levels than the oligodendroglial component. Within an individual tumor, tumor cells adjacent to proliferating microvessels exhibit a stronger Coronin 1C staining than those adjacent to areas of necrosis (Fig. 4B). In addition, the tumor matrix contains a varying number of reactive astrocytes, activated and migrating astrocytic cells, which also highly express Coronin 1C. This also was the case for reactive astrocytes and microglial cells that had been triggered by circumscriptive nontumor lesions such as infarcts and traumatic lesions (Fig. 4C, left image). In contrast, reactive astrocytes in epilepsia-related diffuse astrogliosis less strongly express Coronin 1C.

Figure 4. Coronin 1C expression levels are associated with the malignant phenotype of human gliomas.

Figure 4

Coronin 1C expression levels are associated with the malignant phenotype of human gliomas. Paraffin-embedded tissue sections of human brain tumors were immunostained with a monoclonal antibody directed against Coronin 1C (Coronin 3), followed by biotinylated (more...)

A finding of significance is that Coronin 1C was associated with the malignant phenotype of diffuse gliomas.43 This class of tumors includes astrocytomas, oligodendrogliomas and oligoastrocytomas. Diffuse gliomas clearly have a strong correlation between high Coronin 1C expression levels and increased grade of the malignancy (Fig. 4C, right image). Coronin 1C's demonstrated role in actin dynamics and cell motility suggests that its expression in astroglial cells and in glial tumor cells is related to migration and the malignant phenotype. This hypothesis was tested by shRNA-mediated knockdown of Coronin 1C in U373 and A172 glioblastoma cells.43 Silencing Coronin 1C expression significantly reduces cell proliferation rates, cell migration velocity, formation of invadopodia, secretion of matrix metalloproteinases and invasion into extracellular matrix (Fig. 5A-C and data not shown). Previously, the degree of migration and invasion of tumor cells into healthy tissue was found to be a critical parameter of malignancy in diffuse gliomas and, moreover, a factor that limits the success of surgery.44 Thus, these data provide strong evidence that the expression level of Coronin 1C correlates with the grade of malignancy of diffuse gliomas and that Coronin 1C is significantly involved in tumor cell proliferation, migration, invasion, malignant progression and possibly prognosis of diffuse gliomas.

Figure 5. Coronin 1C influences malignancy-related cellular functions.

Figure 5

Coronin 1C influences malignancy-related cellular functions. A) Proliferation rates of U373 and A172 glioblastoma cells lacking Coronin 1C (Coronin 3) expression (kd) are compared to control cells (ctrl); a certain number of cells was seeded and counted (more...)

Other Coronin Genes and Disease

Coronin 1A (Coronin 1) is highly expressed in the hematopoetic system and regulates F-actin content in thymocytes (discussed below).45 Gene expression array studies suggest that its expression may be altered in lymphomas and other tumors of hematopeotic origin, although there is no apparent trend between cancer progression and Coronin 1A expression.46-50 Since control of actin dynamics is an integral part many disease states, it follows that Coronins could play a role in diseases other than cancer. The best-characterized example of this is Coronin 1A's role in pathogenic bacterial infection.51 The standard immune response to bacterial infection involves macrophages engulfing bacteria via phagocytosis. By utilizing controlled membrane fusion of phagosomes with lysosomes, the macrophages are able to deliver lytic enzymes, acidify phagolysosomes and subsequently destroy the bacteria.52 Nucleation of actin filaments around nascent phagosomes plays an important role in mediating these critical vesicle fusion events and thus the immune response to bacterial infection can be considered an actin-dependent process.53 Pathogenic bacteria such as Mycobacterium tuberculosis can circumvent the response by retaining Coronin 1A (called TACO in the original study) on the phagosome surface, thereby preventing the maturation of the phagosomes and allowing bacterial propagation within the macrophage.51 The mechanism of retention of Coronin 1A on bacterial-containing phagosomes was unclear for many years. A recent study, however, has identified the bacterial protein lipoamide dehydrogenase C (LpdC) as a Coronin 1A binding protein that recruits and retains Coronin 1A on the phagosome.54 In this work, the authors demonstrated that enforced expression of LpdC increased the survival of normally nonpathogenic bacteria in macrophages. Together, these data are highly suggestive that pathogenic bacteria are hijacking Coronin 1A function to alter actin dynamics on macrophage phagosomes and thereby evading the normal immune response.

Alternately, evidence also exists that Coronin 1A's role may be to control calcium dynamics after bacterial infection.55 Macrophages from mice lacking Coronin 1A do not display the persistent increase in calcium levels that are normally displayed after infection. This consequently prevents the activation of signaling pathways via proteins such as the calcium-sensitive phosphatase Calcineurin. While no alterations in actin dependent processes have been detected in Coronin 1A null macrophages, the effects may be linked to more subtle effects on actin that affect the known relationship between cytoskeletal dynamics and calcium trafficking.56,57 Dramatic effects on actin-dependent processes may also be masked by the presence of functionally redundant Coronin isoforms in this cell type.

While Coronin 1A is utilized to the detriment of macrophages after pathogenic bacterial infection, it plays a large role in the normal immune system as the most prominent hematopoietic Coronin.45 For instance, T-cells from Coronin 1A null mice are profoundly defective in both their ability to undergo chemotaxis in vitro and to home to secondary lymphocyte organs in vivo. Together, these defects lead to a decrease in the levels of CD4+ and CD8+ T-cells in the blood, spleen and lymph nodes of Coronin 1A deficient mice. This phenotype was linked to Coronin 1A's ability to control steady-state F-actin levels via regulation of Arp2/3. Loss of Coronin 1A could therefore lead to an increased rate of immune system failure and a consequent susceptibility to numerous diseases, although this hypothesis has yet to be tested. Further information on Coronin proteins in lymphocytes and macrophages can be found in Chapters 8 6th section and 10.

Coronin 1B (Coronin 2) is the most ubiquitously expressed mammalian isoform of Coronin, but relatively few studies have examined its role in disease states.58 It has, however, been linked to neurite formation and axon regeneration following spinal cord injury.59 p53 activation following injury stimulates the formation and growth of neurites that facilitate regeneration of neural pathways and knockdown of Coronin 1B decreases this neurite outgrowth.60 Coronin 1B activity therefore, like that of Coronin 1C, may be linked to neurodegenerative diseases or even control of neuronal plasticity. Further study of Coronin 1B is necessary to facilitate understanding of its role in diseases, but there is strong evidence that it plays an important role in regulating actin dynamics at the leading edge of motile cells.3 Disruption of this process may be important for a number of motility-involving diseases.

Unanswered Questions and Future Directions

Although Coronins (and particularly Coronin 1C (Coronin 3)) have been implicated in disease states, much work remains to establish functional links between transcriptional regulation and phenotypic change. One important area that will require more work is the careful delineation of the Coronin 1C promoter and its immediate upstream inputs. In addition, the biochemical mechanism of Coronin 1C activity needs to be more precisely determined and the relevant binding partners necessary for this activity need to be identified. Understanding these issues may be important for revealing how diverse signaling cascades are dis-regulated in invasive/metastatic cancer. Another area that will require future studies is clarifying the role that Coronin proteins play in development. Continued analysis of mouse gene deletion models should be quite informative in this regard.

Acknowledgements

We would like to thank Dan Zedek, Nancy Thomas and Nick Holoweckyj for contributions to Figure 2. DR is supported by NIH (NRSA, F32 CA128297), CSC is supported by the DFG (NO 113/13-3) and Köln Fortune, JEB is supported by the Melanoma Research Foundation and the Sontag Foundation.

References

1.
Rybakin V, Clemen CS. Coronin proteins as multifunctional regulators of the cytoskeleton and membrane trafficking. Bioessays. 2005;27(6):625–632. [PubMed: 15892111]
2.
Uetrecht AC, Bear JE. Coronins: the return of the crown. Trends Cell Biol. 2006;16(8):421–426. [PubMed: 16806932]
3.
Cai L, Marshall TW, Uetrecht AC. et al. Coronin 1B coordinates Arp2/3 complex and cofilin activities at the leading edge. Cell. 2007;128(5):915–929. [PMC free article: PMC2630706] [PubMed: 17350576]
4.
Rosentreter A, Hofmann A, Xavier CP. et al. Coronin 3 involvement in F-actin-dependent processes at the cell cortex. Exp Cell Res. 2007;313(5):878–895. [PubMed: 17274980]
5.
Wick M, Burger C, Brusselbach S. et al. Identification of serum-inducible genes: different patterns of gene regulation during G0←S and G1←S progression. J Cell Sci. 1994;107(Pt 1):227–239. [PubMed: 8175911]
6.
Chang HY, Sneddon JB, Alizadeh AA. et al. Gene Expression Signature of Fibroblast Serum Response Predicts Human Cancer Progression: Similarities between Tumors and Wounds. PLoS Biol. 2004;2(2):E7. [PMC free article: PMC314300] [PubMed: 14737219]
7.
Tullai JW, Schaffer ME, Mullenbrock S. et al. Immediate-early and delayed primary response genes are distinct in function and genomic architecture. J Biol Chem. 2007;282(33):23981–95. [PMC free article: PMC2039722] [PubMed: 17575275]
8.
Li Z, Van Calcar S, Q u C. et al. A global transcriptional regulatory role for c-Myc in Burkitt's lymphoma cells. Proc Natl Acad Sci USA. 2003;100(14):8164–8169. [PMC free article: PMC166200] [PubMed: 12808131]
9.
Moreno-Bueno G, Cubillo E, Sarrio D. et al. Genetic profiling of epithelial cells expressing e-cadherin repressors reveals a distinct role for snail, slug and e47 factors in epithelial-mesenchymal transition. Cancer Res. 2006;66(19):9543–9556. [PubMed: 17018611]
10.
Moreno-Bueno G, Sanchez-Estevez C, Cassia R. et al. Differential gene expression profile in endometrioid and nonendometrioid endometrial carcinoma: STK15 is frequently overexpressed and amplified in nonendometrioid carcinomas. Cancer Res. 2003;63(18):5697–5702. [PubMed: 14522886]
11.
Winter SC, Buffa FM, Silva P. et al. Relation of a hypoxia metagene derived from head and neck cancer to prognosis of multiple cancers. Cancer Res. 2007;67(7):3441–3449. [PubMed: 17409455]
12.
Nordsmark M, Bentzen SM, Rudat V. et al. Prognostic value of tumor oxygenation in 397 head and neck tumors after primary radiation therapy. An international multi-center study. Radiother Oncol. 2005;77(1):18–24. [PubMed: 16098619]
13.
Zhao H, Kim Y, Wang P. et al. Genome-wide characterization of gene expression variations and DNA copy number changes in prostate cancer cell lines. Prostate. 2005;63(2):187–197. [PubMed: 15486987]
14.
Bonaccorsi L, Muratori M, Carloni V. et al. Androgen receptor and prostate cancer invasion. Int J Androl. 2003;26(1):21–25. [PubMed: 12534934]
15.
Sharpless NE, Chin L. The INK4a/ARF locus and melanoma. Oncogene. 2003;22(20):3092–3098. [PubMed: 12789286]
16.
Chin L, Merlino G, DePinho RA. Malignant melanoma: modern black plague and genetic black box. Genes Dev. 1998;12(22):3467–3481. [PubMed: 9832500]
17.
Cochran AJ. Prediction of outcome for patients with cutaneous melanoma. Pigment Cell Res. 1997;10(3):162–167. [PubMed: 9266604]
18.
Ahmed I. Malignant melanoma: prognostic indicators. Mayo Clin Proc. 1997;72(4):356–361. [PubMed: 9121184]
19.
Davies H, Bignell GR, Cox C. et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949–954. [PubMed: 12068308]
20.
Omholt K, Karsberg S, Platz A. et al. Screening of N-ras codon 61 mutations in paired primary and metastatic cutaneous melanomas: mutations occur early and persist throughout tumor progression. Clin Cancer Res. 2002;8(11):3468–3474. [PubMed: 12429636]
21.
Herlyn M, Satyamoorthy K. Activated ras. Yet another player in melanoma? Am J Pathol. 1996;149(3):739–744. [PMC free article: PMC1865164] [PubMed: 8780377]
22.
Schulze A, Nicke B, Warne PH. et al. The transcriptional response to Raf activation is almost completely dependent on Mitogen-activated Protein Kinase Kinase activity and shows a major autocrine component. Mol Biol Cell. 2004;15(7):3450–3463. [PMC free article: PMC452596] [PubMed: 15090615]
23.
Shields JM, Thomas NE, Cregger M. et al. Lack of extracellular signal-regulated kinase mitogen-activated protein kinase signaling shows a new type of melanoma. Cancer Res. 2007;67(4):1502–1512. [PubMed: 17308088]
24.
Clark EA, Golub TR, Lander ES. et al. Genomic analysis of metastasis reveals an essential role for RhoC. Nature. 2000;406(6795):532–535. [PubMed: 10952316]
25.
Wang W, Wyckoff JB, Goswami S. et al. Coordinated regulation of pathways for enhanced cell motility and chemotaxis is conserved in rat and mouse mammary tumors. Cancer Res. 2007;67(8):3505–3511. [PubMed: 17440055]
26.
Bear JE, Roadcap DW. Unpublished Data. 2007.
27.
Hasse A, Rosentreter A, Spoerl Z. et al. Coronin 3 and its role in murine brain morphogenesis. Eur J Neurosci. 2005;21(5):1155–1168. [PubMed: 15813925]
28.
Berry M. Development of the cerebral neocortex of the rat. In: Gottlieb G, ed. Aspects of Neurogenesis. New York: Academic Press. 1974;2:7–67.
29.
Massey PV, Bashir ZI. Long-term depression: multiple forms and implications for brain function. Trends Neurosci. 2007;30(4):176–184. [PubMed: 17335914]
30.
Matsuzaki M, Honkura N, Ellis-Davies GC. et al. Structural basis of long-term potentiation in single dendritic spines. Nature. 2004;429(6993):761–766. [PMC free article: PMC4158816] [PubMed: 15190253]
31.
Krucker T, Siggins GR, Halpain S. Dynamic actin filaments are required for stable long-term potentiation (LTP) in area CA1 of the hippocampus. Proc Natl Acad Sci USA. 2000;97(12):6856–6861. [PMC free article: PMC18765] [PubMed: 10823894]
32.
Fukazawa Y, Saitoh Y, Ozawa F. et al. Hippocampal LTP is accompanied by enhanced F-actin content within the dendritic spine that is essential for late LTP maintenance in vivo. Neuron. 2003;38(3):447–460. [PubMed: 12741991]
33.
Bjartmar C, Hildebrand C, Loinder K. Morphological heterogeneity of rat oligodendrocytes: Electron microscopic studies on serial sections. Glia. 1994;11:235–244. [PubMed: 7960028]
34.
Coffrey JC, McDermott KW. The regional distribution of myelin oligodendrocyte glycoprotein (MOG) in the developing rat CNS: An in vivo immunohistochemical study. J Neurocytol. 1997;26:149–161. [PubMed: 9192283]
35.
Weitzdoerfer R, Fountoulakis M, Lubec G. Reduction of actin-related protein complex 2/3 in fetal Down syndrome brain. Biochem Biophys Res Commun. 2002;293(2):836–841. [PubMed: 12054546]
36.
Neer EJ, Schmidt CJ, Smith T. LIS is more. Nat Genet. 1993;5(1):3–4. [PubMed: 8220419]
37.
Lo Nigro C, Chong CS, Smith AC. et al. Point mutations and an intragenic deletion in LIS1, the lissencephaly causative gene in isolated lissencephaly sequence and Miller-Dieker syndrome. Hum Mol Genet. 1997;6(2):157–164. [PubMed: 9063735]
38.
Henning KA, Li L, Iyer N. et al. The Cockayne syndrome group A gene encodes a WD repeat protein that interacts with CSB protein and a subunit of RNA polymerase II TFIIH. Cell. 1995;82(4):555–564. [PubMed: 7664335]
39.
Bassi MT, Ramesar RS, Caciotti B. et al. X-linked late-onset sensorineural deafness caused by a deletion involving OA1 and a novel gene containing WD-40 repeats. Am J Hum Genet. 1999;64(6):1604–1616. [PMC free article: PMC1377903] [PubMed: 10330347]
40.
Tullio-Pelet A, Salomon R, Hadj-Rabia S. et al. Mutant WD-repeat protein in triple-A syndrome. Nat Genet. 2000;26(3):332–335. [PubMed: 11062474]
41.
Handschug K, Sperling S, Yoon SJ. et al. Triple A syndrome is caused by mutations in AAAS, a new WD-repeat protein gene. Hum Mol Genet. 2001;10(3):283–290. [PubMed: 11159947]
42.
Spoerl Z, Stumpf M, Noegel AA. et al. Oligomerization, F-actin interaction and membrane association of the ubiquitous mammalian coronin 3 are mediated by its carboxyl terminus. J Biol Chem. 2002;277(50):48858–48867. [PubMed: 12377779]
43.
Thal DR, Xavier CP, Rosentreter A. et al. Expression of coronin 3 in diffuse gliomas is related to malignancy J Pathol In press . [PubMed: 18189330]
44.
Kleihues P, Cavenee WK. Pathology and Genetics: Tumours of the Nervous System. Lyon: IARC-Press; 2000.
45.
Foger N, Rangell L, Danilenko DM. et al. Requirement for coronin 1 in T-lymphocyte trafficking and cellular homeostasis. Science. 2006;313(5788):839–842. [PubMed: 16902139]
46.
Yanagisawa Y, Sato Y, Asahi-Ozaki Y. et al. Effusion and solid lymphomas have distinctive gene and protein expression profiles in an animal model of primary effusion lymphoma. J Pathol. 2006;209(4):464–473. [PubMed: 16741895]
47.
Graham SM, Vass JK, Holyoake TL. et al. Transcriptional analysis of quiescent and proliferating CD34+ human haemopoietic cells from normal and CML sources. Stem Cells. 2007;25(12):3111–20. [PubMed: 17717066]
48.
Wilson CS, Davidson GS, Martin SB. et al. Gene expression profiling of adult acute myeloid leukemia identifies novel biologic clusters for risk classification and outcome prediction. Blood. 2006;108(2):685–696. [PMC free article: PMC1895492] [PubMed: 16597596]
49.
Sasaki H, Nishikata I, Shiraga T. et al. Overexpression of a cell adhesion molecule, TSLC1, as a possible molecular marker for acute-type adult T-cell leukemia. Blood. 2005;105(3):1204–1213. [PubMed: 15471956]
50.
Mahadevan D, Spier C, Della Croce K. et al. Transcript profiling in peripheral T-cell lymphoma, not otherwise specified and diffuse large B-cell lymphoma identifies distinct tumor profile signatures. Mol Cancer Ther. 2005;4(12):1867–1879. [PubMed: 16373702]
51.
Ferrari G, Langen H, Naito M. et al. A coat protein on phagosomes involved in the intracellular survival of mycobacteria. Cell. 1999;97(4):435–447. [PubMed: 10338208]
52.
Vergne I, Chua J, Singh SB. et al. Cell biology of mycobacterium tuberculosis phagosome. Annu Rev Cell Dev Biol. 2004;20:367–394. [PubMed: 15473845]
53.
Anes E, Kuhnel MP, Bos E. et al. Selected lipids activate phagosome actin assembly and maturation resulting in killing of pathogenic mycobacteria. Nat Cell Biol. 2003;5(9):793–802. [PubMed: 12942085]
54.
Deghmane AE, Soulhine H, Bach H. et al. Lipoamide dehydrogenase mediates retention of coronin-1 on BCG vacuoles, leading to arrest in phagosome maturation. J Cell Sci. 2007;120(16):2796–806. [PubMed: 17652161]
55.
Jayachandran R, Sundaramurthy V, Combaluzier B. et al. Survival of mycobacteria in macrophages is mediated by coronin 1-dependent activation of calcineurin. Cell. 2007;130(1):37–50. [PubMed: 17632055]
56.
Parekh AB, Putney JW. Jr. Store-operated calcium channels. Physiol Rev. 2005;85(2):757–810. [PubMed: 15788710]
57.
Gallo EM, Cante-Barrett K, Crabtree GR. Lymphocyte calcium signaling from membrane to nucleus. Nat Immunol. 2006;7(1):25–32. [PubMed: 16357855]
58.
Cai L, Holoweckyj N, Schaller MD. et al. Phosphorylation of coronin 1B by protein kinase C regulates interaction with Arp2/3 and cell motility. J Biol Chem. 2005;280(36):31913–31923. [PubMed: 16027158]
59.
Di Giovanni S, De Biase A, Yakovlev A. et al. In vivo and in vitro characterization of novel neuronal plasticity factors identified following spinal cord injury. J Biol Chem. 2005;280(3):2084–2091. [PubMed: 15522871]
60.
Di Giovanni S, Knights CD, Rao M. et al. The tumor suppressor protein p53 is required for neurite outgrowth and axon regeneration. EMBO J. 2006;25(17):4084–4096. [PMC free article: PMC1560361] [PubMed: 16946709]
61.
Martinez I, Lombardia L, Garcia-Barreno B. et al. Distinct gene subsets are induced at different time points after human respiratory syncytial virus infection of A549 cells. J Gen Virol. 2007;88(Pt 2):570–581. [PubMed: 17251576]
62.
Fujimura N, Vacik T, Machon O. et al. Wnt-mediated down-regulation of Sp1 target genes by a transcriptional repressor Sp5. J Biol Chem. 2007;282(2):1225–1237. [PubMed: 17090534]
63.
Hartman ZC, Kiang A, Everett RS. et al. Adenovirus infection triggers a rapid, MyD88-regulated transcriptome response critical to acute-phase and adaptive immune responses in vivo. J Virol. 2007;81(4):1796–1812. [PMC free article: PMC1797572] [PubMed: 17121790]
64.
Pellagatti A, Cazzola M, Giagounidis AA. et al. Gene expression profiles of CD34+ cells in myelodysplastic syndromes: involvement of interferon-stimulated genes and correlation to FAB subtype and karyotype. Blood. 2006;108(1):337–345. [PubMed: 16527891]
65.
Kakiuchi S, Daigo Y, Ishikawa N. et al. Prediction of sensitivity of advanced nonsmall cell lung cancers to gefitinib (Iressa, ZD1839). Hum Mol Genet. 2004;13(24):3029–3043. [PubMed: 15496427]
66.
Hertel L, Mocarski ES. Global analysis of host cell gene expression late during cytomegalovirus infection reveals extensive dysregulation of cell cycle gene expression and induction of Pseudomitosis independent of US28 function. J Virol. 2004;78(21):11988–12011. [PMC free article: PMC523267] [PubMed: 15479839]
67.
Ning W, Li CJ, Kaminski N. et al. Comprehensive gene expression profiles reveal pathways related to the pathogenesis of chronic obstructive pulmonary disease. Proc Natl Acad Sci USA. 2004;101(41):14895–14900. [PMC free article: PMC522001] [PubMed: 15469929]
68.
Chen H, Huang XN, Stewart AF. et al. Gene expression changes associated with fibronectin-induced cardiac myocyte hypertrophy. Physiol Genomics. 2004;18(3):273–283. [PubMed: 15306692]
69.
Messmer D, Messmer B, Chiorazzi N. The global transcriptional maturation program and stimuli-specific gene expression profiles of human myeloid dendritic cells. Int Immunol. 2003;15(4):491–503. [PubMed: 12663679]
70.
Ross ME, Zhou X, Song G. et al. Classification of pediatric acute lymphoblastic leukemia by gene expression profiling. Blood. 2003;102(8):2951–2959. [PubMed: 12730115]

These authors contributed equally to this work.

Copyright © 2000-2013, Landes Bioscience.
Bookshelf ID: NBK6528

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...