1
|
Gomila Pelegri N, Stanczak AM, Bottomley AL, Milthorpe BK, Gorrie CA, Padula MP, Santos J. Adipose-Derived Stem Cells Spontaneously Express Neural Markers When Grown in a PEG-Based 3D Matrix. Int J Mol Sci 2023; 24:12139. [PMID: 37569515 PMCID: PMC10418654 DOI: 10.3390/ijms241512139] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Neurological diseases are among the leading causes of disability and death worldwide and remain difficult to treat. Tissue engineering offers avenues to test potential treatments; however, the development of biologically accurate models of brain tissues remains challenging. Given their neurogenic potential and availability, adipose-derived stem cells (ADSCs) are of interest for creating neural models. While progress has been made in differentiating ADSCs into neural cells, their differentiation in 3D environments, which are more representative of the in vivo physiological conditions of the nervous system, is crucial. This can be achieved by modulating the 3D matrix composition and stiffness. Human ADSCs were cultured for 14 days in a 1.1 kPa polyethylene glycol-based 3D hydrogel matrix to assess effects on cell morphology, cell viability, proteome changes and spontaneous neural differentiation. Results showed that cells continued to proliferate over the 14-day period and presented a different morphology to 2D cultures, with the cells elongating and aligning with one another. The proteome analysis revealed 439 proteins changed in abundance by >1.5 fold. Cyclic nucleotide 3'-phosphodiesterase (CNPase) markers were identified using immunocytochemistry and confirmed with proteomics. Findings indicate that ADSCs spontaneously increase neural marker expression when grown in an environment with similar mechanical properties to the central nervous system.
Collapse
Affiliation(s)
- Neus Gomila Pelegri
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
- Neural Injury Research Unit, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Aleksandra M. Stanczak
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (A.M.S.); (M.P.P.)
| | - Amy L. Bottomley
- Microbial Imaging Facility, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Bruce K. Milthorpe
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
| | - Catherine A. Gorrie
- Neural Injury Research Unit, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Matthew P. Padula
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (A.M.S.); (M.P.P.)
| | - Jerran Santos
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
| |
Collapse
|
2
|
Cornelius J, Rottner K, Korte M, Michaelsen-Preusse K. Cortactin Contributes to Activity-Dependent Modulation of Spine Actin Dynamics and Spatial Memory Formation. Cells 2021; 10:cells10071835. [PMID: 34360003 PMCID: PMC8303107 DOI: 10.3390/cells10071835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 01/21/2023] Open
Abstract
Postsynaptic structures on excitatory neurons, dendritic spines, are actin-rich. It is well known that actin-binding proteins regulate actin dynamics and by this means orchestrate structural plasticity during the development of the brain, as well as synaptic plasticity mediating learning and memory processes. The actin-binding protein cortactin is localized to pre- and postsynaptic structures and translocates in a stimulus-dependent manner between spines and the dendritic compartment, thereby indicating a crucial role for synaptic plasticity and neuronal function. While it is known that cortactin directly binds F-actin, the Arp2/3 complex important for actin nucleation and branching as well as other factors involved in synaptic plasticity processes, its precise role in modulating actin remodeling in neurons needs to be deciphered. In this study, we characterized the general neuronal function of cortactin in knockout mice. Interestingly, we found that the loss of cortactin leads to deficits in hippocampus-dependent spatial memory formation. This impairment is correlated with a prominent dysregulation of functional and structural plasticity. Additional evidence shows impaired long-term potentiation in cortactin knockout mice together with a complete absence of structural spine plasticity. These phenotypes might at least in part be explained by alterations in the activity-dependent modulation of synaptic actin in cortactin-deficient neurons.
Collapse
Affiliation(s)
- Jonas Cornelius
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany; (J.C.); (M.K.)
| | - Klemens Rottner
- Research Group Molecular Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
- Division of Molecular Cell Biology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany; (J.C.); (M.K.)
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Kristin Michaelsen-Preusse
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany; (J.C.); (M.K.)
- Correspondence:
| |
Collapse
|
3
|
Kubo Y, Baba K, Toriyama M, Minegishi T, Sugiura T, Kozawa S, Ikeda K, Inagaki N. Shootin1-cortactin interaction mediates signal-force transduction for axon outgrowth. J Cell Biol 2015; 210:663-76. [PMID: 26261183 PMCID: PMC4539990 DOI: 10.1083/jcb.201505011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/26/2015] [Indexed: 11/22/2022] Open
Abstract
The shootin1–cortactin interaction participates in netrin-1–induced F-actin–adhesion coupling and in the promotion of traction forces for axon outgrowth. Motile cells transduce environmental chemical signals into mechanical forces to achieve properly controlled migration. This signal–force transduction is thought to require regulated mechanical coupling between actin filaments (F-actins), which undergo retrograde flow at the cellular leading edge, and cell adhesions via linker “clutch” molecules. However, the molecular machinery mediating this regulatory coupling remains unclear. Here we show that the F-actin binding molecule cortactin directly interacts with a clutch molecule, shootin1, in axonal growth cones, thereby mediating the linkage between F-actin retrograde flow and cell adhesions through L1-CAM. Shootin1–cortactin interaction was enhanced by shootin1 phosphorylation by Pak1, which is activated by the axonal chemoattractant netrin-1. We provide evidence that shootin1–cortactin interaction participates in netrin-1–induced F-actin adhesion coupling and in the promotion of traction forces for axon outgrowth. Under cell signaling, this regulatory F-actin adhesion coupling in growth cones cooperates with actin polymerization for efficient cellular motility.
Collapse
Affiliation(s)
- Yusuke Kubo
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Kentarou Baba
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Michinori Toriyama
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Takunori Minegishi
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Tadao Sugiura
- Laboratory of Biomedical Imaging, Graduate School of Information Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Satoshi Kozawa
- Mathematical Informatics, Graduate School of Information Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Kazushi Ikeda
- Mathematical Informatics, Graduate School of Information Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| |
Collapse
|
4
|
He Y, Ren Y, Wu B, Decourt B, Lee AC, Taylor A, Suter DM. Src and cortactin promote lamellipodia protrusion and filopodia formation and stability in growth cones. Mol Biol Cell 2015. [PMID: 26224308 PMCID: PMC4569314 DOI: 10.1091/mbc.e15-03-0142] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
How Src tyrosine kinase and cortactin control actin organization and dynamics in neuronal growth cones is not well understood. Using multiple high-resolution imaging techniques, this study shows that Src and cortactin control the persistence of lamellipodial protrusion as well as the formation, stability, and elongation of filopodia in growth cones. Src tyrosine kinases have been implicated in axonal growth and guidance; however, the underlying cellular mechanisms are not well understood. Specifically, it is unclear which aspects of actin organization and dynamics are regulated by Src in neuronal growth cones. Here, we investigated the function of Src2 and one of its substrates, cortactin, in lamellipodia and filopodia of Aplysia growth cones. We found that up-regulation of Src2 activation state or cortactin increased lamellipodial length, protrusion time, and actin network density, whereas down-regulation had opposite effects. Furthermore, Src2 or cortactin up-regulation increased filopodial density, length, and protrusion time, whereas down-regulation promoted lateral movements of filopodia. Fluorescent speckle microscopy revealed that rates of actin assembly and retrograde flow were not affected in either case. In summary, our results support a model in which Src and cortactin regulate growth cone motility by increasing actin network density and protrusion persistence of lamellipodia by controlling the state of actin-driven protrusion versus retraction. In addition, both proteins promote the formation and stability of actin bundles in filopodia.
Collapse
Affiliation(s)
- Yingpei He
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Yuan Ren
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Bingbing Wu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Boris Decourt
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Aih Cheun Lee
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Aaron Taylor
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907 Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907 )
| |
Collapse
|
5
|
PKC-epsilon activation is required for recognition memory in the rat. Behav Brain Res 2013; 253:280-9. [PMID: 23911427 DOI: 10.1016/j.bbr.2013.07.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 07/18/2013] [Accepted: 07/22/2013] [Indexed: 11/20/2022]
Abstract
Activation of PKCɛ, an abundant and developmentally regulated PKC isoform in the brain, has been implicated in memory throughout life and across species. Yet, direct evidence for a mechanistic role for PKCɛ in memory is still lacking. Hence, we sought to evaluate this in rats, using short-term treatments with two PKCɛ-selective peptides, the inhibitory ɛV1-2 and the activating ψɛRACK, and the novel object recognition task (NORT). Our results show that the PKCɛ-selective activator ψɛRACK, did not have a significant effect on recognition memory. In the short time frames used, however, inhibition of PKCɛ activation with the peptide inhibitor ɛV1-2 significantly impaired recognition memory. Moreover, when we addressed at the molecular level the immediate proximal signalling events of PKCɛ activation in acutely dissected rat hippocampi, we found that ψɛRACK increased in a time-dependent manner phosphorylation of MARCKS and activation of Src, Raf, and finally ERK1/2, whereas ɛV1-2 inhibited all basal activity of this pathway. Taken together, these findings present the first direct evidence that PKCɛ activation is an essential molecular component of recognition memory and point toward the use of systemically administered PKCɛ-regulating peptides as memory study tools and putative therapeutic agents.
Collapse
|
6
|
Chen Y, Fan JX, Zhang ZL, Wang G, Cheng X, Chuai M, Lee KKH, Yang X. The negative influence of high-glucose ambience on neurogenesis in developing quail embryos. PLoS One 2013; 8:e66646. [PMID: 23818954 PMCID: PMC3688607 DOI: 10.1371/journal.pone.0066646] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 05/08/2013] [Indexed: 12/19/2022] Open
Abstract
Gestational diabetes is defined as glucose intolerance during pregnancy and it is presented as high blood glucose levels during the onset pregnancy. This condition has an adverse impact on fetal development but the mechanism involved is still not fully understood. In this study, we investigated the effects of high glucose on the developing quail embryo, especially its impact on the development of the nervous system. We established that high glucose altered the central nervous system mophologically, such that neural tube defects (NTDs) developed. In addition, we found that high glucose impaired nerve differentiation at dorsal root ganglia and in the developing limb buds, as revealed by neurofilament (NF) immunofluorescent staining. The dorsal root ganglia are normally derived from neural crest cells (NCCs), so we examine the delamination of NCCs from dorsal side of the neural tube. We established that high glucose was detrimental to the NCCs, in vivo and in vitro. High glucose also negatively affected neural differentiation by reducing the number and length of neurites emanating from neurons in culture. We established that high glucose exposure caused an increase in reactive oxidative species (ROS) generation by primary cultured neurons. We hypothesized that excess ROS was the factor responsible for impairing neuron development and differentiation. We provided evidence for our hypothesis by showing that the addition of vitamin C (a powerful antioxidant) could rescue the damaging effects of high glucose on cultured neurons.
Collapse
Affiliation(s)
- Yao Chen
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Jian-xia Fan
- Department of Gynecology and Obstetrics, International Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhao-long Zhang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Guang Wang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Xin Cheng
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Manli Chuai
- Division of Cell and Developmental Biology, University of Dundee, Dundee, United Kingdom
| | - Kenneth Ka Ho Lee
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
- * E-mail: (XY); (KKHL)
| | - Xuesong Yang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
- Institute of Fetal-Preterm Labor Medicine, Jinan University, Guangzhou, China
- * E-mail: (XY); (KKHL)
| |
Collapse
|
7
|
Cheng X, Wang G, Ma ZL, Chen YY, Fan JJ, Zhang ZL, Lee KKH, Luo HM, Yang X. Exposure to 2,5-hexanedione can induce neural malformations in chick embryos. Neurotoxicology 2012; 33:1239-47. [DOI: 10.1016/j.neuro.2012.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 06/18/2012] [Accepted: 07/17/2012] [Indexed: 11/26/2022]
|
8
|
Asimaki O, Leondaritis G, Lois G, Sakellaridis N, Mangoura D. Cannabinoid 1 receptor-dependent transactivation of fibroblast growth factor receptor 1 emanates from lipid rafts and amplifies extracellular signal-regulated kinase 1/2 activation in embryonic cortical neurons. J Neurochem 2011; 116:866-73. [DOI: 10.1111/j.1471-4159.2010.07030.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
9
|
Asimaki O, Mangoura D. Cannabinoid receptor 1 induces a biphasic ERK activation via multiprotein signaling complex formation of proximal kinases PKCε, Src, and Fyn in primary neurons. Neurochem Int 2010; 58:135-44. [PMID: 21074588 DOI: 10.1016/j.neuint.2010.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 11/02/2010] [Accepted: 11/03/2010] [Indexed: 11/17/2022]
Abstract
Cannabinoid receptors 1 (CB1Rs) play important roles in the regulation of dendritic branching, synapse density, and synaptic transmission through multiple G-protein-coupled signaling systems, including the activation of the extracellular signal-regulated kinases ERK1/2. The proximal signaling interactions leading to ERK1/2 activation by CB1R in CNS remain, however, unclear. Here, we present evidence that the CB1R agonist methanandamide induced a biphasic and sustained activation of ERK1/2 in primary neurons derived from E7 telencephalon. We show that E7 neurons natively express high levels of CB1R message and protein, the majority of which associates with PKCɛ at basal conditions. We now demonstrate that the first peak of ERK activation by CB1R was mediated by the sequential activation of G(q), PLC, and PKCɛ, selectively, and that the CB1R-activated PKCɛ acutely formed transient signaling modules containing activated Src and Fyn. A second pool of CB1Rs, coupled to PTX-sensitive activation of G(i/o), utilized as effectors additional Src and Fyn molecules to generate a second, additional wave of ERK activation at 15 min. Concurrently to these intermolecular signaling interactions, cytoskeleton-associated proteins MARCKS and p120catenin were drastically modified by phosphorylation of PKC and Src, respectively. These receptor-proximal signaling events correlated well with induction of neuritic outgrowth in the long term. Our data provide evidence for multiprotein signaling complex formation in the coupling of CB1R to activation of ERK in CNS neurons, and may elucidate several of the less understood acute effects of cannabinoid drugs.
Collapse
Affiliation(s)
- Olga Asimaki
- Developmental Neurobiology and Neurochemistry Group, Basic Neurosciences, Center for Preventive Medicine, Neurosciences and Social Psychiatry, Biomedical Research Foundation of the Academy of Athens, 4, Soranou Ephessiou Street, 11527 Athens, Greece
| | | |
Collapse
|
10
|
Brown JA, Bridgman PC. Disruption of the cytoskeleton during Semaphorin 3A induced growth cone collapse correlates with differences in actin organization and associated binding proteins. Dev Neurobiol 2009; 69:633-46. [PMID: 19513995 DOI: 10.1002/dneu.20732] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Repulsive guidance cues induce growth cone collapse or collapse and retraction. Collapse results from disruption and loss of the actin cytoskeleton. Actin-rich regions of growth cones contain binding proteins that influence filament organization, such as Arp2/3, cortactin, and fascin, but little is known about the role that these proteins play in collapse. Here, we show that Semaphorin 3A (Sema 3A), which is repulsive to mouse dorsal root ganglion neurons, has unequal effects on actin binding proteins and their associated filaments. The immunofluorescence staining intensity of Arp-2 and cortactin decreases relative to total protein; whereas in unextracted growth cones fascin increases. Fascin and myosin IIB staining redistribute and show increased overlap. The degree of actin filament loss during collapse correlates with filament superstructures detected by rotary shadow electron microscopy. Collapse results in the loss of branched f-actin meshworks, while actin bundles are partially retained to varying degrees. Taken together with the known affects of Sema 3A on actin, this suggests a model for collapse that follows a sequence; depolymerization of actin meshworks followed by partial depolymerization of fascin associated actin bundles and their movement to the neurite to complete collapse. The relocated fascin associated actin bundles may provide the substrate for actomyosin contractions that produce retraction.
Collapse
Affiliation(s)
- Jacquelyn A Brown
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
11
|
Fantozzi I, Grall D, Cagnol S, Stanchi F, Sudaka A, Brunstein MC, Bozec A, Fischel JL, Milano G, Van Obberghen-Schilling E. Overexpression of cortactin in head and neck squamous cell carcinomas can be uncoupled from augmented EGF receptor expression. Acta Oncol 2008; 47:1502-12. [PMID: 18607838 DOI: 10.1080/02841860802089801] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND The gene encoding cortactin, CTTN (locus 11q13), an actin-binding substrate of Src kinases, is frequently amplified in breast and head and neck squamous cell carcinomas (HNSCC) and cortactin overexpression is thought to contribute in a significant way to the invasive phenotype of these tumors. Elevated Epidermal Growth Factor receptor (EGFR) expression is also commonly observed in HNSCC and has been associated with poor prognosis and resistance to cytotoxic agents, including ionizing radiation. It has been suggested that cortactin overexpression may increase EGFR levels in these tumors by affecting receptor downregulation, however we recently found by multivariate analysis, that cortactin expression status remained an independent prognostic factor for local recurrence, disease-free survival, and overall survival. MATERIAL AND METHODS To examine the potential link between cortactin overexpression and EGFR status, we compared cortactin and EGFR levels in a series of tumor lines derived from HNSCC. RNAi-mediated silencing was performed in cortactin overexpressing cells and in vivo tumoral potential with respect to cortactin and EGFR status was analyzed. RESULTS AND DISCUSSION Cortactin and EGFR levels were not strictly coupled in these lines and cortactin depletion did not decrease steady state receptor levels, although it did affect the epithelial to mesenchymal phenotypic conversion of cells. These results, together with clinical findings point to the existence of an EGFR-independent role of cortactin in HNSCC that may have important implications regarding the design of targeted therapies to combat tumor spread.
Collapse
|
12
|
Abstract
Cortactin, an actin filament-binding protein and target of multiple kinases, has emerged as a central element connecting signaling pathways with cytoskeleton restructuring. It is involved in a perplexingly diverse array of cellular processes, including cell motility, invasiveness, synaptogenesis, endocytosis, intercellular contact assembly, and host-pathogen interactions, where the common denominator appears to be a role in the coordination of membrane dynamics with cytoskeletal remodeling. Although in recent years our knowledge about cortactin has increased exponentially, the exact mechanisms underlying its fundamental roles remain to be defined.
Collapse
Affiliation(s)
- Laura I Cosen-Binker
- Saint Michael's Hospital Research Institute, Department of Surgery, University of Toronto, Ontario, Canada
| | | |
Collapse
|
13
|
Mangoura D, Sun Y, Li C, Singh D, Gutmann DH, Flores A, Ahmed M, Vallianatos G. Phosphorylation of neurofibromin by PKC is a possible molecular switch in EGF receptor signaling in neural cells. Oncogene 2006; 25:735-45. [PMID: 16314845 DOI: 10.1038/sj.onc.1209113] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Children with neurofibromatosis (NF1) typically develop central nervous system (CNS) abnormalities, including aberrant proliferation of astrocytes and formation of benign astrocytomas. The NF1 gene encodes neurofibromin, a Ras-GAP, highly expressed in developing neural cells; the mechanism of regulation of neurofibromin as a Ras-GAP, remains however unknown. We now show that, in response to EGF, neurofibromin is in vivo phosphorylated on serine residues by PKC-alpha, in human, rat, and avian CNS cells and cell lines. EGF-induced PKC phosphorylation was prominent in the cysteine/serine-rich domain (CSRD) of neurofibromin, which lies in the N-terminus and upstream of the Ras-GAP domain (GRD), and this modification significantly increased the association of neurofibromin with actin in co-immunoprecipitations. In addition, we show that Ras activation in response to EGF was significantly lowered when C62B cells overexpressed a construct encoding both CSRD + GRD. Moreover, when PKC-alpha was downregulated, the Ras-GAP activity of CSRD + GRD was significantly diminished, whereas overexpressed GRD alone acted as a weaker GAP and in a PKC-independent manner. Most importantly, functional Ras inhibition and EGF signaling shifts were established at the single cell level in C6-derived cell lines stably overexpressing CSRD + GRD, when transient co-overexpression of Ras and PKC-depletion prior to stimulation with EGF-induced mitosis. Taken together, these data provide the first evidence of a functional, allosteric regulation of GRD by CSRD, which requires neurofibromin phosphorylation by PKC and association with the actin cytoskeleton. Our data may suggest a novel mechanism for regulating biological responses to EGF and provide a new aspect for the understanding of the aberrant proliferation seen in the CNS of children with NF1.
Collapse
Affiliation(s)
- D Mangoura
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA. and Neurosciences Division, Institute for Biomedical Research, Athens, Greece
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Mangoura D, Theofilopoulos S, Karouzaki S, Tsirimonaki E. 12-O-tetradecanoyl-phorbol-13-acetate-dependent up-regulation of dopaminergic gene expression requires Ras and neurofibromin in human IMR-32 neuroblastoma. J Neurochem 2006; 97 Suppl 1:97-103. [PMID: 16635257 DOI: 10.1111/j.1471-4159.2005.03483.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The dopaminergic transcriptional programme is highly regulated during development and in the adult, in response to activation of membrane receptor signalling cascades. Gene expression of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine synthesis, is known to be regulated by receptors that act through protein kinase C (PKC) or Ras signalling. To investigate possible interactions between these two pathways before they converge on Raf activation, we evaluated whether phorbol ester (12-O-tetradecanoyl-phorbol-13-acetate, TPA)-dependent PKC activation required Ras for regulation of TH expression in IMR-32 cells. We found that long-term treatment with TPA, which induces down-regulation of PKC-alpha, led to induction of both protein and message levels of TH by autocrine factors. This was dependent on endogenous Ras, but independent of the transcription factor Nurr1. Moreover, this mechanism of action mimicked the effects of overexpression of the Ras-GAP domain of neurofibromin, GAP-related domain (GRD) I, which is part of the upstream mechanism for regulation of Ras activation and a PKC-alpha substrate. Overexpression of Ras also led to transcriptional and translational up-regulation of TH, independent of Nurr1 induction, as well as distinct phenotypic changes consistent with cell hypertrophy and increased secretory activity shown by induction of expression of vesicular monoamine transporter 2 and synaptosomal-associated protein-25. Most interestingly, overexpression of GRDI and down-regulation of the endogenous GRDII neurofibromin led to significant increases in Nurr1 message, possibly reflecting a transcriptional hierarchy during development. Taken together, these studies suggest that PKC-alpha, neurofibromin and Ras are essential in regulation of TH gene expression in IMR-32 cells.
Collapse
Affiliation(s)
- Dimitra Mangoura
- Neurosciences Division, Foundation for Biomedical Research of the Academy of Athens (IIBEAA), Athens, Greece.
| | | | | | | |
Collapse
|
15
|
Fischer TM, Steinmetz PN, Odde DJ. Robust micromechanical neurite elicitation in synapse-competent neurons via magnetic bead force application. Ann Biomed Eng 2005; 33:1229-37. [PMID: 16133929 DOI: 10.1007/s10439-005-5509-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2005] [Accepted: 04/25/2005] [Indexed: 11/27/2022]
Abstract
The ability to engineer living networks of interconnected neurons with specified connectivity would facilitate the study of synaptogenesis and information processing in the nervous system. Previously, we found that a neurite can be elicited from embryonic chick forebrain neurons by direct mechanical means using magnetic bead force application (MBFA); however, our previous studies and others focused on young, synapse-incompetent neurons. To address this issue, we tested cultures of embryonic chick forebrain neurons of varying age and found that neurites could be micromechanically elicited via MBFA at all ages tested, which ranged between 7 and 22 embryonic equivalent (EE) days (days in ovo plus days in vitro). The probability of neurite initiation was at least 40% for all ages, with a maximum of approximately 80% after 2-4 days in vitro, and a decrease to approximately 60% by day 10 in vitro. The force required to elicit a neurite was approximately 1500 pN with a minimum of approximately 700 pN at embryonic equivalent day 14. The probability of success was similar for two rates of force application (10 and 500 pN/s). Neurite initiation via micromechanical force is robust with respect to cell age, and micromechanical force can induce neurites in synapse-competent neurons.
Collapse
Affiliation(s)
- Trent M Fischer
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
16
|
van Rossum AGSH, Schuuring-Scholtes E, Seggelen VVBV, Kluin PM, Schuuring E. Comparative genome analysis of cortactin and HS1: the significance of the F-actin binding repeat domain. BMC Genomics 2005; 6:15. [PMID: 15710041 PMCID: PMC554100 DOI: 10.1186/1471-2164-6-15] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2004] [Accepted: 02/14/2005] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND In human carcinomas, overexpression of cortactin correlates with poor prognosis. Cortactin is an F-actin-binding protein involved in cytoskeletal rearrangements and cell migration by promoting actin-related protein (Arp)2/3 mediated actin polymerization. It shares a high amino acid sequence and structural similarity to hematopoietic lineage cell-specific protein 1 (HS1) although their functions differ considerable. In this manuscript we describe the genomic organization of these two genes in a variety of species by a combination of cloning and database searches. Based on our analysis, we predict the genesis of the actin-binding repeat domain during evolution. RESULTS Cortactin homologues exist in sponges, worms, shrimps, insects, urochordates, fishes, amphibians, birds and mammalians, whereas HS1 exists in vertebrates only, suggesting that both genes have been derived from an ancestor cortactin gene by duplication. In agreement with this, comparative genome analysis revealed very similar exon-intron structures and sequence homologies, especially over the regions that encode the characteristic highly conserved F-actin-binding repeat domain. Cortactin splice variants affecting this F-actin-binding domain were identified not only in mammalians, but also in amphibians, fishes and birds. In mammalians, cortactin is ubiquitously expressed except in hematopoietic cells, whereas HS1 is mainly expressed in hematopoietic cells. In accordance with their distinct tissue specificity, the putative promoter region of cortactin is different from HS1. CONCLUSIONS Comparative analysis of the genomic organization and amino acid sequences of cortactin and HS1 provides inside into their origin and evolution. Our analysis shows that both genes originated from a gene duplication event and subsequently HS1 lost two repeats, whereas cortactin gained one repeat. Our analysis genetically underscores the significance of the F-actin binding domain in cytoskeletal remodeling, which is of importance for the major role of HS1 in apoptosis and for cortactin in cell migration.
Collapse
MESH Headings
- Actin-Related Protein 2/chemistry
- Actin-Related Protein 3/chemistry
- Actins/chemistry
- Adaptor Proteins, Signal Transducing
- Alternative Splicing
- Animals
- Apoptosis
- Blood Proteins/chemistry
- Cell Line, Tumor
- Cell Lineage
- Cell Movement
- Cloning, Molecular
- Cortactin/chemistry
- Cytoskeleton/metabolism
- DNA, Complementary/metabolism
- Databases as Topic
- Databases, Factual
- Databases, Genetic
- Evolution, Molecular
- Exons
- Gene Duplication
- Genome
- Hematopoietic Stem Cells/cytology
- Humans
- Introns
- Models, Genetic
- Prognosis
- Promoter Regions, Genetic
- Protein Binding
- Protein Structure, Tertiary
- Proteins/chemistry
Collapse
Affiliation(s)
- Agnes GSH van Rossum
- Department of Pathology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
- Division of Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Ellen Schuuring-Scholtes
- Department of Pathology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | | | - Philip M Kluin
- Department of Pathology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
- Department of Pathology, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Ed Schuuring
- Department of Pathology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
- Department of Pathology, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
17
|
James J, Das AV, Rahnenführer J, Ahmad I. Cellular and molecular characterization of early and late retinal stem cells/progenitors: Differential regulation of proliferation and context dependent role of Notch signaling. ACTA ACUST UNITED AC 2004; 61:359-76. [PMID: 15452852 DOI: 10.1002/neu.20064] [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] [Indexed: 11/05/2022]
Abstract
Retinal stem cells/progenitors that define the evolutionarily conserved early and late stages of retinal histogenesis are known to have distinct competence to give rise to stage-specific retinal cell types. However, the information regarding their innate proliferative behavior and phenotypic potential in terms of generating neurons and glia is lacking. Here we demonstrate that, like their counterparts in other central nervous system (CNS) regions during early and late stages of embryonic development, the early and late retinal stem cells/progenitors display different proliferative response to fibroblast growth factor 2 (FGF2) and epidermal growth factor (EGF) and bias towards generating neurons or glia. Although the former predominantly generate neurons, the latter are partial towards giving rise to glia. Transcription profiling identified classes of genes that are differentially expressed in early and late retinal stem cells/progenitors in proliferating conditions and suggested that the distinct proliferative response to FGF2 and EGF is likely due to differential expression of FGF receptor 1 (FGFR1) and EGF receptor (EGFR). However, the proliferative maintenance of retinal stem cells/progenitors is likely to include other signaling pathways such as those mediated by insulin-like growth factors (IGFs) and stem cell factor (SCF). Transcription profiling of early and late retinal stem cells/progenitors in proliferating and differentiating conditions suggested a context dependent role for Notch signaling, which may constitute one of the mechanisms underlying the stage-dependent phenotypic potential of retinal stem cells/progenitors.
Collapse
MESH Headings
- Age Factors
- Animals
- Bromodeoxyuridine/metabolism
- Cell Count/methods
- Cell Differentiation/physiology
- Cell Proliferation
- Cells, Cultured
- Drug Interactions
- Embryo, Mammalian
- Enzyme Inhibitors/pharmacology
- Epidermal Growth Factor/pharmacology
- ErbB Receptors
- Female
- Fibroblast Growth Factor 2/pharmacology
- Fluorescent Antibody Technique/methods
- Gene Expression Regulation, Developmental
- Glial Fibrillary Acidic Protein/metabolism
- Glycoproteins/metabolism
- Intermediate Filament Proteins/metabolism
- Male
- Membrane Proteins/physiology
- Microtubule-Associated Proteins/metabolism
- Nerve Tissue Proteins/metabolism
- Nestin
- Neurons/enzymology
- Neurons/physiology
- Oligonucleotide Array Sequence Analysis/methods
- Pregnancy
- Proto-Oncogene Proteins c-kit/metabolism
- RNA, Messenger/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 1
- Receptor, IGF Type 2/metabolism
- Receptors, Fibroblast Growth Factor/metabolism
- Receptors, Notch
- Retina/cytology
- Retina/embryology
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Signal Transduction/physiology
- Stem Cells/physiology
Collapse
Affiliation(s)
- Jackson James
- Department of Ophthalmology, Lied Transplant Center (LTC 11715), University of Nebraska Medical Center, 600 South 42nd Street, Omaha, Nebraska 68198-6395, USA
| | | | | | | |
Collapse
|
18
|
Dudek SM, Jacobson JR, Chiang ET, Birukov KG, Wang P, Zhan X, Garcia JGN. Pulmonary Endothelial Cell Barrier Enhancement by Sphingosine 1-Phosphate. J Biol Chem 2004; 279:24692-700. [PMID: 15056655 DOI: 10.1074/jbc.m313969200] [Citation(s) in RCA: 234] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We recently reported the critical importance of Rac GTPase-dependent cortical actin rearrangement in the augmentation of pulmonary endothelial cell (EC) barrier function by sphingosine 1-phosphate (S1P). We now describe functional roles for the actin-binding proteins cortactin and EC myosin light chain kinase (MLCK) in mediating this response. Antisense down-regulation of cortactin protein expression significantly inhibits S1P-induced barrier enhancement in cultured human pulmonary artery EC as measured by transendothelial electrical resistance (TER). Immunofluorescence studies reveal rapid, Rac-dependent translocation of cortactin to the expanded cortical actin band following S1P challenge, where colocalization with EC MLCK occurs within 5 min. Adenoviral overexpression of a Rac dominant negative mutant attenuates TER elevation by S1P. S1P also induces a rapid increase in cortactin tyrosine phosphorylation (within 30 s) critical to subsequent barrier enhancement, since EC transfected with a tyrosine-deficient mutant cortactin exhibit a blunted TER response. Direct binding of EC MLCK to the cortactin Src homology 3 domain appears essential to S1P barrier regulation, since cortactin blocking peptide inhibits both S1P-induced MLC phosphorylation and peak S1P-induced TER values. These data support novel roles for the cytoskeletal proteins cortactin and EC MLCK in mediating lung vascular barrier augmentation evoked by S1P.
Collapse
Affiliation(s)
- Steven M Dudek
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Domowicz MS, Mangoura D, Schwartz NB. Aggrecan regulates telencephalic neuronal aggregation in culture. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 143:207-16. [PMID: 12855192 DOI: 10.1016/s0165-3806(03)00133-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Proteoglycans have been suggested to play roles in pattern formation in the developing central nervous system. In the chick embryo, aggrecan, a chondroitin sulfate proteoglycan, has a regionally-specific and developmentally-regulated expression profile. Telencephalic neuronal cultures, when aggregated, exhibit aggrecan expression patterns comparable to those observed in vivo. The chicken mutation nanomelia produces a truncated aggrecan species that cannot be processed further and is not secreted. Neurons from normal and nanomelic chick embryo telencephalon were scored for aggregate formation and analyzed for distribution of aggrecan protein and expression of aggrecan mRNA. Distinctly different pattern formation, with respect to aggregate size (smaller) and number (fewer) were observed in poly-L-lysine plated neuronal cultures derived from nanomelic embryos when compared to those derived from normal embryos. Significantly, the nanomelic phenotype was subsequently rescued upon addition of the brain-specific form of aggrecan. Modulation of neuronal aggregate formation was mimicked by treatment with chondroitinase ABC but not other glycanases, and was rescued by addition of chondroitin 6-sulfate to the culture media. Lastly, although broad and diffuse distribution of aggrecan among the cell aggregates in the culture paradigm was observed by immunocytochemistry, mRNA in situ hybridization revealed that only a small population of cells in the center of the aggregates was responsible for the production of the secreted aggrecan found associated with neuronal aggregates. These studies suggest a function for aggrecan as a diffusible signal in CNS histomorphogenesis.
Collapse
Affiliation(s)
- Miriam S Domowicz
- Departments of Pediatrics and Biochemistry & Molecular Biology, Committee on Developmental Biology, The University of Chicago, 5841 S. Maryland Avenue, MC 5058, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
20
|
Li C, Cheng Y, Gutmann DA, Mangoura D. Differential localization of the neurofibromatosis 1 (NF1) gene product, neurofibromin, with the F-actin or microtubule cytoskeleton during differentiation of telencephalic neurons. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2001; 130:231-48. [PMID: 11675125 DOI: 10.1016/s0165-3806(01)00190-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The protein product of the neurofibromatosis 1 gene, neurofibromin, is abundantly expressed in the cerebral cortex during development, but its physiological role remains unknown. To gain insights into the functions of neurofibromin in neurons, we examined patterns of expression and subcellular localization of neurofibromin during neuronal differentiation. Western blot analysis of telencephali homogenates throughout chick embryogenesis revealed that neurofibromin expression increased during embryonic development. Further analysis showed that telencephalic neurons were also enriched in neurofibromin in culture and that a biphasic gain in expression correlated well with both phases of differentiation in culture, first with a massive outgrowth of processes and gains in neurotransmitter phenotype differentiation, and then with synapse formation. Compared to proteins associated with distinct cytoskeleton systems, the pattern of neurofibromin expression correlated closely with that of the cortical cytoskeleton protein paxillin. Moreover, analysis of immunofluorescence staining of neurofibromin showed that in the presence of a protein crosslinker which preserves both soluble and filamentous cytoskeleton proteins after extraction with Triton X-100, neurofibromin colocalized with F-actin only during the first differentiation phase. This colocalization persisted when the actin cytoskeleton was collapsed with cytochalasin D treatment. In contrast, during the second phase of differentiation neurofibromin colocalized with microtubules, but not F-actin, and the staining pattern was disrupted with nocodazole, but not cytochalasin. A constant finding under all conditions was the presence of neurofibromin in the nucleus, which supports the idea that the bipartite nuclear targeting sequence between residues 2555 and 2572 of neurofibromin may be functional. In summary, we have shown that telencephalic neurons and astroblasts are enriched in neurofibromin and that the subcellular targeting of neurofibromin toward the actin or the microtubule cytoskeleton is developmentally regulated.
Collapse
Affiliation(s)
- C Li
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|